1
|
Yeow ZY, Sarju S, Chang FC, Xu LY, van Breugel M, Holland AJ. Mesoscale regulation of microtubule-organizing centers by the E3 ligase TRIM37. Nat Struct Mol Biol 2025:10.1038/s41594-025-01540-6. [PMID: 40415023 DOI: 10.1038/s41594-025-01540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/19/2025] [Indexed: 05/27/2025]
Abstract
Centrosomes ensure accurate chromosome segregation during cell division. Although the regulation of centrosome number is well established, less is known about the suppression of noncentrosomal microtubule-organizing centers (ncMTOCs). The E3 ligase TRIM37, implicated in Mulibrey nanism and 17q23-amplified cancers, has emerged as a key regulator of both centrosomes and ncMTOCs. Yet, the mechanism by which TRIM37 achieves enzymatic activation to target these mesoscale structures had thus far remained unknown. Here we elucidate the activation process of TRIM37, unveiling a process that initiates with TRAF domain-directed substrate recognition followed by B-box domain-mediated oligomerization and culminates in RING domain dimerization. Using optogenetics, we demonstrate that the E3 activity of TRIM37 is directly coupled to the assembly state of its substrates, being activated only when centrosomal proteins cluster into higher-order assemblies resembling MTOCs. This regulatory framework provides a mechanistic basis for understanding TRIM37-driven pathologies and echoes the restriction of the human immunodeficiency virus capsid by TRIM5, thus unveiling a conserved activation blueprint among TRIM proteins to control turnover of complexes assembled at the mesoscale level.
Collapse
Affiliation(s)
- Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sonia Sarju
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fang-Chi Chang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lance Y Xu
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark van Breugel
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Rendo V, Schubert M, Khuu N, Suarez Peredo Rodriguez MF, Whyte D, Ling X, van den Brink A, Huang K, Swift M, He Y, Zerbib J, Smith R, Raaijmakers J, Bandopadhayay P, Guenther LM, Hwang JH, Iniguez A, Moody S, Seo JH, Stover EH, Garraway L, Hahn WC, Stegmaier K, Medema RH, Chowdhury D, Colomé-Tatché M, Ben-David U, Beroukhim R, Foijer F. A compendium of Amplification-Related Gain Of Sensitivity genes in human cancer. Nat Commun 2025; 16:1077. [PMID: 39870664 PMCID: PMC11772776 DOI: 10.1038/s41467-025-56301-2] [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: 07/03/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
While the effect of amplification-induced oncogene expression in cancer is known, the impact of copy-number gains on "bystander" genes is less understood. We create a comprehensive map of dosage compensation in cancer by integrating expression and copy number profiles from over 8000 tumors in The Cancer Genome Atlas and cell lines from the Cancer Cell Line Encyclopedia. Additionally, we analyze 17 cancer open reading frame screens to identify genes toxic to cancer cells when overexpressed. Combining these approaches, we propose a class of 'Amplification-Related Gain Of Sensitivity' (ARGOS) genes located in commonly amplified regions, yet expressed at lower levels than expected by their copy number, and toxic when overexpressed. We validate RBM14 as an ARGOS gene in lung and breast cancer cells, and suggest a toxicity mechanism involving altered DNA damage response and STING signaling. We additionally observe increased patient survival in a radiation-treated cancer cohort with RBM14 amplification.
Collapse
Affiliation(s)
- Veronica Rendo
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Michael Schubert
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, Netherlands.
- Institute of Computational Biology, Helmholtz Munich, Neuherberg, Germany.
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Nicholas Khuu
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Declan Whyte
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, Netherlands
| | - Xiao Ling
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, Netherlands
| | - Anouk van den Brink
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, Netherlands
| | - Kaimeng Huang
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michelle Swift
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yizhou He
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Johanna Zerbib
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ross Smith
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jonne Raaijmakers
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Pratiti Bandopadhayay
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pediatrics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lillian M Guenther
- St. Jude Children's Research Hospital, Department of Oncology, Memphis, TN, USA
| | - Justin H Hwang
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Amanda Iniguez
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Moody
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ji-Heui Seo
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elizabeth H Stover
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Levi Garraway
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - William C Hahn
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kimberly Stegmaier
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pediatrics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - René H Medema
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dipanjan Chowdhury
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Colomé-Tatché
- Institute of Computational Biology, Helmholtz Munich, Neuherberg, Germany
- Biomedical Center (BMC), Physiological Chemistry, Ludwig Maximilians University, Munich, Germany
| | - Uri Ben-David
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Rameen Beroukhim
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
3
|
Yeow ZY, Sarju S, Breugel MV, Holland AJ. Mesoscale regulation of MTOCs by the E3 ligase TRIM37. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617407. [PMID: 39416078 PMCID: PMC11482927 DOI: 10.1101/2024.10.09.617407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Centrosomes ensure accurate chromosome segregation during cell division. Although the regulation of centrosome number is well-established, less is known about the suppression of non-centrosomal MTOCs (ncMTOCs). The E3 ligase TRIM37, implicated in Mulibrey nanism and 17q23-amplified cancers, has emerged as a key regulator of both centrosomes and ncMTOCs. Yet, the mechanism by which TRIM37 achieves enzymatic activation to target these mesoscale structures had remained unknown. Here, we elucidate TRIM37's activation process, beginning with TRAF domain-directed substrate recognition, progressing through B-box domain-mediated oligomerization, and culminating in RING domain dimerization. Using optogenetics, we demonstrate that TRIM37's E3 activity is directly coupled to the assembly state of its substrates, activating only when centrosomal proteins cluster into higher-order assemblies resembling MTOCs. This regulatory framework provides a mechanistic basis for understanding TRIM37-driven pathologies and, by echoing TRIM5's restriction of the HIV capsid, unveils a conserved activation blueprint among TRIM proteins for controlling mesoscale assembly turnover.
Collapse
Affiliation(s)
- Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sonia Sarju
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark V Breugel
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 2AT, UK
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Lin S, Xiong J, Zhou F, Fu J, Luo H, Wan Z, Luo J, Cao K. Molecular mechanism of RBM14-mediated promotion of proliferation, migration, and invasion in osteosarcoma. Transl Cancer Res 2024; 13:2122-2140. [PMID: 38881928 PMCID: PMC11170514 DOI: 10.21037/tcr-23-2070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/17/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Osteosarcoma (OS) is an exceptionally aggressive bone neoplasm that predominantly impacts the paediatric and adolescent population, exhibiting unfavourable prognosis. The importance of RNA binding motif protein 14 (RBM14) in the aetiology of OS is not well understood, despite its established involvement in several other types of cancer. METHODS In this study, we conducted an analysis of the expression profiles of RBM14 in cancer tissues and cell lines. To achieve this, we will utilised data obtained from various databases including The Cancer Genome Atlas Program (TCGA) project, The Genotype-Tissue Expression (GTEx) Project, Gene Expression Omnibus (GEO) database, and cancer cell line encyclopedia (CCLE) data. Furthermore, this study also aims to examine the effects of RBM14 on the proliferation, migration, and invasive properties of OS cells using cell functional gain and loss studies. In this study, we carried out an in-depth investigation to explore possible molecular pathways that underlie the regulation of the malignant phenotype found in OS by RBM14. This investigation involved integrating data from RBM14 overexpression, RBM14 knockdown RNA-seq experiments, and an array comprising 6,096 perturbed genes obtained from the Genetic Perturbation Similarity Analysis Database (GPSAdb). This research offers an opportunity to build a robust conceptual framework for the potential advancement of novel therapeutic approaches that are especially aimed at attacking OS. RESULTS RBM14 plays an active role in OS by significantly contributing to the enhancement of cellular proliferation, migration, and invasion. At the molecular level, it is probable that RBM14 exerts control over the malignant characteristics of OS through its modulation of the Hippo signalling system. CONCLUSIONS The above-mentioned findings underscore the significant importance of RBM14 as an intriguing target for therapy for the mitigation and management of OS. This particular protein holds an excellent opportunity for the development of novel and efficacious therapeutic approaches that possess the potential to yield favorable results for patients affected with OS.
Collapse
Affiliation(s)
- Sijian Lin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiachao Xiong
- The Orthopaedic Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Faxin Zhou
- The Orthopaedic Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jimin Fu
- The Orthopaedic Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hao Luo
- The Orthopaedic Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zongmiao Wan
- The Orthopaedic Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Cao
- The Orthopaedic Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Kweon TH, Jung H, Ko JY, Kang J, Kim W, Kim Y, Kim HB, Yi EC, Ku NO, Cho JW, Yang WH. O-GlcNAcylation of RBM14 contributes to elevated cellular O-GlcNAc through regulation of OGA protein stability. Cell Rep 2024; 43:114163. [PMID: 38678556 DOI: 10.1016/j.celrep.2024.114163] [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: 11/01/2023] [Revised: 03/18/2024] [Accepted: 04/11/2024] [Indexed: 05/01/2024] Open
Abstract
Dysregulation of O-GlcNAcylation has emerged as a potential biomarker for several diseases, particularly cancer. The role of OGT (O-GlcNAc transferase) in maintaining O-GlcNAc homeostasis has been extensively studied; nevertheless, the regulation of OGA (O-GlcNAcase) in cancer remains elusive. Here, we demonstrated that the multifunctional protein RBM14 is a regulator of cellular O-GlcNAcylation. By investigating the correlation between elevated O-GlcNAcylation and increased RBM14 expression in lung cancer cells, we discovered that RBM14 promotes ubiquitin-dependent proteasomal degradation of OGA, ultimately mediating cellular O-GlcNAcylation levels. In addition, RBM14 itself is O-GlcNAcylated at serine 521, regulating its interaction with the E3 ligase TRIM33, consequently affecting OGA protein stability. Moreover, we demonstrated that mutation of serine 521 to alanine abrogated the oncogenic properties of RBM14. Collectively, our findings reveal a previously unknown mechanism for the regulation of OGA and suggest a potential therapeutic target for the treatment of cancers with dysregulated O-GlcNAcylation.
Collapse
Affiliation(s)
- Tae Hyun Kweon
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 03722, Republic of Korea; Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyeryeon Jung
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul 03080, Republic of Korea
| | - Jeong Yeon Ko
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jingu Kang
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 03722, Republic of Korea; Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Wonyoung Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yeolhoe Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Han Byeol Kim
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul 03080, Republic of Korea
| | - Nam-On Ku
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 03722, Republic of Korea
| | - Jin Won Cho
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 03722, Republic of Korea; Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Won Ho Yang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
6
|
Li Z, Liao Y, Tang C, Xu L, Peng B, Xu X. RBM14 promotes DNA end resection during homologous recombination repair. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1864-1873. [PMID: 37559455 PMCID: PMC10753362 DOI: 10.3724/abbs.2023104] [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: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 08/11/2023] Open
Abstract
DNA double-strand break (DSB) repair by homologous recombination (HR) is crucial for the maintenance of genome stability and integrity. In this study, we aim to identify novel RNA binding proteins (RBPs) involved in HR repair because little is known about RBP function in HR. For this purpose, we carry out pulldown assays using a synthetic ssDNA/dsDNA structure coated with replication protein A (RPA) to mimic resected DNA, a crucial intermediate in HR-mediated DSB repair. Using this approach, we identify RNA-binding motif protein 14 (RBM14) as a potential binding partner. We further show that RBM14 interacts with an essential HR repair factor, CtIP. RBM14 is crucial for CtIP recruitment to DSB sites and for subsequent RPA coating and RAD51 replacement, facilitating efficient HR repair. Moreover, inhibition of RBM14 expression sensitizes cancer cells to X-ray irradiation. Together, our results demonstrate that RBM14 promotes DNA end resection to ensure HR repair and may serve as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Zheng Li
- College of Life SciencesCapital Normal UniversityBeijing100048China
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer CenterMarshall Laboratory of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518060China
| | - Yanting Liao
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer CenterMarshall Laboratory of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518060China
| | - Chen Tang
- State Key Laboratory of Agro-biotechnology and MOA Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijing100091China
- Shenzhen University General Hospital-Dehua Hospital Joint Research Center on Precision Medicine (sgh-dhhCPM)Dehua HospitalDehua362500China
| | - Linli Xu
- College of Life SciencesCapital Normal UniversityBeijing100048China
| | - Bin Peng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer CenterMarshall Laboratory of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518060China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer CenterMarshall Laboratory of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518060China
| |
Collapse
|
7
|
Pierron M, Woglar A, Busso C, Jha K, Mikeladze‐Dvali T, Croisier M, Gönczy P. Centriole elimination during Caenorhabditis elegans oogenesis initiates with loss of the central tube protein SAS-1. EMBO J 2023; 42:e115076. [PMID: 37987153 PMCID: PMC10711648 DOI: 10.15252/embj.2023115076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023] Open
Abstract
In most metazoans, centrioles are lost during oogenesis, ensuring that the zygote is endowed with the correct number of two centrioles, which are paternally contributed. How centriole architecture is dismantled during oogenesis is not understood. Here, we analyze with unprecedent detail the ultrastructural and molecular changes during oogenesis centriole elimination in Caenorhabditis elegans. Centriole elimination begins with loss of the so-called central tube and organelle widening, followed by microtubule disassembly. The resulting cluster of centriolar proteins then disappears gradually, usually moving in a microtubule- and dynein-dependent manner to the plasma membrane. Our analysis indicates that neither Polo-like kinases nor the PCM, which modulate oogenesis centriole elimination in Drosophila, do so in C. elegans. Furthermore, we demonstrate that the central tube protein SAS-1 normally departs initially from the organelle, which loses integrity earlier in sas-1 mutants. Overall, our work provides novel mechanistic insights regarding the fundamental process of oogenesis centriole elimination.
Collapse
Affiliation(s)
- Marie Pierron
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Alexander Woglar
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Keshav Jha
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | | | - Marie Croisier
- BIO‐EM platform, School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| |
Collapse
|
8
|
Kalbfuss N, Gönczy P. Towards understanding centriole elimination. Open Biol 2023; 13:230222. [PMID: 37963546 PMCID: PMC10645514 DOI: 10.1098/rsob.230222] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/14/2023] [Indexed: 11/16/2023] Open
Abstract
Centrioles are microtubule-based structures crucial for forming flagella, cilia and centrosomes. Through these roles, centrioles are critical notably for proper cell motility, signalling and division. Recent years have advanced significantly our understanding of the mechanisms governing centriole assembly and architecture. Although centrioles are typically very stable organelles, persisting over many cell cycles, they can also be eliminated in some cases. Here, we review instances of centriole elimination in a range of species and cell types. Moreover, we discuss potential mechanisms that enable the switch from a stable organelle to a vanishing one. Further work is expected to provide novel insights into centriole elimination mechanisms in health and disease, thereby also enabling scientists to readily manipulate organelle fate.
Collapse
Affiliation(s)
- Nils Kalbfuss
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
9
|
Xu D, Qin R, Li M, Shen J, Mao Y, Tang K, Zhang A, Wang D, Shi Y. Identification of a novel cell cycle-related risk signature predicting prognosis in patients with pancreatic adenocarcinoma. Medicine (Baltimore) 2022; 101:e29683. [PMID: 36401386 PMCID: PMC9678543 DOI: 10.1097/md.0000000000029683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Growing evidence have indicated that cell cycle-related genes (CRGs) play an essential role in the progression of pancreatic adenocarcinoma (PAAD). Nevertheless, the application of CRGs in estimating the prognosis of PAAD patients is still lacking. This study aimed to establish a risk signature based on CRGs that can predict patients' overall survival for PAAD. METHODS The expression and corresponding clinical data of PAAD patients from The Cancer Genome Atlas database and 200 cell cycle-related genes from the MSigDB were used for the generation and validation of the signature. LASSO Cox regression was applied to build the prediction model. The diagnostic value of signature was evaluated by receiver operating characteristic curves. Univariate and multivariate regression was used to construct the nomogram providing the clinicians a useful tool. RESULTS A total of 103 CRGs were identified. Seven genes (RBM14, SMAD3, CENPA, KIF23, NUSAP1, INCENP, SMC4) with non-zero coefficients in LASSO analysis were used to construct the prognostic signature. The 7-gene signature significantly stratified patients into high- and low-risk groups in terms of overall survival, and the area under the receiver operating characteristic curve of 5-year survival reached 0.749. Multivariate analysis showed that the signature is an independent prognostic factor. We then mapped a nomogram to predict 1-, 3-, and 5-year survival for PAAD patients. The calibration curves indicated that the model was reliable. Finally, we discovered that TP53 and KRAS mutated most frequently in low and high-risk groups, respectively. CONCLUSION Our findings suggested that the seven genes identified in this study are valuable prognostic predictors for patients with PAAD. These findings provided us with a novel insight that it is useful for understanding cell cycle mechanisms and for identifying patients with PAAD with poor prognosis.
Collapse
Affiliation(s)
- Dapeng Xu
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Rong Qin
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Ming Li
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Jun Shen
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yongmin Mao
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Kai Tang
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Aiguo Zhang
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Dafeng Wang
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yingzuo Shi
- Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
- * Correspondence: Yingzuo Shi, Department of Pediatric Surgery, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China (e-mail: )
| |
Collapse
|
10
|
Narabayashi H, Koma C, Nakata K, Ikegami M, Nakanishi Y, Ogihara J, Tsuda M, Hosono A, Hanazawa S, Takahashi K. Gut microbiota-dependent adaptor molecule recruits DNA methyltransferase to the TLR4 gene in colonic epithelial cells to suppress inflammatory reactions. Front Mol Biosci 2022; 9:1005136. [PMID: 36339704 PMCID: PMC9634067 DOI: 10.3389/fmolb.2022.1005136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
The intestine is inhabited by a large number of commensal bacteria that are immunologically non-self, potentially causing inflammation. However, in a healthy intestine, inflammation is strictly controlled at low levels to maintain homeostasis. We previously reported that the gut microbiota induce DNA methylation of the gene encoding Toll-like receptor (TLR) 4, a pattern recognition receptor that recognizes lipopolysaccharides of gram-negative bacteria, in colonic epithelial cells, suggesting its role in controlling intestinal inflammation. However, there remains a question of how gut microbiota cause methylation of only specific genes including TLR4, despite the fact that DNA methyltransferase (DNMT) is common to all genes targeted for methylation. Here, we identified RBM14 as an adaptor molecule that recruits DNMT to the TLR4 gene. RBM14 was shown to bind DNMT3 and be expressed at significantly higher levels in an intestinal epithelial cell (IEC) line with hypermethylated TLR4 gene than in an IEC line with hypomethylated TLR4 gene. In addition, RBM14 interacted with DNA regions of the TLR4 gene, and knockdown of RBM14 suppressed DNA methylation of the TLR4 gene in IECs. Furthermore, RBM14 expression was higher in colonic epithelial cells of conventional mice than in those of germ-free mice. Collectively, these results indicate that the gut microbiota induce methylation of the TLR4 gene in colonic epithelial cells by upregulating RBM14, which can recruit DNMT3 to the gene. The regulation of adaptor molecules such as RBM14, which bind to specific target genes and recruit DNMT, can explain, at least in part, how gut microbiota contribute to the maintenance of intestinal homeostasis through epigenetic control of specific gene expression in IECs.
Collapse
Affiliation(s)
- Hikari Narabayashi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Chiharu Koma
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Kazuaki Nakata
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Mion Ikegami
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Yusuke Nakanishi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Jun Ogihara
- Department of Chemistry and Life Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Masato Tsuda
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Akira Hosono
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Shigemasa Hanazawa
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Kyoko Takahashi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
- *Correspondence: Kyoko Takahashi,
| |
Collapse
|
11
|
Takumi K, Kitagawa D. Experimental and Natural Induction of de novo Centriole Formation. Front Cell Dev Biol 2022; 10:861864. [PMID: 35445021 PMCID: PMC9014216 DOI: 10.3389/fcell.2022.861864] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 01/11/2023] Open
Abstract
In cycling cells, new centrioles are assembled in the vicinity of pre-existing centrioles. Although this canonical centriole duplication is a tightly regulated process in animal cells, centrioles can also form in the absence of pre-existing centrioles; this process is termed de novo centriole formation. De novo centriole formation is triggered by the removal of all pre-existing centrioles in the cell in various manners. Moreover, overexpression of polo-like kinase 4 (Plk4), a master regulatory kinase for centriole biogenesis, can induce de novo centriole formation in some cell types. Under these conditions, structurally and functionally normal centrioles can be formed de novo. While de novo centriole formation is normally suppressed in cells with intact centrioles, depletion of certain suppressor proteins leads to the ectopic formation of centriole-related protein aggregates in the cytoplasm. It has been shown that de novo centriole formation also occurs naturally in some species. For instance, during the multiciliogenesis of vertebrate epithelial cells, massive de novo centriole amplification occurs to form numerous motile cilia. In this review, we summarize the previous findings on de novo centriole formation, particularly under experimental conditions, and discuss its regulatory mechanisms.
Collapse
Affiliation(s)
- Kasuga Takumi
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Qin H, Qu Y, Yuan YF, Li YY, Qiao J. RBM14 Modulates Tubulin Acetylation and Regulates Spindle Morphology During Meiotic Maturation in Mouse Oocytes. Front Cell Dev Biol 2021; 9:635728. [PMID: 33604343 PMCID: PMC7884444 DOI: 10.3389/fcell.2021.635728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/12/2021] [Indexed: 02/05/2023] Open
Abstract
RBM14 is an RNA-binding protein that regulates spindle integrity in mitosis; however, its functions during meiosis are still unclear. In this study, we discovered that RBM14 expression was down-regulated in oocytes from old mice. The RBM14 distribution at different stages of meiosis was explored, while it presents overlapped localization patterns with α-tubulin in MI- and MII-stage oocytes. Treatment of MI-stage oocytes with spindle-perturbing agents revealed that RBM14 was co-localized with microtubules. RBM14 knockdown with RBM14-specific morpholino showed that RBM14-depleted oocytes underwent symmetric division compared to the controls. RBM14 knockdown also resulted in spindle defects and chromosome abnormalities during oocyte maturation, presumably due to α-tubulin hyperacetylation. Co-immunoprecipitation analysis demonstrated that RBM14 is interacted with endogenous α-tubulin in mammalian cells. These findings indicate that RBM14 is an essential modulator of oocyte meiotic maturation by regulating α-tubulin acetylation to affect spindle morphology and chromosome alignment. Consequently, RBM14 represents a potential biomarker of oocyte quality and a novel therapeutic target in women with oocyte maturation failure.
Collapse
Affiliation(s)
- Hao Qin
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Peking University, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yi Qu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Peking University, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yi-Feng Yuan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Peking University, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yang-Yang Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Peking University, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Peking University, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Balestra FR, Domínguez-Calvo A, Wolf B, Busso C, Buff A, Averink T, Lipsanen-Nyman M, Huertas P, Ríos RM, Gönczy P. TRIM37 prevents formation of centriolar protein assemblies by regulating Centrobin. eLife 2021; 10:62640. [PMID: 33491649 PMCID: PMC7870141 DOI: 10.7554/elife.62640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
TRIM37 is an E3 ubiquitin ligase mutated in Mulibrey nanism, a disease with impaired organ growth and increased tumor formation. TRIM37 depletion from tissue culture cells results in supernumerary foci bearing the centriolar protein Centrin. Here, we characterize these centriolar protein assemblies (Cenpas) to uncover the mechanism of action of TRIM37. We find that an atypical de novo assembly pathway can generate Cenpas that act as microtubule-organizing centers (MTOCs), including in Mulibrey patient cells. Correlative light electron microscopy reveals that Cenpas are centriole-related or electron-dense structures with stripes. TRIM37 regulates the stability and solubility of Centrobin, which accumulates in elongated entities resembling the striped electron dense structures upon TRIM37 depletion. Furthermore, Cenpas formation upon TRIM37 depletion requires PLK4, as well as two parallel pathways relying respectively on Centrobin and PLK1. Overall, our work uncovers how TRIM37 prevents Cenpas formation, which would otherwise threaten genome integrity.
Collapse
Affiliation(s)
- Fernando R Balestra
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Andrés Domínguez-Calvo
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Alizée Buff
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Tessa Averink
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Marita Lipsanen-Nyman
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pablo Huertas
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Rosa M Ríos
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
14
|
Loss of DIAPH3, a Formin Family Protein, Leads to Cytokinetic Failure Only under High Temperature Conditions in Mouse FM3A Cells. Int J Mol Sci 2020; 21:ijms21228493. [PMID: 33187357 PMCID: PMC7696919 DOI: 10.3390/ijms21228493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 11/17/2022] Open
Abstract
Cell division is essential for the maintenance of life and involves chromosome segregation and subsequent cytokinesis. The processes are tightly regulated at both the spatial and temporal level by various genes, and failures in this regulation are associated with oncogenesis. Here, we investigated the gene responsible for defects in cell division by using murine temperature-sensitive (ts) mutant strains, tsFT101 and tsFT50 cells. The ts mutants normally grow in a low temperature environment (32 °C) but fail to divide in a high temperature environment (39 °C). Exome sequencing and over-expression analyses identified Diaph3, a member of the formin family, as the cause of the temperature sensitivity observed in tsFT101 and tsFT50 cells. Interestingly, Diaph3 knockout cells showed abnormality in cytokinesis at 39 °C, and the phenotype was rescued by re-expression of Diaph3 WT, but not Diaph1 and Diaph2, other members of the formin family. Furthermore, Diaph3 knockout cells cultured at 39 °C showed a significant increase in the level of acetylated α-tubulin, an index of stabilized microtubules, and the level was reduced by Diaph3 expression. These results suggest that Diaph3 is required for cytokinesis only under high temperature conditions. Therefore, our study provides a new insight into the mechanisms by which regulatory factors of cell division function in a temperature-dependent manner.
Collapse
|
15
|
A Genetic Screen for Human Genes Suppressing FUS Induced Toxicity in Yeast. G3-GENES GENOMES GENETICS 2020; 10:1843-1852. [PMID: 32276960 PMCID: PMC7263679 DOI: 10.1534/g3.120.401164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
FUS is a nucleic acid binding protein that, when mutated, cause a subset of familial amyotrophic lateral sclerosis (ALS). Expression of FUS in yeast recapitulates several pathological features of the disease-causing mutant proteins, including nuclear to cytoplasmic translocation, formation of cytoplasmic inclusions, and cytotoxicity. Genetic screens using the yeast model of FUS have identified yeast genes and their corresponding human homologs suppressing FUS induced toxicity in yeast, neurons and animal models. To expand the search for human suppressor genes of FUS induced toxicity, we carried out a genome-scale genetic screen using a newly constructed library containing 13570 human genes cloned in an inducible yeast-expression vector. Through multiple rounds of verification, we found 37 human genes that, when overexpressed, suppress FUS induced toxicity in yeast. Human genes with DNA or RNA binding functions are overrepresented among the identified suppressor genes, supporting that perturbations of RNA metabolism is a key underlying mechanism of FUS toxicity.
Collapse
|
16
|
Ito KK, Watanabe K, Kitagawa D. The Emerging Role of ncRNAs and RNA-Binding Proteins in Mitotic Apparatus Formation. Noncoding RNA 2020; 6:E13. [PMID: 32245090 PMCID: PMC7151635 DOI: 10.3390/ncrna6010013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022] Open
Abstract
Mounting experimental evidence shows that non-coding RNAs (ncRNAs) serve a wide variety of biological functions. Recent studies suggest that a part of ncRNAs are critically important for supporting the structure of subcellular architectures. Here, we summarize the current literature demonstrating the role of ncRNAs and RNA-binding proteins in regulating the assembly of mitotic apparatus, especially focusing on centrosomes, kinetochores, and mitotic spindles.
Collapse
Affiliation(s)
| | | | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan; (K.K.I.); (K.W.)
| |
Collapse
|
17
|
Kunimoto H, Inoue A, Kojima H, Yang J, Zhao H, Tsuruta D, Nakajima K. RBM10 regulates centriole duplication in HepG2 cells by ectopically assembling PLK4‐STIL complexes in the nucleus. Genes Cells 2020; 25:100-110. [DOI: 10.1111/gtc.12741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Hiroyuki Kunimoto
- Department of Immunology Graduate School of Medicine Osaka City University Osaka Japan
| | - Akira Inoue
- Department of Immunology Graduate School of Medicine Osaka City University Osaka Japan
| | - Hirotada Kojima
- Department of Immunology Graduate School of Medicine Osaka City University Osaka Japan
| | - Junhao Yang
- Department of Immunology Graduate School of Medicine Osaka City University Osaka Japan
| | - Hong Zhao
- Department of Immunology Graduate School of Medicine Osaka City University Osaka Japan
| | - Daisuke Tsuruta
- Department of Dermatology Graduate School of Medicine Osaka City University Osaka Japan
| | - Koichi Nakajima
- Department of Immunology Graduate School of Medicine Osaka City University Osaka Japan
| |
Collapse
|
18
|
Li J, Wang C, Feng G, Zhang L, Chen G, Sun H, Wang J, Zhang Y, Zhou Q, Li W. Rbm14 maintains the integrity of genomic DNA during early mouse embryogenesis via mediating alternative splicing. Cell Prolif 2020; 53:e12724. [PMID: 31794640 PMCID: PMC6985654 DOI: 10.1111/cpr.12724] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE In this study, we generated an Rbm14 knockout mouse model to explore its functions during early mouse embryogenesis. MATERIALS AND METHODS The Rbm14 knockout mouse model was generated by a combination of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and microinjection techniques. The developmental defects of the knockout embryos were characterized by histological analyses. The accumulation of DNA damage in mouse embryonic stem cells (ESCs) was detected by γH2AX staining and comet assay. The altered mRNA splicing of DNA damage response (DDR)-related genes was detected by RNA-Seq analysis and confirmed by semi-quantitative PCR. The interaction of RBM14 with alternative splicing-related genes was detected by immunoprecipitation-mass spectra (IP-MS) and confirmed by co-immunoprecipitation (Co-IP). RESULTS Rbm14 knockout in mice results in apoptosis and cell proliferation defects in early post-implantation epiblast cells, leading to gastrulation disruption and embryonic lethality. FACS and immunostaining demonstrate accumulation of DNA damage in Rbm14 knockout ES cells. We also identified altered splicing of DDR-related genes in the knockout mouse ESCs by RNA-Seq, indicating that RBM14-mediated alternative splicing is required for the maintenance of genome integrity during early mouse embryogenesis. CONCLUSIONS Our work reveals that Rbm14 plays an essential role in the maintenance of genome integrity during early mouse embryonic development by regulating alternative splicing of DDR-related genes.
Collapse
Affiliation(s)
- Jing Li
- School of Life SciencesUniversity of Science and Technology of ChinaHefeiChina
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Chenxin Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Linlin Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Guilai Chen
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Hao Sun
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jiaqiang Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- College of Life ScienceNortheast Agricultural University of ChinaHarbinChina
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Wei Li
- School of Life SciencesUniversity of Science and Technology of ChinaHefeiChina
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
19
|
NudC-like protein 2 restrains centriole amplification by stabilizing HERC2. Cell Death Dis 2019; 10:628. [PMID: 31427565 PMCID: PMC6700069 DOI: 10.1038/s41419-019-1843-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/27/2019] [Accepted: 07/26/2019] [Indexed: 01/02/2023]
Abstract
Centriole duplication is tightly controlled to occur once per cell cycle, and disruption of this synchrony causes centriole amplification, which is frequently observed in many cancers. Our previous work showed that nuclear distribution gene C (NudC)-like protein 2 (NudCL2) localizes to centrosomes; however, little is known about the role of NudCL2 in the regulation of centrosome function. Here, we find that NudCL2 is required for accurate centriole duplication by stabilizing the E3 ligase HECT domain and RCC1-like domain-containing protein 2 (HERC2). Knockout (KO) of NudCL2 using CRISPR/Cas9-based genome editing or depletion of NudCL2 using small interfering RNA causes significant centriole amplification. Overexpression of NudCL2 significantly suppresses hydroxyurea-induced centriole overduplication. Quantitative proteomic analysis reveals that HERC2 is downregulated in NudCL2 KO cells. NudCL2 is shown to interact with and stabilize HERC2. Depletion of HERC2 leads to the similar defects to that in NudCL2-downregulated cells, and ectopic expression of HERC2 effectively rescues the centriole amplification caused by the loss of NudCL2, whereas the defects induced by HERC2 depletion cannot be reversed by exogenous expression of NudCL2. Either loss of NudCL2 or depletion of HERC2 leads to the accumulation of ubiquitin-specific peptidase 33 (USP33), a centrosomal protein that positively regulates centriole duplication. Moreover, knockdown of USP33 reverses centriole amplification in both NudCL2 KO and HERC2-depleted cells. Taken together, our data suggest that NudCL2 plays an important role in maintaining the fidelity of centriole duplication by stabilizing HERC2 to control USP33 protein levels, providing a previously undescribed mechanism restraining centriole amplification.
Collapse
|
20
|
Watanabe K, Takao D, Ito KK, Takahashi M, Kitagawa D. The Cep57-pericentrin module organizes PCM expansion and centriole engagement. Nat Commun 2019; 10:931. [PMID: 30804344 PMCID: PMC6389942 DOI: 10.1038/s41467-019-08862-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/31/2019] [Indexed: 02/07/2023] Open
Abstract
Centriole duplication occurs once per cell cycle to ensure robust formation of bipolar spindles and chromosome segregation. Each newly-formed daughter centriole remains connected to its mother centriole until late mitosis. The disengagement of the centriole pair is required for centriole duplication. However, the mechanisms underlying centriole engagement remain poorly understood. Here, we show that Cep57 is required for pericentriolar material (PCM) organization that regulates centriole engagement. Depletion of Cep57 causes PCM disorganization and precocious centriole disengagement during mitosis. The disengaged daughter centrioles acquire ectopic microtubule-organizing-center activity, which results in chromosome mis-segregation. Similar defects are observed in mosaic variegated aneuploidy syndrome patient cells with cep57 mutations. We also find that Cep57 binds to the well-conserved PACT domain of pericentrin. Microcephaly osteodysplastic primordial dwarfism disease pericentrin mutations impair the Cep57-pericentrin interaction and lead to PCM disorganization. Together, our work demonstrates that Cep57 provides a critical interface between the centriole core and PCM. Centriole disengagement occurs towards mitotic exit and involves cleavage of pericentrin, a component of the pericentriolar material. Here the authors show that depletion of the centrosomal protein Cep57 leads to precocious centriole disengagement, and that Cep57 binds pericentrin.
Collapse
Affiliation(s)
- Koki Watanabe
- Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.,Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, 240-0193, Japan.,Department of Physiological Chemistry, Graduate school of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Daisuke Takao
- Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.,Department of Physiological Chemistry, Graduate school of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Kei K Ito
- Department of Physiological Chemistry, Graduate school of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Mikiko Takahashi
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Tokyo, 164-8530, Japan
| | - Daiju Kitagawa
- Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan. .,Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, 240-0193, Japan. .,Department of Physiological Chemistry, Graduate school of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
21
|
Chen G, Zhang D, Zhang L, Feng G, Zhang B, Wu Y, Li W, Zhang Y, Hu B. RBM14 is indispensable for pluripotency maintenance and mesoderm development of mouse embryonic stem cells. Biochem Biophys Res Commun 2018; 501:259-265. [PMID: 29729270 DOI: 10.1016/j.bbrc.2018.04.231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
The pluripotency of embryonic stem cells (ESCs) is maintained by core pluripotency transcription factors, cofactors and several signaling pathways. RBM14 is a component of the para-speckle complex, which has been implicated in multiple important biological processes. The role of RBM14 in ESCs and lineage differentiation remains to be elucidated. In the present study, we provided evidence that RBM14 plays important roles in maintaining pluripotency and in the early differentiation of ESCs. RBM14 was demonstrated to be expressed in mouse embryonic stem cells (mESCs) and localized in the nucleus. RBM14 expression was depleted in mESCs using clustered regularly interspaced short palindromic repeats (CRISPR) technology. Our results also showed that RBM14 depletion altered the gene expression profiles of mESCs. In particular, pluripotency-associated genes and genes involved in the Wnt and TGF-β signaling pathways were downregulated in RBM14 knockout mESCs. Furthermore, RBM14 was found to be essential for mesoderm development in vitro and in vivo. The specific effects of RBM14 depletion were verified by conducting a rescue experiment. Our findings demonstrated that RBM14 not only plays an important role in maintaining the pluripotency of mESCs but is also indispensable for mesoderm development.
Collapse
Affiliation(s)
- Guilai Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Linlin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Boya Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yihui Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China.
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
22
|
Di Francesco L, Verrico A, Asteriti IA, Rovella P, Cirigliano P, Guarguaglini G, Schininà ME, Lavia P. Visualization of human karyopherin beta-1/importin beta-1 interactions with protein partners in mitotic cells by co-immunoprecipitation and proximity ligation assays. Sci Rep 2018; 8:1850. [PMID: 29382863 PMCID: PMC5789818 DOI: 10.1038/s41598-018-19351-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 12/29/2017] [Indexed: 12/29/2022] Open
Abstract
Karyopherin beta-1/Importin beta-1 is a conserved nuclear transport receptor, acting in protein nuclear import in interphase and as a global regulator of mitosis. These pleiotropic functions reflect its ability to interact with, and regulate, different pathways during the cell cycle, operating as a major effector of the GTPase RAN. Importin beta-1 is overexpressed in cancers characterized by high genetic instability, an observation that highlights the importance of identifying its partners in mitosis. Here we present the first comprehensive profile of importin beta-1 interactors from human mitotic cells. By combining co-immunoprecipitation and proteome-wide mass spectrometry analysis of synchronized cell extracts, we identified expected (e.g., RAN and SUMO pathway factors) and novel mitotic interactors of importin beta-1, many with RNA-binding ability, that had not been previously associated with importin beta-1. These data complement interactomic studies of interphase transport pathways. We further developed automated proximity ligation assay (PLA) protocols to validate selected interactors. We succeeded in obtaining spatial and temporal resolution of genuine importin beta-1 interactions, which were visualized and localized in situ in intact mitotic cells. Further developments of PLA protocols will be helpful to dissect importin beta-1-orchestrated pathways during mitosis.
Collapse
Affiliation(s)
- Laura Di Francesco
- Dipartimento di Scienze Biochimiche, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.,Unit of Human Microbiome, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Annalisa Verrico
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, Via degli Apuli 4, 00185, Rome, Italy
| | - Italia Anna Asteriti
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, Via degli Apuli 4, 00185, Rome, Italy
| | - Paola Rovella
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, Via degli Apuli 4, 00185, Rome, Italy
| | | | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, Via degli Apuli 4, 00185, Rome, Italy
| | - Maria Eugenia Schininà
- Dipartimento di Scienze Biochimiche, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Patrizia Lavia
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, Via degli Apuli 4, 00185, Rome, Italy.
| |
Collapse
|
23
|
Firmin FF, Oger F, Gheeraert C, Dubois-Chevalier J, Vercoutter-Edouart AS, Alzaid F, Mazuy C, Dehondt H, Alexandre J, Derudas B, Dhalluin Q, Ploton M, Berthier A, Woitrain E, Lefebvre T, Venteclef N, Pattou F, Staels B, Eeckhoute J, Lefebvre P. The RBM14/CoAA-interacting, long intergenic non-coding RNA Paral1 regulates adipogenesis and coactivates the nuclear receptor PPARγ. Sci Rep 2017; 7:14087. [PMID: 29075020 PMCID: PMC5658386 DOI: 10.1038/s41598-017-14570-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/12/2017] [Indexed: 02/03/2023] Open
Abstract
Adipocyte differentiation and function relies on a network of transcription factors, which is disrupted in obesity-associated low grade, chronic inflammation leading to adipose tissue dysfunction. In this context, there is a need for a thorough understanding of the transcriptional regulatory network involved in adipose tissue pathophysiology. Recent advances in the functional annotation of the genome has highlighted the role of non-coding RNAs in cellular differentiation processes in coordination with transcription factors. Using an unbiased genome-wide approach, we identified and characterized a novel long intergenic non-coding RNA (lincRNA) strongly induced during adipocyte differentiation. This lincRNA favors adipocyte differentiation and coactivates the master adipogenic regulator peroxisome proliferator-activated receptor gamma (PPARγ) through interaction with the paraspeckle component and hnRNP-like RNA binding protein 14 (RBM14/NCoAA), and was therefore called PPARγ-activator RBM14-associated lncRNA (Paral1). Paral1 expression is restricted to adipocytes and decreased in humans with increasing body mass index. A decreased expression was also observed in diet-induced or genetic mouse models of obesity and this down-regulation was mimicked in vitro by TNF treatment. In conclusion, we have identified a novel component of the adipogenic transcriptional regulatory network defining the lincRNA Paral1 as an obesity-sensitive regulator of adipocyte differentiation and function.
Collapse
Affiliation(s)
- François F Firmin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Frederik Oger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Céline Gheeraert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Julie Dubois-Chevalier
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Anne-Sophie Vercoutter-Edouart
- CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FRABio FR 3688, Univ, Lille, Villeneuve d'Ascq, F-59650, France
| | - Fawaz Alzaid
- INSERM UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, F-75006, France
| | - Claire Mazuy
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Hélène Dehondt
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Jeremy Alexandre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Bruno Derudas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Quentin Dhalluin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Maheul Ploton
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Alexandre Berthier
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Eloise Woitrain
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Tony Lefebvre
- CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FRABio FR 3688, Univ, Lille, Villeneuve d'Ascq, F-59650, France
| | - Nicolas Venteclef
- INSERM UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, F-75006, France
| | - François Pattou
- Univ. Lille, Inserm, CHU Lille, U1190- EGID, F-59000, Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Jérôme Eeckhoute
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Philippe Lefebvre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France.
| |
Collapse
|
24
|
RNA processing as an alternative route to attack glioblastoma. Hum Genet 2017; 136:1129-1141. [PMID: 28608251 DOI: 10.1007/s00439-017-1819-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023]
Abstract
Genomic analyses have become an important tool to identify new avenues for therapy. This is especially true for cancer types with extremely poor outcomes, since our lack of effective therapies offers no tangible clinical starting point to build upon. The highly malignant brain tumor glioblastoma (GBM) exemplifies such a refractory cancer, with only 15 month average patient survival. Analyses of several hundred GBM samples compiled by the TCGA (The Cancer Genome Atlas) have produced an extensive transcriptomic map, identified prevalent chromosomal alterations, and defined important driver mutations. Unfortunately, clinical trials based on these results have not yet delivered an improvement on outcome. It is, therefore, necessary to characterize other regulatory routes known for playing a role in tumor relapse and response to treatment. Alternative splicing affects more than 90% of the human coding genes and it is an important source for transcript variation and gene regulation. Mutations and alterations in splicing factors are highly prevalent in multiple cancers, demonstrating the potential for splicing to act as a tumor driver. As a result, numerous genes are expressed as cancer-specific splicing isoforms that are functionally distinct from the canonical isoforms found in normal tissue. These include genes that regulate cancer-critical pathways such as apoptosis, DNA repair, cell proliferation, and migration. Splicing defects can even induce genomic instability, a common characteristic of cancer, and a driver of tumor evolution. Importantly, components of the splicing machinery are targetable; multiple drugs can inhibit splicing factors or promote changes in splicing which could be exploited to begin improving clinical outcomes. Here, we review the current literature and present a case for exploring RNA processing as therapeutic route for the treatment of GBM.
Collapse
|
25
|
Skrlj B, Konc J, Kunej T. Identification of Sequence Variants within Experimentally Validated Protein Interaction Sites Provides New Insights into Molecular Mechanisms of Disease Development. Mol Inform 2017; 36. [DOI: 10.1002/minf.201700017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 04/17/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Blaz Skrlj
- Department of molecular modeling; National Institute of Chemistry; Hajdrihova 19 Ljubljana Slovenia
- Jožef Stefan International Postgraduate School; Jamova cesta 39 1000 Ljubljana Slovenia
| | - Janez Konc
- Department of molecular modeling; National Institute of Chemistry; Hajdrihova 19 Ljubljana Slovenia
| | - Tanja Kunej
- Department of Animal Science, Biotechnical Faculty; University of Ljubljana
| |
Collapse
|
26
|
Tsuchiya Y, Yoshiba S, Gupta A, Watanabe K, Kitagawa D. Cep295 is a conserved scaffold protein required for generation of a bona fide mother centriole. Nat Commun 2016; 7:12567. [PMID: 27562453 PMCID: PMC5007451 DOI: 10.1038/ncomms12567] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/13/2016] [Indexed: 01/05/2023] Open
Abstract
Centrioles surrounded by pericentriolar material (PCM) serve as the core structure of the centrosome. A newly formed daughter centriole grows into a functional mother centriole. However, the underlying mechanisms remain poorly understood. Here we show that Cep295, an evolutionarily conserved protein, is required for generation of a bona fide mother centriole organizing a functional centrosome. We find that Cep295 is recruited to the proximal centriole wall in the early stages of procentriole assembly. Cep295 then acts as a scaffold for the proper assembly of the daughter centriole. We also find that Cep295 binds directly to and recruits Cep192 onto the daughter centriole wall, which presumably endows the function of the new mother centriole for PCM assembly, microtubule-organizing centre activity and the ability for centriole formation. These findings led us to propose that Cep295 acts upstream of the conserved pathway for centriole formation and promotes the daughter-to-mother centriole conversion.
Collapse
Affiliation(s)
- Yuki Tsuchiya
- Department of Molecular Genetics, Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka 411-8540, Japan
| | - Satoko Yoshiba
- Department of Molecular Genetics, Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Akshari Gupta
- Department of Molecular Genetics, Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka 411-8540, Japan
| | - Koki Watanabe
- Department of Molecular Genetics, Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka 411-8540, Japan
| | - Daiju Kitagawa
- Department of Molecular Genetics, Division of Centrosome Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
27
|
Shiratsuchi G, Kitagawa D. Suppression of ectopic assembly of centriole proteins ensures mitotic spindle integrity. Mol Cell Oncol 2016; 2:e1002717. [PMID: 27308496 PMCID: PMC4905339 DOI: 10.1080/23723556.2014.1002717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 10/25/2022]
Abstract
Abnormalities in maintaining the appropriate number of centrioles could be the origin of genome instability in tumor formation. Recently, we demonstrated that ectopic formation of aberrant centriole-related structures occurs even in the presence of pre-existing centrioles, leading to mitotic spindle defects and possibly contributing to tumorigenesis.
Collapse
Affiliation(s)
- Gen Shiratsuchi
- Centrosome Biology Laboratory; Center for Frontier Research; National Institute of Genetics ; Mishima, Shizuoka, Japan
| | - Daiju Kitagawa
- Centrosome Biology Laboratory; Center for Frontier Research; National Institute of Genetics ; Mishima, Shizuoka, Japan
| |
Collapse
|
28
|
Rao SR, Flores-Rodriguez N, Page SL, Wong C, Robinson PJ, Chircop M. The Clathrin-dependent Spindle Proteome. Mol Cell Proteomics 2016; 15:2537-53. [PMID: 27174698 DOI: 10.1074/mcp.m115.054809] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Indexed: 01/07/2023] Open
Abstract
The mitotic spindle is required for chromosome congression and subsequent equal segregation of sister chromatids. These processes involve a complex network of signaling molecules located at the spindle. The endocytic protein, clathrin, has a "moonlighting" role during mitosis, whereby it stabilizes the mitotic spindle. The signaling pathways that clathrin participates in to achieve mitotic spindle stability are unknown. Here, we assessed the mitotic spindle proteome and phosphoproteome in clathrin-depleted cells using quantitative MS/MS (data are available via ProteomeXchange with identifier PXD001603). We report a spindle proteome that consists of 3046 proteins and a spindle phosphoproteome consisting of 5157 phosphosites in 1641 phosphoproteins. Of these, 2908 (95.4%) proteins and 1636 (99.7%) phosphoproteins are known or predicted spindle-associated proteins. Clathrin-depletion from spindles resulted in dysregulation of 121 proteins and perturbed signaling to 47 phosphosites. The majority of these proteins increased in mitotic spindle abundance and six of these were validated by immunofluorescence microscopy. Functional pathway analysis confirmed the reported role of clathrin in mitotic spindle stabilization for chromosome alignment and highlighted possible new mechanisms of clathrin action. The data also revealed a novel second mitotic role for clathrin in bipolar spindle formation.
Collapse
Affiliation(s)
- Sushma R Rao
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Neftali Flores-Rodriguez
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Scott L Page
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Chin Wong
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Phillip J Robinson
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Megan Chircop
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| |
Collapse
|
29
|
Opposing post-translational modifications regulate Cep76 function to suppress centriole amplification. Oncogene 2016; 35:5377-5387. [PMID: 27065328 PMCID: PMC5125818 DOI: 10.1038/onc.2016.74] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 02/03/2016] [Accepted: 02/19/2016] [Indexed: 02/07/2023]
Abstract
Centrioles are critical for many cellular processes including cell division and cilia assembly. The number of centrioles within a cell is under strict control, and deregulation of centriole copy number is a hallmark of cancer. The molecular mechanisms that halt centriole amplification have not been fully elucidated. Here, we found that centrosomal protein of 76 kDa (Cep76), previously shown to restrain centriole amplification, interacts with cyclin-dependent kinase 2 (CDK2) and is a bona fide substrate of this kinase. Cep76 is preferentially phosphorylated by cyclin A/CDK2 at a single site S83, and this event is crucial to suppress centriole amplification in S phase. A novel Cep76 mutation S83C identified in a cancer patient fails to prevent centriole amplification. Mechanistically, Cep76 phosphorylation inhibits activation of polo-like kinase 1 (Plk1), thereby blocking premature centriole disengagement and subsequent amplification. Cep76 can also be acetylated, and enforced acetylation at K279 dampens the protein’s ability to inhibit amplification and precludes S83 phosphorylation. Acetylation of Cep76 normally occurs in G2 phase and correlates with loss of protein function. Our data suggest that temporal changes in posttranslational modifications of Cep76 during the cell cycle regulate its capacity to suppress centriole amplification, and its deregulation may contribute to malignancy.
Collapse
|
30
|
Abstract
Over a century ago, centrosome aberrations were postulated to cause cancer by promoting genome instability. The mechanisms governing centrosome assembly and function are increasingly well understood, allowing for a timely reappraisal of this postulate. This Review discusses recent advances that shed new light on the relationship between centrosomes and cancer, and raise the possibility that centrosome aberrations contribute to this disease in different ways than initially envisaged.
Collapse
Affiliation(s)
- Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
31
|
Van de Mark D, Kong D, Loncarek J, Stearns T. MDM1 is a microtubule-binding protein that negatively regulates centriole duplication. Mol Biol Cell 2015; 26:3788-802. [PMID: 26337392 PMCID: PMC4626064 DOI: 10.1091/mbc.e15-04-0235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/28/2015] [Indexed: 12/03/2022] Open
Abstract
MDM1 is a microtubule-binding protein that localizes to centrioles. 3D-SIM microscopy shows MDM1 to be closely associated with the centriole barrel, likely residing in the centriole lumen. MDM1 overexpression and depletion experiments suggest that MDM1 is a negative regulator of centriole duplication. Mouse double-minute 1 (Mdm1) was originally identified as a gene amplified in transformed mouse cells and more recently as being highly up-regulated during differentiation of multiciliated epithelial cells, a specialized cell type having hundreds of centrioles and motile cilia. Here we show that the MDM1 protein localizes to centrioles of dividing cells and differentiating multiciliated cells. 3D-SIM microscopy showed that MDM1 is closely associated with the centriole barrel, likely residing in the centriole lumen. Overexpression of MDM1 suppressed centriole duplication, whereas depletion of MDM1 resulted in an increase in granular material that likely represents early intermediates in centriole formation. We show that MDM1 binds microtubules in vivo and in vitro. We identified a repeat motif in MDM1 that is required for efficient microtubule binding and found that these repeats are also present in CCSAP, another microtubule-binding protein. We propose that MDM1 is a negative regulator of centriole duplication and that its function is mediated through microtubule binding.
Collapse
Affiliation(s)
| | - Dong Kong
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research-Frederick, National Cancer Institute, National Institutes of Health, Frederick, MD 21702
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research-Frederick, National Cancer Institute, National Institutes of Health, Frederick, MD 21702
| | - Tim Stearns
- Department of Biology, Stanford University, Stanford, CA 94305 Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305
| |
Collapse
|
32
|
Hennig S, Kong G, Mannen T, Sadowska A, Kobelke S, Blythe A, Knott GJ, Iyer KS, Ho D, Newcombe EA, Hosoki K, Goshima N, Kawaguchi T, Hatters D, Trinkle-Mulcahy L, Hirose T, Bond CS, Fox AH. Prion-like domains in RNA binding proteins are essential for building subnuclear paraspeckles. J Cell Biol 2015; 210:529-39. [PMID: 26283796 PMCID: PMC4539981 DOI: 10.1083/jcb.201504117] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 07/02/2015] [Indexed: 12/21/2022] Open
Abstract
Prion-like domains (PLDs) are low complexity sequences found in RNA binding proteins associated with the neurodegenerative disorder amyotrophic lateral sclerosis. Recently, PLDs have been implicated in mediating gene regulation via liquid-phase transitions that drive ribonucleoprotein granule assembly. In this paper, we report many PLDs in proteins associated with paraspeckles, subnuclear bodies that form around long noncoding RNA. We mapped the interactome network of paraspeckle proteins, finding enrichment of PLDs. We show that one protein, RBM14, connects key paraspeckle subcomplexes via interactions mediated by its PLD. We further show that the RBM14 PLD, as well as the PLD of another essential paraspeckle protein, FUS, is required to rescue paraspeckle formation in cells in which their endogenous counterpart has been knocked down. Similar to FUS, the RBM14 PLD also forms hydrogels with amyloid-like properties. These results suggest a role for PLD-mediated liquid-phase transitions in paraspeckle formation, highlighting this nuclear body as an excellent model system for understanding the perturbation of such processes in neurodegeneration.
Collapse
Affiliation(s)
- Sven Hennig
- The Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia The Centre for Medical Research, The University of Western Australia, Crawley, WA 6009, Australia
| | - Geraldine Kong
- The Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia The Centre for Medical Research, The University of Western Australia, Crawley, WA 6009, Australia
| | - Taro Mannen
- Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Agata Sadowska
- The Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia The Centre for Medical Research, The University of Western Australia, Crawley, WA 6009, Australia
| | - Simon Kobelke
- The Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia The Centre for Medical Research, The University of Western Australia, Crawley, WA 6009, Australia
| | - Amanda Blythe
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - Gavin J Knott
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - K Swaminathan Iyer
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - Diwei Ho
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - Estella A Newcombe
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kana Hosoki
- Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Naoki Goshima
- Molecular Profiling Research Center for Drug Discovery, National Institute for Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Tetsuya Kawaguchi
- Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Danny Hatters
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Laura Trinkle-Mulcahy
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5 Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Tetsuro Hirose
- Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Charles S Bond
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - Archa H Fox
- The Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia The Centre for Medical Research, The University of Western Australia, Crawley, WA 6009, Australia
| |
Collapse
|