1
|
Uechi H, Sridharan S, Nijssen J, Bilstein J, Iglesias-Artola JM, Kishigami S, Casablancas-Antras V, Poser I, Martinez EJ, Boczek E, Wagner M, Tomschke N, de Jesus Domingues AM, Pal A, Doeleman T, Kour S, Anderson EN, Stein F, Lee HO, Zhang X, Fritsch AW, Jahnel M, Fürsch J, Murthy AC, Alberti S, Bickle M, Fawzi NL, Nadler A, David DC, Pandey UB, Hermann A, Stengel F, Davis BG, Baldwin AJ, Savitski MM, Hyman AA, Wheeler RJ. Small-molecule dissolution of stress granules by redox modulation benefits ALS models. Nat Chem Biol 2025:10.1038/s41589-025-01893-5. [PMID: 40369342 DOI: 10.1038/s41589-025-01893-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/26/2025] [Indexed: 05/16/2025]
Abstract
Neurodegenerative diseases, such as amyotrophic lateral sclerosis, are often associated with mutations in stress granule proteins. Aberrant stress granule condensate formation is associated with disease, making it a potential target for pharmacological intervention. Here, we identified lipoamide, a small molecule that specifically prevents cytoplasmic condensation of stress granule proteins. Thermal proteome profiling showed that lipoamide stabilizes intrinsically disordered domain-containing proteins, including SRSF1 and SFPQ, which are stress granule proteins necessary for lipoamide activity. SFPQ has redox-state-specific condensate dissolving behavior, which is modulated by the redox-active lipoamide dithiolane ring. In animals, lipoamide ameliorates aging-associated aggregation of a stress granule reporter protein, improves neuronal morphology and recovers motor defects caused by amyotrophic lateral sclerosis-associated FUS and TDP-43 mutants. Thus, lipoamide is a well-tolerated small-molecule modulator of stress granule condensation, and dissection of its molecular mechanism identified a cellular pathway for redox regulation of stress granule formation.
Collapse
Affiliation(s)
- Hiroyuki Uechi
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Sindhuja Sridharan
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jik Nijssen
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jessica Bilstein
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Satoshi Kishigami
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Virginia Casablancas-Antras
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Ina Poser
- Dewpoint Therapeutics, Dresden, Germany
| | | | | | | | - Nadine Tomschke
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - António M de Jesus Domingues
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Dewpoint Therapeutics, Dresden, Germany
| | - Arun Pal
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Dresden High Magnetic Field Laboratory (HLD), Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Thom Doeleman
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Sukhleen Kour
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric Nathaniel Anderson
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Hyun O Lee
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Xiaojie Zhang
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Anatol W Fritsch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marcus Jahnel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
- Biotechnology Center (BIOTEC), CMCB, TU Dresden, Dresden, Germany
| | - Julius Fürsch
- University of Konstanz, Department of Biology, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Anastasia C Murthy
- Graduate Program in Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
| | - Simon Alberti
- Biotechnology Center (BIOTEC), CMCB, TU Dresden, Dresden, Germany
| | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Institute for Translational Bioengineering, pRED, Roche, Basel, Switzerland
| | - Nicolas L Fawzi
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
| | - André Nadler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Della C David
- German Centre for Neurodegenerative Diseases, Tübingen, Germany
- Babraham Institute, Cambridge, UK
| | - Udai B Pandey
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andreas Hermann
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock, Germany
- Translational Neurodegeneration Section 'Albrecht Kossel', Department of Neurology, University Medical Center Rostock, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Florian Stengel
- University of Konstanz, Department of Biology, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Benjamin G Davis
- Department of Chemistry, University of Oxford, Oxford, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
- The Rosalind Franklin Institute, Harwell, UK
| | - Andrew J Baldwin
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- The Rosalind Franklin Institute, Harwell, UK
| | - Mikhail M Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Richard J Wheeler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK.
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
Ly J, Tao YF, Di Bernardo M, Khalizeva E, Giuliano CJ, Lourido S, Fleming MD, Cheeseman IM. Alternative start codon selection shapes mitochondrial function during evolution, homeostasis, and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645657. [PMID: 40196624 PMCID: PMC11974929 DOI: 10.1101/2025.03.27.645657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Mitochondrial endosymbiosis was a pivotal event in eukaryotic evolution, requiring core proteins to adapt to function both within the mitochondria and in the host cell. Here, we systematically profile the localization of protein isoforms generated by alternate start codon selection during translation. We identify hundreds of pairs of differentially-localized protein isoforms, many of which affect mitochondrial targeting and are essential for mitochondrial function. The emergence of dual-localized mitochondrial protein isoforms coincides with mitochondrial acquisition during early eukaryotic evolution. We further reveal that eukaryotes use diverse mechanisms-such as leaky ribosome scanning, alternative transcription, and paralog duplication-to maintain the production of dual-localized isoforms. Finally, we identify multiple isoforms that are specifically dysregulated by rare disease patient mutations and demonstrate how these mutations can help explain unique clinical presentations. Together, our findings illuminate the evolutionary and pathological relevance of alternative translation initiation, offering new insights into the molecular underpinnings of mitochondrial biology.
Collapse
Affiliation(s)
- Jimmy Ly
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Yi Fei Tao
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Matteo Di Bernardo
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Ekaterina Khalizeva
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Christopher J. Giuliano
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Mark D. Fleming
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
3
|
Kanie T, Ng R, Abbott KL, Tanvir NM, Lorentzen E, Pongs O, Jackson PK. Myristoylated Neuronal Calcium Sensor-1 captures the preciliary vesicle at distal appendages. eLife 2025; 14:e85998. [PMID: 39882855 PMCID: PMC11984960 DOI: 10.7554/elife.85998] [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: 01/06/2023] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
The primary cilium is a microtubule-based organelle that cycles through assembly and disassembly. In many cell types, formation of the cilium is initiated by recruitment of preciliary vesicles to the distal appendage of the mother centriole. However, the distal appendage mechanism that directly captures preciliary vesicles is yet to be identified. In an accompanying paper, we show that the distal appendage protein, CEP89, is important for the preciliary vesicle recruitment, but not for other steps of cilium formation (Kanie et al., 2025). The lack of a membrane-binding motif in CEP89 suggests that it may indirectly recruit preciliary vesicles via another binding partner. Here, we identify Neuronal Calcium Sensor-1 (NCS1) as a stoichiometric interactor of CEP89. NCS1 localizes to the position between CEP89 and the centriole-associated vesicle marker, RAB34, at the distal appendage. This localization was completely abolished in CEP89 knockouts, suggesting that CEP89 recruits NCS1 to the distal appendage. Similar to CEP89 knockouts, preciliary vesicle recruitment as well as subsequent cilium formation was perturbed in NCS1 knockout cells. The ability of NCS1 to recruit the preciliary vesicle is dependent on its myristoylation motif and NCS1 knockout cells expressing a myristoylation defective mutant failed to rescue the vesicle recruitment defect despite localizing properly to the centriole. In sum, our analysis reveals the first known mechanism for how the distal appendage recruits the preciliary vesicles.
Collapse
Affiliation(s)
- Tomoharu Kanie
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford UniversityStanfordUnited States
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Roy Ng
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford UniversityStanfordUnited States
| | - Keene L Abbott
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford UniversityStanfordUnited States
| | | | - Esben Lorentzen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Olaf Pongs
- Institute for Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland UniversitySaarbrückenGermany
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford UniversityStanfordUnited States
| |
Collapse
|
4
|
Smith EM, Ly J, Haug S, Cheeseman IM. Molecular determinants of RNase MRP specificity and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635360. [PMID: 39974906 PMCID: PMC11838342 DOI: 10.1101/2025.01.28.635360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
RNase MRP and RNase P are evolutionarily related complexes that facilitate rRNA and tRNA biogenesis, respectively. The two enzymes share nearly all protein subunits and have evolutionarily related catalytic RNAs. Notably, RNase P includes a unique subunit, Rpp21, whereas no RNase MRP-specific proteins have been found in humans, limiting molecular analyses of RNase MRP function. Here, we identify the RNase MRP-specific protein, C18orf21/RMRPP1. RMRPP1 and Rpp21 display significant structural homology, but we identify specific regions that drive interactions with their respective complexes. Additionally, we reveal that RNase MRP is required for 40S, but not 60S, ribosome biogenesis uncovering an alternative pathway for ribosome assembly. Finally, we identify Nepro as an essential rRNA processing factor that associates with the RNase MRP complex. Together, our findings elucidate the molecular determinants of RNase MRP function and underscore its critical role in ribosome biogenesis.
Collapse
Affiliation(s)
- Eric M. Smith
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jimmy Ly
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sofia Haug
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
5
|
Ly J, Xiang K, Su KC, Sissoko GB, Bartel DP, Cheeseman IM. Nuclear release of eIF1 restricts start-codon selection during mitosis. Nature 2024; 635:490-498. [PMID: 39443796 PMCID: PMC11605796 DOI: 10.1038/s41586-024-08088-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Regulated start-codon selection has the potential to reshape the proteome through the differential production of upstream open reading frames, canonical proteins, and alternative translational isoforms1-3. However, conditions under which start codon selection is altered remain poorly defined. Here, using transcriptome-wide translation-initiation-site profiling4, we reveal a global increase in the stringency of start-codon selection during mammalian mitosis. Low-efficiency initiation sites are preferentially repressed in mitosis, resulting in pervasive changes in the translation of thousands of start sites and their corresponding protein products. This enhanced stringency of start-codon selection during mitosis results from increased association between the 40S ribosome and the key regulator of start-codon selection, eIF1. We find that increased eIF1-40S ribosome interaction during mitosis is mediated by the release of a nuclear pool of eIF1 upon nuclear envelope breakdown. Selectively depleting the nuclear pool of eIF1 eliminates the change to translational stringency during mitosis, resulting in altered synthesis of thousands of protein isoforms. In addition, preventing mitotic translational rewiring results in substantially increased cell death and decreased mitotic slippage in cells that experience a mitotic delay induced by anti-mitotic chemotherapies. Thus, cells globally control stringency of translation initiation, which has critical roles during the mammalian cell cycle in preserving mitotic cell physiology.
Collapse
Affiliation(s)
- Jimmy Ly
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kehui Xiang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Kuan-Chung Su
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gunter B Sissoko
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David P Bartel
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Ly J, Blengini CS, Cady SL, Schindler K, Cheeseman IM. A conserved germline-specific Dsn1 alternative splice isoform supports oocyte and embryo development. Curr Biol 2024; 34:4307-4317.e6. [PMID: 39178843 PMCID: PMC11421959 DOI: 10.1016/j.cub.2024.07.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/19/2024] [Accepted: 07/26/2024] [Indexed: 08/26/2024]
Abstract
The chromosome segregation and cell division programs associated with somatic mitosis and germline meiosis display dramatic differences such as kinetochore orientation, cohesin removal, or the presence of a gap phase.1,2,3,4,5,6 These changes in chromosome segregation require alterations to the established cell division machinery.5,6 It remains unclear what aspects of kinetochore function and its regulatory control differ between the mitotic and meiotic cell divisions to rewire these core processes. Alternative RNA splicing can generate distinct protein isoforms to allow for the differential control of cell processes across cell types. However, alternative splice isoforms that differentially modulate distinct cell division programs have remained elusive. Here, we demonstrate that mammalian germ cells express an alternative mRNA splice isoform for the kinetochore component, DSN1, a subunit of the MIS12 complex that links the centromeres to spindle microtubules during chromosome segregation. This germline DSN1 isoform bypasses the requirement for Aurora kinase phosphorylation for its centromere localization due to the absence of a key regulatory region allowing DSN1 to display persistent centromere localization. Expression of the germline DSN1 isoform in somatic cells results in constitutive kinetochore localization, chromosome segregation errors, and growth defects, providing an explanation for its tight cell-type-specific expression. Reciprocally, precisely eliminating expression of the germline-specific DSN1 splice isoform in mouse models disrupts oocyte maturation and early embryonic divisions coupled with a reduction in fertility. Together, this work identifies a germline-specific splice isoform for a chromosome segregation component and implicates its role in mammalian fertility.
Collapse
Affiliation(s)
- Jimmy Ly
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Sarah L Cady
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Karen Schindler
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
7
|
Johnstone M, Leck A, Lange T, Wilcher K, Shephard MS, Paranjpe A, Schutte S, Wells S, Kappes F, Salomonis N, Privette Vinnedge LM. The chromatin remodeler DEK promotes proliferation of mammary epithelium and is associated with H3K27me3 epigenetic modifications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612116. [PMID: 39314335 PMCID: PMC11419013 DOI: 10.1101/2024.09.09.612116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The DEK chromatin remodeling protein was previously shown to confer oncogenic phenotypes to human and mouse mammary epithelial cells using in vitro and knockout mouse models. However, its functional role in normal mammary gland epithelium remained unexplored. We developed two novel mouse models to study the role of Dek in normal mammary gland biology in vivo . Mammary gland-specific Dek over-expression in mice resulted in hyperproliferation of cells that visually resembled alveolar cells, and a transcriptional profile that indicated increased expression of cell cycle, mammary stem/progenitor, and lactation-associated genes. Conversely, Dek knockout mice exhibited an alveologenesis or lactation defect, resulting in dramatically reduced pup survival. Analysis of previously published single-cell RNA-sequencing of mouse mammary glands revealed that Dek is most highly expressed in mammary stem cells and alveolar progenitor cells, and to a lesser extent in basal epithelial cells, supporting the observed phenotypes. Mechanistically, we discovered that Dek is a modifier of Ezh2 methyltransferase activity, upregulating the levels of histone H3 trimethylation on lysine 27 (H3K27me3) to control gene transcription. Combined, this work indicates that Dek promotes proliferation of mammary epithelial cells via cell cycle deregulation. Furthermore, we report a novel function for Dek in alveologenesis and histone H3 K27 trimethylation.
Collapse
|
8
|
Khurana S, Varma D, Foltz DR. Contribution of CENP-F to FOXM1-Mediated Discordant Centromere and Kinetochore Transcriptional Regulation. Mol Cell Biol 2024; 44:209-225. [PMID: 38779933 PMCID: PMC11204039 DOI: 10.1080/10985549.2024.2350543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Proper chromosome segregation is required to ensure chromosomal stability. The centromere (CEN) is a unique chromatin domain defined by CENP-A and is responsible for recruiting the kinetochore (KT) during mitosis, ultimately regulating microtubule spindle attachment and mitotic checkpoint function. Upregulation of many CEN/KT genes is commonly observed in cancer. Here, we show that although FOXM1 occupies promoters of many CEN/KT genes with MYBL2, FOXM1 overexpression alone is insufficient to drive the FOXM1-correlated transcriptional program. CENP-F is canonically an outer kinetochore component; however, it functions with FOXM1 to coregulate G2/M transcription and proper chromosome segregation. Loss of CENP-F results in altered chromatin accessibility at G2/M genes and reduced FOXM1-MBB complex formation. We show that coordinated CENP-FFOXM1 transcriptional regulation is a cancer-specific function. We observe a small subset of CEN/KT genes including CENP-C, that are not regulated by FOXM1. Upregulation of CENP-C in the context of CENP-A overexpression leads to increased chromosome missegregation and cell death suggesting that escape of CENP-C from FOXM1 regulation is a cancer survival mechanism. Together, we show that FOXM1 and CENP-F coordinately regulate G2/M genes, and this coordination is specific to a subset of genes to allow for maintenance of chromosome instability levels and subsequent cell survival.
Collapse
Affiliation(s)
- Sakshi Khurana
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dileep Varma
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Cellular and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel R. Foltz
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
9
|
Ly J, Xiang K, Su KC, Sissoko GB, Bartel DP, Cheeseman IM. Nuclear release of eIF1 globally increases stringency of start-codon selection to preserve mitotic arrest physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588385. [PMID: 38617206 PMCID: PMC11014515 DOI: 10.1101/2024.04.06.588385] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Regulated start-codon selection has the potential to reshape the proteome through the differential production of uORFs, canonical proteins, and alternative translational isoforms. However, conditions under which start-codon selection is altered remain poorly defined. Here, using transcriptome-wide translation initiation site profiling, we reveal a global increase in the stringency of start-codon selection during mammalian mitosis. Low-efficiency initiation sites are preferentially repressed in mitosis, resulting in pervasive changes in the translation of thousands of start sites and their corresponding protein products. This increased stringency of start-codon selection during mitosis results from increased interactions between the key regulator of start-codon selection, eIF1, and the 40S ribosome. We find that increased eIF1-40S ribosome interactions during mitosis are mediated by the release of a nuclear pool of eIF1 upon nuclear envelope breakdown. Selectively depleting the nuclear pool of eIF1 eliminates the changes to translational stringency during mitosis, resulting in altered mitotic proteome composition. In addition, preventing mitotic translational rewiring results in substantially increased cell death and decreased mitotic slippage following treatment with anti-mitotic chemotherapeutics. Thus, cells globally control translation initiation stringency with critical roles during the mammalian cell cycle to preserve mitotic cell physiology.
Collapse
|
10
|
Ly J, Blengini CS, Cady SL, Schindler K, Cheeseman IM. A conserved germline-specific Dsn1 alternative splice isoform supports oocyte and embryo development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589883. [PMID: 38659852 PMCID: PMC11042369 DOI: 10.1101/2024.04.17.589883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Alternative mRNA splicing can generate distinct protein isoforms to allow for the differential control of cell processes across cell types. However, alternative splice isoforms that differentially modulate distinct cell division programs have remained elusive. Here, we demonstrate that mammalian germ cells express an alternate mRNA splice isoform for the kinetochore component, DSN1, a subunit of the MIS12 complex that links the centromeres to spindle microtubules during chromosome segregation. This germline DSN1 isoform bypasses the requirement for Aurora kinase phosphorylation for its centromere localization due to the absence of a key regulatory region allowing DSN1 to display persistent centromere localization. Expression of the germline DSN1 isoform in somatic cells results in constitutive kinetochore localization, chromosome segregation errors, and growth defects, providing an explanation for its tight cell type-specific expression. Reciprocally, precisely eliminating expression of the germline DSN1 splice isoform in mouse models disrupts oocyte maturation and early embryonic divisions coupled with a reduction in fertility. Together, this work identifies a germline-specific splice isoform for a chromosome segregation component and implicates its role in mammalian fertility.
Collapse
Affiliation(s)
- Jimmy Ly
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Cecilia S. Blengini
- Department of Genetics, Rutgers University, Piscataway, New Jersey, United States
| | - Sarah L. Cady
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Karen Schindler
- Department of Genetics, Rutgers University, Piscataway, New Jersey, United States
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
11
|
Weber J, Legal T, Lezcano AP, Gluszek-Kustusz A, Paterson C, Eibes S, Barisic M, Davies OR, Welburn JPI. A conserved CENP-E region mediates BubR1-independent recruitment to the outer corona at mitotic onset. Curr Biol 2024; 34:1133-1141.e4. [PMID: 38354735 DOI: 10.1016/j.cub.2024.01.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024]
Abstract
The outer corona plays an essential role at the onset of mitosis by expanding to maximize microtubule attachment to kinetochores.1,2 The low-density structure of the corona forms through the expansion of unattached kinetochores. It comprises the RZZ complex, the dynein adaptor Spindly, the plus-end directed microtubule motor centromere protein E (CENP-E), and the Mad1/Mad2 spindle-assembly checkpoint proteins.3,4,5,6,7,8,9,10 CENP-E specifically associates with unattached kinetochores to facilitate chromosome congression,11,12,13,14,15,16 interacting with BubR1 at the kinetochore through its C-terminal region (2091-2358).17,18,19,20,21 We recently showed that CENP-E recruitment to BubR1 at the kinetochores is both rapid and essential for correct chromosome alignment. However, CENP-E is also recruited to the outer corona by a second, slower pathway that is currently undefined.19 Here, we show that BubR1-independent localization of CENP-E is mediated by a conserved loop that is essential for outer-corona targeting. We provide a structural model of the entire CENP-E kinetochore-targeting domain combining X-ray crystallography and Alphafold2. We reveal that maximal recruitment of CENP-E to unattached kinetochores critically depends on BubR1 and the outer corona, including dynein. Ectopic expression of the CENP-E C-terminal domain recruits the RZZ complex, Mad1, and Spindly, and prevents kinetochore biorientation in cells. We propose that BubR1-recruited CENP-E, in addition to its essential role in chromosome alignment to the metaphase plate, contributes to the recruitment of outer corona proteins through interactions with the CENP-E corona-targeting domain to facilitate the rapid capture of microtubules for efficient chromosome alignment and mitotic progression.
Collapse
Affiliation(s)
- Jeraldine Weber
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK
| | - Thibault Legal
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK
| | - Alicia Perez Lezcano
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK
| | - Agata Gluszek-Kustusz
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK
| | - Calum Paterson
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK
| | - Susana Eibes
- Cell Division and Cytoskeleton, Danish Cancer Institute, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Institute, Strandboulevarden 49, 2100 Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 3C Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Owen R Davies
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK
| | - Julie P I Welburn
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3BF, UK.
| |
Collapse
|
12
|
Sissoko GB, Tarasovetc EV, Marescal O, Grishchuk EL, Cheeseman IM. Higher-order protein assembly controls kinetochore formation. Nat Cell Biol 2024; 26:45-56. [PMID: 38168769 PMCID: PMC10842828 DOI: 10.1038/s41556-023-01313-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 11/13/2023] [Indexed: 01/05/2024]
Abstract
To faithfully segregate chromosomes during vertebrate mitosis, kinetochore-microtubule interactions must be restricted to a single site on each chromosome. Prior work on pair-wise kinetochore protein interactions has been unable to identify the mechanisms that prevent outer kinetochore formation in regions with a low density of CENP-A nucleosomes. To investigate the impact of higher-order assembly on kinetochore formation, we generated oligomers of the inner kinetochore protein CENP-T using two distinct, genetically engineered systems in human cells. Although individual CENP-T molecules interact poorly with outer kinetochore proteins, oligomers that mimic centromeric CENP-T density trigger the robust formation of functional, cytoplasmic kinetochore-like particles. Both in cells and in vitro, each molecule of oligomerized CENP-T recruits substantially higher levels of outer kinetochore components than monomeric CENP-T molecules. Our work suggests that the density dependence of CENP-T restricts outer kinetochore recruitment to centromeres, where densely packed CENP-A recruits a high local concentration of inner kinetochore proteins.
Collapse
Affiliation(s)
- Gunter B Sissoko
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ekaterina V Tarasovetc
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Océane Marescal
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ekaterina L Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
13
|
Khurana S, Foltz DR. Contribution of CENP-F to FOXM1-mediated discordant centromere and kinetochore transcriptional regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573453. [PMID: 38234763 PMCID: PMC10793414 DOI: 10.1101/2023.12.27.573453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Proper chromosome segregation is required to ensure genomic and chromosomal stability. The centromere is a unique chromatin domain present throughout the cell cycle on each chromosome defined by the CENP-A nucleosome. Centromeres (CEN) are responsible for recruiting the kinetochore (KT) during mitosis, ultimately regulating spindle attachment and mitotic checkpoint function. Upregulation of many genes that encode the CEN/KT proteins is commonly observed in cancer. Here, we show although that FOXM1 occupies the promoters of many CEN/KT genes with MYBL2, occupancy is insufficient alone to drive the FOXM1 correlated transcriptional program. We show that CENP-F, a component of the outer kinetochore, functions with FOXM1 to coregulate G2/M transcription and proper chromosome segregation. Loss of CENP-F results in alteration of chromatin accessibility at G2/M genes, including CENP-A, and leads to reduced FOXM1-MBB complex formation. The FOXM1-CENP-F transcriptional coordination is a cancer-specific function. We observed that a few CEN/KT genes escape FOXM1 regulation such as CENP-C which when upregulated with CENP-A, leads to increased chromosome misegregation and cell death. Together, we show that the FOXM1 and CENP-F coordinately regulate G2/M gene expression, and this coordination is specific to a subset of genes to allow for proliferation and maintenance of chromosome stability for cancer cell survival.
Collapse
Affiliation(s)
- Sakshi Khurana
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Simpsom Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Daniel R. Foltz
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Simpsom Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
14
|
Gluszek‐Kustusz A, Craske B, Legal T, McHugh T, Welburn JPI. Phosphorylation controls spatial and temporal activities of motor-PRC1 complexes to complete mitosis. EMBO J 2023; 42:e113647. [PMID: 37592895 PMCID: PMC10620760 DOI: 10.15252/embj.2023113647] [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: 01/31/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023] Open
Abstract
During mitosis, spindle architecture alters as chromosomes segregate into daughter cells. The microtubule crosslinker protein regulator of cytokinesis 1 (PRC1) is essential for spindle stability, chromosome segregation and completion of cytokinesis, but how it recruits motors to the central spindle to coordinate the segregation of chromosomes is unknown. Here, we combine structural and cell biology approaches to show that the human CENP-E motor, which is essential for chromosome capture and alignment by microtubules, binds to PRC1 through a conserved hydrophobic motif. This binding mechanism is also used by Kinesin-4 Kif4A:PRC1. Using in vitro reconstitution, we demonstrate that CENP-E slides antiparallel PRC1-crosslinked microtubules. We find that the regulation of CENP-E -PRC1 interaction is spatially and temporally coupled with relocalization to overlapping microtubules in anaphase. Finally, we demonstrate that the PRC1-microtubule motor interaction is essential in anaphase to control chromosome partitioning, retain central spindle integrity and ensure cytokinesis. Taken together our findings reveal the molecular basis for the cell cycle regulation of motor-PRC1 complexes to couple chromosome segregation and cytokinesis.
Collapse
Affiliation(s)
- Agata Gluszek‐Kustusz
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Benjamin Craske
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Thibault Legal
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
- McGill UniversityMontrealQCCanada
| | - Toni McHugh
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Julie PI Welburn
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
15
|
Wong-Dilworth L, Rodilla-Ramirez C, Fox E, Restel SD, Stockhammer A, Adarska P, Bottanelli F. STED imaging of endogenously tagged ARF GTPases reveals their distinct nanoscale localizations. J Cell Biol 2023; 222:e202205107. [PMID: 37102998 PMCID: PMC10140647 DOI: 10.1083/jcb.202205107] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/10/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
ADP-ribosylation factor (ARF) GTPases are major regulators of cellular membrane homeostasis. High sequence similarity and multiple, possibly redundant functions of the five human ARFs make investigating their function a challenging task. To shed light on the roles of the different Golgi-localized ARF members in membrane trafficking, we generated CRISPR-Cas9 knockins (KIs) of type I (ARF1 and ARF3) and type II ARFs (ARF4 and ARF5) and mapped their nanoscale localization with stimulated emission depletion (STED) super-resolution microscopy. We find ARF1, ARF4, and ARF5 on segregated nanodomains on the cis-Golgi and ER-Golgi intermediate compartments (ERGIC), revealing distinct roles in COPI recruitment on early secretory membranes. Interestingly, ARF4 and ARF5 define Golgi-tethered ERGIC elements decorated by COPI and devoid of ARF1. Differential localization of ARF1 and ARF4 on peripheral ERGICs suggests the presence of functionally different classes of intermediate compartments that could regulate bi-directional transport between the ER and the Golgi. Furthermore, ARF1 and ARF3 localize to segregated nanodomains on the trans-Golgi network (TGN) and are found on TGN-derived post-Golgi tubules, strengthening the idea of distinct roles in post-Golgi sorting. This work provides the first map of the nanoscale organization of human ARF GTPases on cellular membranes and sets the stage to dissect their numerous cellular roles.
Collapse
Affiliation(s)
| | | | - Eleanor Fox
- Institut für Biochemie, Freie Universität Berlin, Berlin, Germany
| | | | | | - Petia Adarska
- Institut für Biochemie, Freie Universität Berlin, Berlin, Germany
| | | |
Collapse
|
16
|
Tsang MJ, Cheeseman IM. Alternative CDC20 translational isoforms tune mitotic arrest duration. Nature 2023; 617:154-161. [PMID: 37100900 PMCID: PMC10461078 DOI: 10.1038/s41586-023-05943-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/13/2023] [Indexed: 04/28/2023]
Abstract
Mitotic defects activate the spindle-assembly checkpoint, which inhibits the anaphase-promoting complex co-activator CDC20 to induce a prolonged cell cycle arrest1,2. Once errors are corrected, the spindle-assembly checkpoint is silenced, allowing anaphase onset to occur. However, in the presence of persistent unresolvable errors, cells can undergo 'mitotic slippage', exiting mitosis into a tetraploid G1 state and escaping the cell death that results from a prolonged arrest. The molecular logic that enables cells to balance these duelling mitotic arrest and slippage behaviours remains unclear. Here we demonstrate that human cells modulate the duration of their mitotic arrest through the presence of conserved, alternative CDC20 translational isoforms. Downstream translation initiation results in a truncated CDC20 isoform that is resistant to spindle-assembly-checkpoint-mediated inhibition and promotes mitotic exit even in the presence of mitotic perturbations. Our study supports a model in which the relative levels of CDC20 translational isoforms control the duration of mitotic arrest. During a prolonged mitotic arrest, new protein synthesis and differential CDC20 isoform turnover create a timer, with mitotic exit occurring once the truncated Met43 isoform achieves sufficient levels. Targeted molecular changes or naturally occurring cancer mutations that alter CDC20 isoform ratios or its translational control modulate mitotic arrest duration and anti-mitotic drug sensitivity, with potential implications for the diagnosis and treatment of human cancers.
Collapse
Affiliation(s)
- Mary-Jane Tsang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
17
|
Kanie T, Ng R, Abbott KL, Pongs O, Jackson PK. Myristoylated Neuronal Calcium Sensor-1 captures the ciliary vesicle at distal appendages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.523037. [PMID: 36712037 PMCID: PMC9881967 DOI: 10.1101/2023.01.06.523037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The primary cilium is a microtubule-based organelle that cycles through assembly and disassembly. In many cell types, formation of the cilium is initiated by recruitment of ciliary vesicles to the distal appendage of the mother centriole. However, the distal appendage mechanism that directly captures ciliary vesicles is yet to be identified. In an accompanying paper, we show that the distal appendage protein, CEP89, is important for thef ciliary vesicle recruitment, but not for other steps of cilium formation (Tomoharu Kanie, Love, Fisher, Gustavsson, & Jackson, 2023). The lack of a membrane binding motif in CEP89 suggests that it may indirectly recruit ciliary vesicles via another binding partner. Here, we identify Neuronal Calcium Sensor-1 (NCS1) as a stoichiometric interactor of CEP89. NCS1 localizes to the position between CEP89 and a ciliary vesicle marker, RAB34, at the distal appendage. This localization was completely abolished in CEP89 knockouts, suggesting that CEP89 recruits NCS1 to the distal appendage. Similarly to CEP89 knockouts, ciliary vesicle recruitment as well as subsequent cilium formation was perturbed in NCS1 knockout cells. The ability of NCS1 to recruit the ciliary vesicle is dependent on its myristoylation motif and NCS1 knockout cells expressing myristoylation defective mutant failed to rescue the vesicle recruitment defect despite localizing proper localization to the centriole. In sum, our analysis reveals the first known mechanism for how the distal appendage recruits the ciliary vesicles.
Collapse
Affiliation(s)
- Tomoharu Kanie
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford University, Stanford, CA, 94305
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, OK, 73112
| | - Roy Ng
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford University, Stanford, CA, 94305
| | - Keene L. Abbott
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford University, Stanford, CA, 94305
| | - Olaf Pongs
- Institute for Physiology, Center for Integrative Physiology and Molecular Medicine (CIPPM), Saarland University, Homburg, Germany
| | - Peter K. Jackson
- Baxter Laboratory, Department of Microbiology & Immunology and Department of Pathology, Stanford University, Stanford, CA, 94305
| |
Collapse
|
18
|
Kaja A, Barman P, Guha S, Bhaumik SR. Tandem Affinity Purification and Mass-Spectrometric Analysis of FACT and Associated Proteins. Methods Mol Biol 2023; 2701:209-227. [PMID: 37574485 DOI: 10.1007/978-1-0716-3373-1_14] [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] [Indexed: 08/15/2023]
Abstract
Isolation of a protein/complex is important for its biochemical and structural characterization with mechanistic insights. TAP (tandem affinity purification) strategy allows rapid isolation of cellular proteins/complexes with a high level of purity. This methodology involves an immuno-affinity-based purification followed by a conformation-based isolation to obtain a highly homogeneous protein/complex. Here, we describe the TAP-mediated isolation of endogenous FACT (facilitates chromatin transcription; a heterodimer), an essential histone chaperone associated with BER (base excision repair). However, it is not clearly understood how FACT regulates BER. Such knowledge would advance our understanding of BER with implications in disease pathogenesis, since BER is an evolutionarily conserved process that is linked to various diseases including ageing, neurodegenerative disorders, and cancers. Using isolated FACT by TAP methodology, one can study the mechanisms of action of FACT in BER. Further, isolated FACT can be used for studies in other DNA transactions such as transcription and replication, as FACT is involved in these processes. Furthermore, TAP-mediated isolation strategy can be combined with mass spectrometry to identify the protein interaction partners of FACT.
Collapse
Affiliation(s)
- Amala Kaja
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Priyanka Barman
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Shalini Guha
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Sukesh R Bhaumik
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA.
| |
Collapse
|
19
|
Jung-Garcia Y, Maiques O, Monger J, Rodriguez-Hernandez I, Fanshawe B, Domart MC, Renshaw MJ, Marti RM, Matias-Guiu X, Collinson LM, Sanz-Moreno V, Carlton JG. LAP1 supports nuclear adaptability during constrained melanoma cell migration and invasion. Nat Cell Biol 2023; 25:108-119. [PMID: 36624187 PMCID: PMC9859759 DOI: 10.1038/s41556-022-01042-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2022] [Indexed: 01/11/2023]
Abstract
Metastasis involves dissemination of cancer cells away from a primary tumour and colonization at distal sites. During this process, the mechanical properties of the nucleus must be tuned since they pose a challenge to the negotiation of physical constraints imposed by the microenvironment and tissue structure. We discovered increased expression of the inner nuclear membrane protein LAP1 in metastatic melanoma cells, at the invasive front of human primary melanoma tumours and in metastases. Human cells express two LAP1 isoforms (LAP1B and LAP1C), which differ in their amino terminus. Here, using in vitro and in vivo models that recapitulate human melanoma progression, we found that expression of the shorter isoform, LAP1C, supports nuclear envelope blebbing, constrained migration and invasion by allowing a weaker coupling between the nuclear envelope and the nuclear lamina. We propose that LAP1 renders the nucleus highly adaptable and contributes to melanoma aggressiveness.
Collapse
Affiliation(s)
- Yaiza Jung-Garcia
- Organelle Dynamics Laboratory, The Francis Crick Institute, London, UK
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Oscar Maiques
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Joanne Monger
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
| | - Irene Rodriguez-Hernandez
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Bruce Fanshawe
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Marie-Charlotte Domart
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Matthew J Renshaw
- Advanced Light Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rosa M Marti
- Department of Dermatology, Hospital Universitari Arnau de Vilanova, University of Lleida, IRB Lleida, CIBERONC, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRB Lleida, CIBERONC, Lleida, Spain
| | - Lucy M Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Victoria Sanz-Moreno
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK.
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.
| | - Jeremy G Carlton
- Organelle Dynamics Laboratory, The Francis Crick Institute, London, UK.
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.
| |
Collapse
|
20
|
Funk L, Su KC, Ly J, Feldman D, Singh A, Moodie B, Blainey PC, Cheeseman IM. The phenotypic landscape of essential human genes. Cell 2022; 185:4634-4653.e22. [PMID: 36347254 PMCID: PMC10482496 DOI: 10.1016/j.cell.2022.10.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/01/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022]
Abstract
Understanding the basis for cellular growth, proliferation, and function requires determining the roles of essential genes in diverse cellular processes, including visualizing their contributions to cellular organization and morphology. Here, we combined pooled CRISPR-Cas9-based functional screening of 5,072 fitness-conferring genes in human HeLa cells with microscopy-based imaging of DNA, the DNA damage response, actin, and microtubules. Analysis of >31 million individual cells identified measurable phenotypes for >90% of gene knockouts, implicating gene targets in specific cellular processes. Clustering of phenotypic similarities based on hundreds of quantitative parameters further revealed co-functional genes across diverse cellular activities, providing predictions for gene functions and associations. By conducting pooled live-cell screening of ∼450,000 cell division events for 239 genes, we additionally identified diverse genes with functional contributions to chromosome segregation. Our work establishes a resource detailing the consequences of disrupting core cellular processes that represents the functional landscape of essential human genes.
Collapse
Affiliation(s)
- Luke Funk
- Broad Institute of MIT and Harvard, 415 Main St., Cambridge, MA 02142, USA; Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Kuan-Chung Su
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jimmy Ly
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - David Feldman
- Broad Institute of MIT and Harvard, 415 Main St., Cambridge, MA 02142, USA
| | - Avtar Singh
- Broad Institute of MIT and Harvard, 415 Main St., Cambridge, MA 02142, USA
| | - Brittania Moodie
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Paul C Blainey
- Broad Institute of MIT and Harvard, 415 Main St., Cambridge, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02142, USA.
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
21
|
Navarro AP, Cheeseman IM. Identification of a Golgi-localized peptide reveals a minimal Golgi-targeting motif. Mol Biol Cell 2022; 33:ar110. [PMID: 35921174 DOI: 10.1091/mbc.e22-03-0091] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Prior work has identified signal sequences and motifs that are necessary and sufficient to target proteins to specific subcellular regions and organelles such as the plasma membrane, nucleus, endoplasmic reticulum, and mitochondria. In contrast, minimal sequence motifs that are sufficient for Golgi localization remain largely elusive. In this work, we identified a 37-amino acid alternative open reading frame (altORF) within the mRNA of the centromere protein CENP-R. This altORF peptide localizes specifically to the cytoplasmic surface of the Golgi apparatus. Through mutational analysis, we identify a minimal 10-amino acid sequence and a critical cysteine residue that are necessary and sufficient for Golgi localization. Pharmacological perturbations suggest that this peptide undergoes lipid modification to promote its localization. Together, our work defines a minimal sequence that is sufficient for Golgi targeting and provide a valuable Golgi marker for live cell imaging.
Collapse
Affiliation(s)
- Alexandra P Navarro
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
22
|
Navarro AP, Cheeseman IM. Dynamic cell cycle-dependent phosphorylation modulates CENP-L-CENP-N centromere recruitment. Mol Biol Cell 2022; 33:ar87. [PMID: 35830614 PMCID: PMC9582625 DOI: 10.1091/mbc.e22-06-0239] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The kinetochore is a macromolecular structure that is needed to ensure proper chromosome segregation during each cellular division. The kinetochore is assembled upon a platform of the 16-subunit constitutive centromere-associated network (CCAN), which is present at centromeres throughout the cell cycle. The nature and regulation of CCAN assembly, interactions, and dynamics needed to facilitate changing centromere properties and requirements remain to be fully elucidated. The CENP-LN complex is a CCAN component that displays unique cell cycle–dependent localization behavior, peaking in the S phase. Here, we demonstrate that phosphorylation of CENP-L and CENP-N controls CENP-LN complex formation and localization in a cell cycle–dependent manner. Mimicking constitutive phosphorylation of either CENP-L or CENP-N or simultaneously preventing phosphorylation of both proteins prevents CENP-LN localization and disrupts chromosome segregation. Our work suggests that cycles of phosphorylation and dephosphorylation are critical for CENP-LN complex recruitment and dynamics at kinetochores to enable cell cycle–dependent CCAN reorganization.
Collapse
Affiliation(s)
- Alexandra P Navarro
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
23
|
Holzer E, Rumpf-Kienzl C, Falk S, Dammermann A. A modified TurboID approach identifies tissue-specific centriolar components in C. elegans. PLoS Genet 2022; 18:e1010150. [PMID: 35442950 PMCID: PMC9020716 DOI: 10.1371/journal.pgen.1010150] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/15/2022] [Indexed: 01/26/2023] Open
Abstract
Proximity-dependent labeling approaches such as BioID have been a great boon to studies of protein-protein interactions in the context of cytoskeletal structures such as centrosomes which are poorly amenable to traditional biochemical approaches like immunoprecipitation and tandem affinity purification. Yet, these methods have so far not been applied extensively to invertebrate experimental models such as C. elegans given the long labeling times required for the original promiscuous biotin ligase variant BirA*. Here, we show that the recently developed variant TurboID successfully probes the interactomes of both stably associated (SPD-5) and dynamically localized (PLK-1) centrosomal components. We further develop an indirect proximity labeling method employing a GFP nanobody-TurboID fusion, which allows the identification of protein interactors in a tissue-specific manner in the context of the whole animal. Critically, this approach utilizes available endogenous GFP fusions, avoiding the need to generate multiple additional strains for each target protein and the potential complications associated with overexpressing the protein from transgenes. Using this method, we identify homologs of two highly conserved centriolar components, Cep97 and BLD10/Cep135, which are present in various somatic tissues of the worm. Surprisingly, neither protein is expressed in early embryos, likely explaining why these proteins have escaped attention until now. Our work expands the experimental repertoire for C. elegans and opens the door for further studies of tissue-specific variation in centrosome architecture.
Collapse
Affiliation(s)
- Elisabeth Holzer
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | | | - Sebastian Falk
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | | |
Collapse
|
24
|
Fusi L, Paudel R, Meder K, Schlosser A, Schrama D, Goebeler M, Schmidt M. Interaction of transcription factor FoxO3 with histone acetyltransferase complex subunit TRRAP Modulates Gene Expression and Apoptosis. J Biol Chem 2022; 298:101714. [PMID: 35151693 PMCID: PMC8914384 DOI: 10.1016/j.jbc.2022.101714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/28/2021] [Accepted: 01/19/2022] [Indexed: 11/01/2022] Open
Abstract
Forkhead box O (FoxO) transcription factors are conserved proteins involved in the regulation of life span and age-related diseases, such as diabetes and cancer. Stress stimuli or growth factor deprivation promotes nuclear localization and activation of FoxO proteins, which—depending on the cellular context—can lead to cell cycle arrest or apoptosis. In endothelial cells (ECs), they further regulate angiogenesis and may promote inflammation and vessel destabilization implicating a role of FoxOs in vascular diseases. In several cancers, FoxOs exert a tumor-suppressive function by regulating proliferation and survival. We and others have previously shown that FoxOs can regulate these processes via two different mechanisms: by direct binding to forkhead-responsive elements at the promoter of target genes or by a poorly understood alternative process that does not require direct DNA binding and regulates key targets in primary human ECs. Here, we performed an interaction study in ECs to identify new nuclear FoxO3 interaction partners that might contribute to FoxO-dependent gene regulation. Mass spectrometry analysis of FoxO3-interacting proteins revealed transformation/transcription domain–associated protein (TRRAP), a member of multiple histone acetyltransferase complexes, as a novel binding partner of FoxO family proteins. We demonstrate that TRRAP is required to support FoxO3 transactivation and FoxO3-dependent G1 arrest and apoptosis in ECs via transcriptional activation of the cyclin-dependent kinase inhibitor p27kip1 and the proapoptotic B-cell lymphoma 2 family member, BIM. Moreover, FoxO–TRRAP interaction could explain FoxO-induced alternative gene regulation via TRRAP-dependent recruitment to target promoters lacking forkhead-responsive element sequences.
Collapse
|
25
|
Kumari A, Kumar C, Pergu R, Kumar M, Mahale SP, Wasnik N, Mylavarapu SVS. Phosphorylation and Pin1 binding to the LIC1 subunit selectively regulate mitotic dynein functions. J Cell Biol 2021; 220:212736. [PMID: 34709360 PMCID: PMC8562849 DOI: 10.1083/jcb.202005184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 05/13/2021] [Accepted: 09/22/2021] [Indexed: 01/31/2023] Open
Abstract
The dynein motor performs multiple functions in mitosis by engaging with a wide cargo spectrum. One way to regulate dynein's cargo-binding selectivity is through the C-terminal domain (CTD) of its light intermediate chain 1 subunit (LIC1), which binds directly with cargo adaptors. Here we show that mitotic phosphorylation of LIC1-CTD at its three cdk1 sites is required for proper mitotic progression, for dynein loading onto prometaphase kinetochores, and for spindle assembly checkpoint inactivation in human cells. Mitotic LIC1-CTD phosphorylation also engages the prolyl isomerase Pin1 predominantly to Hook2-dynein-Nde1-Lis1 complexes, but not to dynein-spindly-dynactin complexes. LIC1-CTD dephosphorylation abrogates dynein-Pin1 binding, promotes prophase centrosome-nuclear envelope detachment, and impairs metaphase chromosome congression and mitotic Golgi fragmentation, without affecting interphase membrane transport. Phosphomutation of a conserved LIC1-CTD SP site in zebrafish leads to early developmental defects. Our work reveals that LIC1-CTD phosphorylation differentially regulates distinct mitotic dynein pools and suggests the evolutionary conservation of this phosphoregulation.
Collapse
Affiliation(s)
- Amrita Kumari
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India.,Manipal Academy of Higher Education, Manipal Karnataka, India
| | - Chandan Kumar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India
| | - Rajaiah Pergu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India.,Manipal Academy of Higher Education, Manipal Karnataka, India
| | - Megha Kumar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India.,Council of Scientific and Industrial Research, Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research, New Delhi, India
| | - Sagar P Mahale
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India.,Manipal Academy of Higher Education, Manipal Karnataka, India
| | - Neeraj Wasnik
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India
| | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, third Milestone Faridabad-Gurgaon Expressway, Faridabad Haryana, India.,Manipal Academy of Higher Education, Manipal Karnataka, India
| |
Collapse
|
26
|
Maier NK, Ma J, Lampson MA, Cheeseman IM. Separase cleaves the kinetochore protein Meikin at the meiosis I/II transition. Dev Cell 2021; 56:2192-2206.e8. [PMID: 34331869 PMCID: PMC8355204 DOI: 10.1016/j.devcel.2021.06.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/03/2021] [Accepted: 06/25/2021] [Indexed: 12/19/2022]
Abstract
To generate haploid gametes, germ cells undergo two consecutive meiotic divisions requiring key changes to the cell division machinery. Here, we demonstrate that the protease separase rewires key cell division processes at the meiosis I/II transition by cleaving the meiosis-specific protein Meikin. Separase proteolysis does not inactivate Meikin but instead alters its function to create a distinct activity state. Full-length Meikin and the C-terminal Meikin separase cleavage product both localize to kinetochores, bind to Plk1 kinase, and promote Rec8 cleavage, but our results reveal distinct roles for these proteins in controlling meiosis. Mutations that prevent Meikin cleavage or that conditionally inactivate Meikin at anaphase I result in defective meiosis II chromosome alignment in mouse oocytes. Finally, as oocytes exit meiosis, C-Meikin is eliminated by APC/C-mediated degradation prior to the first mitotic division. Thus, multiple regulatory events irreversibly modulate Meikin activity during successive meiotic divisions to rewire the cell division machinery at two distinct transitions.
Collapse
Affiliation(s)
- Nolan K Maier
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jun Ma
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
27
|
Hwang SH, Somatilaka BN, White K, Mukhopadhyay S. Ciliary and extraciliary Gpr161 pools repress hedgehog signaling in a tissue-specific manner. eLife 2021; 10:67121. [PMID: 34346313 PMCID: PMC8378848 DOI: 10.7554/elife.67121] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022] Open
Abstract
The role of compartmentalized signaling in primary cilia during tissue morphogenesis is not well understood. The cilia localized G protein-coupled receptor, Gpr161, represses hedgehog pathway via cAMP signaling. We engineered a knock-in at the Gpr161 locus in mice to generate a variant (Gpr161mut1), which was ciliary localization defective but cAMP signaling competent. Tissue phenotypes from hedgehog signaling depend on downstream bifunctional Gli transcriptional factors functioning as activators or repressors. Compared to knockout (ko), Gpr161mut1/ko had delayed embryonic lethality, moderately increased hedgehog targets, and partially down-regulated Gli3 repressor. Unlike ko, the Gpr161mut1/ko neural tube did not show Gli2 activator-dependent expansion of ventral-most progenitors. Instead, the intermediate neural tube showed progenitor expansion that depends on loss of Gli3 repressor. Increased extraciliary receptor levels in Gpr161mut1/mut1 prevented ventralization. Morphogenesis in limb buds and midface requires Gli repressor; these tissues in Gpr161mut1/mut1 manifested hedgehog hyperactivation phenotypes—polydactyly and midfacial widening. Thus, ciliary and extraciliary Gpr161 pools likely establish tissue-specific Gli repressor thresholds in determining morpho-phenotypic outcomes.
Collapse
Affiliation(s)
- Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Bandarigoda N Somatilaka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kevin White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
28
|
Gatta AT, Olmos Y, Stoten CL, Chen Q, Rosenthal PB, Carlton JG. CDK1 controls CHMP7-dependent nuclear envelope reformation. eLife 2021; 10:59999. [PMID: 34286694 PMCID: PMC8324300 DOI: 10.7554/elife.59999] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Through membrane sealing and disassembly of spindle microtubules, the Endosomal Sorting Complex Required for Transport-III (ESCRT-III) machinery has emerged as a key player in the regeneration of a sealed nuclear envelope (NE) during mitotic exit, and in the repair of this organelle during interphase rupture. ESCRT-III assembly at the NE occurs transiently during mitotic (M) exit and is initiated when CHMP7, an ER-localised ESCRT-II/ESCRT-III hybrid protein, interacts with the Inner Nuclear Membrane (INM) protein LEM2. Whilst classical nucleocytoplasmic transport mechanisms have been proposed to separate LEM2 and CHMP7 during interphase, it is unclear how CHMP7 assembly is suppressed in mitosis when NE and ER identities are mixed. Here, we use live cell imaging and protein biochemistry to examine the biology of these proteins during M-exit. Firstly, we show that CHMP7 plays an important role in the dissolution of LEM2 clusters that form at the NE during M-exit. Secondly, we show that CDK1 phosphorylates CHMP7 upon M-entry at Ser3 and Ser441 and that this phosphorylation reduces CHMP7's interaction with LEM2, limiting its assembly during M-phase. We show that spatiotemporal differences in the dephosphorylation of CHMP7 license its assembly at the NE during telophase, but restrict its assembly on the ER at this time. Without CDK1 phosphorylation, CHMP7 undergoes inappropriate assembly in the peripheral ER during M-exit, capturing LEM2 and downstream ESCRT-III components. Lastly, we establish that a microtubule network is dispensable for ESCRT-III assembly at the reforming nuclear envelope. These data identify a key cell-cycle control programme allowing ESCRT-III-dependent nuclear regeneration.
Collapse
Affiliation(s)
- Alberto T Gatta
- School of Cancer and Pharmaceutical Sciences, King's College, London, United Kingdom.,Organelle Dynamics Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Yolanda Olmos
- School of Cancer and Pharmaceutical Sciences, King's College, London, United Kingdom.,Organelle Dynamics Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Caroline L Stoten
- School of Cancer and Pharmaceutical Sciences, King's College, London, United Kingdom.,Organelle Dynamics Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Qu Chen
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Peter B Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Jeremy G Carlton
- School of Cancer and Pharmaceutical Sciences, King's College, London, United Kingdom.,Organelle Dynamics Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
29
|
Gabriel L, Srinivasan B, Kuś K, Mata JF, João Amorim M, Jansen LET, Athanasiadis A. Enrichment of Zα domains at cytoplasmic stress granules is due to their innate ability to bind to nucleic acids. J Cell Sci 2021; 134:268376. [PMID: 34037233 DOI: 10.1242/jcs.258446] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/08/2021] [Indexed: 01/14/2023] Open
Abstract
Zα domains recognize the left-handed helical Z conformation of double-stranded nucleic acids. They are found in proteins involved in the nucleic acid sensory pathway of the vertebrate innate immune system and host evasion by viral pathogens. Previously, it has been demonstrated that ADAR1 (encoded by ADAR in humans) and DAI (also known as ZBP1) localize to cytoplasmic stress granules (SGs), and this localization is mediated by their Zα domains. To investigate the mechanism, we determined the interactions and localization pattern for the N-terminal region of human DAI (ZαβDAI), which harbours two Zα domains, and for a ZαβDAI mutant deficient in nucleic acid binding. Electrophoretic mobility shift assays demonstrated the ability of ZαβDAI to bind to hyperedited nucleic acids, which are enriched in SGs. Furthermore, using immunofluorescence and immunoprecipitation coupled with mass spectrometry, we identified several interacting partners of the ZαβDAI-RNA complex in vivo under conditions of arsenite-induced stress. These interactions are lost upon loss of nucleic acid-binding ability or upon RNase treatment. Thus, we posit that the mechanism for the translocation of Zα domain-containing proteins to SGs is mainly mediated by the nucleic acid-binding ability of their Zα domains. This article has an associated First Person interview with Bharath Srinivasan, joint first author of the paper.
Collapse
Affiliation(s)
- Luisa Gabriel
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| | - Bharath Srinivasan
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| | - Krzysztof Kuś
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| | - João F Mata
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| | - Maria João Amorim
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| | - Lars E T Jansen
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| | - Alekos Athanasiadis
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, Oeiras 2781-156, Portugal
| |
Collapse
|
30
|
Nguyen AL, Fadel MD, Cheeseman IM. Differential requirements for the CENP-O complex reveal parallel PLK1 kinetochore recruitment pathways. Mol Biol Cell 2021; 32:712-721. [PMID: 33596090 PMCID: PMC8108507 DOI: 10.1091/mbc.e20-11-0751] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 01/09/2023] Open
Abstract
Similar to other core biological processes, the vast majority of cell division components are essential for viability across human cell lines. However, recent genome-wide screens have identified a number of proteins that exhibit cell line-specific essentiality. Defining the behaviors of these proteins is critical to our understanding of complex biological processes. Here, we harness differential essentiality to reveal the contributions of the four-subunit centromere-localized CENP-O complex, whose precise function has been difficult to define. Our results support a model in which the CENP-O complex and BUB1 act in parallel pathways to recruit a threshold level of PLK1 to mitotic kinetochores, ensuring accurate chromosome segregation. We demonstrate that targeted changes to either pathway sensitizes cells to the loss of the other component, resulting in cell-state dependent requirements. This approach also highlights the advantage of comparing phenotypes across diverse cell lines to define critical functional contributions and behaviors that could be exploited for the targeted treatment of disease.
Collapse
Affiliation(s)
| | - Marie Diane Fadel
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
31
|
SOGA1 and SOGA2/MTCL1 are CLASP-interacting proteins required for faithful chromosome segregation in human cells. Chromosome Res 2021; 29:159-173. [PMID: 33587225 DOI: 10.1007/s10577-021-09651-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 10/22/2022]
Abstract
CLASPs are key modulators of microtubule dynamics throughout the cell cycle. During mitosis, CLASPs independently associate with growing microtubule plus-ends and kinetochores and play essential roles in chromosome segregation. In a proteomic survey for human CLASP1-interacting proteins during mitosis, we have previously identified SOGA1 and SOGA2/MTCL1, whose mitotic roles remained uncharacterized. Here we performed an initial functional characterization of human SOGA1 and SOGA2/MTCL1 during mitosis. Using specific polyclonal antibodies raised against SOGA proteins, we confirmed their expression and reciprocal interaction with CLASP1 and CLASP2 during mitosis. In addition, we found that both SOGA1 and SOGA2/MTCL1 are phospho-regulated during mitosis by CDK1. Immunofluorescence analysis revealed that SOGA2/MTCL1 co-localizes with mitotic spindle microtubules and spindle poles throughout mitosis and both SOGA proteins are enriched at the midbody during mitotic exit/cytokinesis. GFP-tagging of SOGA2/MTCL1 further revealed a microtubule-independent localization at kinetochores. Live-cell imaging after siRNA-mediated knockdown of SOGA1 and SOGA2/MTCL1 showed that they are independently required for distinct aspects of chromosome segregation. Thus, SOGA1 and SOGA2/MTCL1 are bona fide CLASP-interacting proteins during mitosis required for faithful chromosome segregation in human cells.
Collapse
|
32
|
Chen Z, Chen J. Mass spectrometry-based protein‒protein interaction techniques and their applications in studies of DNA damage repair. J Zhejiang Univ Sci B 2021; 22:1-20. [PMID: 33448183 PMCID: PMC7818012 DOI: 10.1631/jzus.b2000356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
Abstract
Proteins are major functional units that are tightly connected to form complex and dynamic networks. These networks enable cells and organisms to operate properly and respond efficiently to environmental cues. Over the past decades, many biochemical methods have been developed to search for protein-binding partners in order to understand how protein networks are constructed and connected. At the same time, rapid development in proteomics and mass spectrometry (MS) techniques makes it possible to identify interacting proteins and build comprehensive protein‒protein interaction networks. The resulting interactomes and networks have proven informative in the investigation of biological functions, such as in the field of DNA damage repair. In recent years, a number of proteins involved in DNA damage response and DNA repair pathways have been uncovered with MS-based protein‒protein interaction studies. As the technologies for enriching associated proteins and MS become more sophisticated, the studies of protein‒protein interactions are entering a new era. In this review, we summarize the strategies and recent developments for exploring protein‒protein interaction. In addition, we discuss the application of these tools in the investigation of protein‒protein interaction networks involved in DNA damage response and DNA repair.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Sreevalsan S, Döring M, Paszkowski-Rogacz M, Brux M, Blanck C, Meyer M, Momburg F, Buchholz F, Theis M. MLLT6 maintains PD-L1 expression and mediates tumor immune resistance. EMBO Rep 2020; 21:e50155. [PMID: 33063451 PMCID: PMC7726806 DOI: 10.15252/embr.202050155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 12/26/2022] Open
Abstract
Tumor cells subvert immune surveillance by harnessing signals from immune checkpoints to acquire immune resistance. The protein PD‐L1 is an important component in this process, and inhibition of PD‐L1 elicits durable anti‐tumor responses in a broad spectrum of cancers. However, immune checkpoint inhibition that target known pathways is not universally effective. A better understanding of the genetic repertoire underlying these processes is necessary to expand our knowledge in tumor immunity and to facilitate identification of alternative targets. Here, we present a CRISPR/Cas9 screen in human cancer cells to identify genes that confer tumors with the ability to evade the cytotoxic effects of the immune system. We show that the transcriptional regulator MLLT6 (AF17) is required for efficient PD‐L1 protein expression and cell surface presentation in cancer cells. MLLT6 depletion alleviates suppression of CD8+ cytotoxic T cell‐mediated cytolysis. Furthermore, cancer cells lacking MLLT6 exhibit impaired STAT1 signaling and are insensitive to interferon‐γ‐induced stimulation of IDO1, GBP5, CD74, and MHC class II genes. Collectively, our findings establish MLLT6 as a regulator of oncogenic and interferon‐γ‐associated immune resistance.
Collapse
Affiliation(s)
- Sandeep Sreevalsan
- National Center for Tumor Diseases (NCT/UCC) Dresden, German Cancer Research Center (DKFZ), University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Marietta Döring
- National Center for Tumor Diseases (NCT/UCC) Dresden, German Cancer Research Center (DKFZ), University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Maciej Paszkowski-Rogacz
- Medical Systems Biology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Melanie Brux
- National Center for Tumor Diseases (NCT/UCC) Dresden, German Cancer Research Center (DKFZ), University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Carolina Blanck
- Medical Systems Biology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marten Meyer
- Antigen Presentation & T/NK Cell Activation Group, Clinical Cooperation Unit 'Applied Tumor Immunity', German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Momburg
- Antigen Presentation & T/NK Cell Activation Group, Clinical Cooperation Unit 'Applied Tumor Immunity', German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Buchholz
- National Center for Tumor Diseases (NCT/UCC) Dresden, German Cancer Research Center (DKFZ), University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Medical Systems Biology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mirko Theis
- National Center for Tumor Diseases (NCT/UCC) Dresden, German Cancer Research Center (DKFZ), University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Medical Systems Biology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
34
|
Dou Y, Kalmykova S, Pashkova M, Oghbaie M, Jiang H, Molloy KR, Chait BT, Rout MP, Fenyö D, Jensen TH, Altukhov I, LaCava J. Affinity proteomic dissection of the human nuclear cap-binding complex interactome. Nucleic Acids Res 2020; 48:10456-10469. [PMID: 32960270 PMCID: PMC7544204 DOI: 10.1093/nar/gkaa743] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
A 5′,7-methylguanosine cap is a quintessential feature of RNA polymerase II-transcribed RNAs, and a textbook aspect of co-transcriptional RNA processing. The cap is bound by the cap-binding complex (CBC), canonically consisting of nuclear cap-binding proteins 1 and 2 (NCBP1/2). Interest in the CBC has recently renewed due to its participation in RNA-fate decisions via interactions with RNA productive factors as well as with adapters of the degradative RNA exosome. A novel cap-binding protein, NCBP3, was recently proposed to form an alternative CBC together with NCBP1, and to interact with the canonical CBC along with the protein SRRT. The theme of post-transcriptional RNA fate, and how it relates to co-transcriptional ribonucleoprotein assembly, is abundant with complicated, ambiguous, and likely incomplete models. In an effort to clarify the compositions of NCBP1-, 2- and 3-related macromolecular assemblies, we have applied an affinity capture-based interactome screen where the experimental design and data processing have been modified to quantitatively identify interactome differences between targets under a range of experimental conditions. This study generated a comprehensive view of NCBP-protein interactions in the ribonucleoprotein context and demonstrates the potential of our approach to benefit the interpretation of complex biological pathways.
Collapse
Affiliation(s)
- Yuhui Dou
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Maria Pashkova
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Mehrnoosh Oghbaie
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, USA.,European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hua Jiang
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, USA
| | - Kelly R Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, USA
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, NYU Langone Health, New York, USA
| | - Torben Heick Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Ilya Altukhov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - John LaCava
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, USA.,European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
35
|
Legal T, Hayward D, Gluszek-Kustusz A, Blackburn EA, Spanos C, Rappsilber J, Gruneberg U, Welburn JPI. The C-terminal helix of BubR1 is essential for CENP-E-dependent chromosome alignment. J Cell Sci 2020; 133:jcs246025. [PMID: 32665320 PMCID: PMC7473641 DOI: 10.1242/jcs.246025] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
During cell division, misaligned chromosomes are captured and aligned by motors before their segregation. The CENP-E motor is recruited to polar unattached kinetochores to facilitate chromosome alignment. The spindle checkpoint protein BubR1 (also known as BUB1B) has been reported as a CENP-E interacting partner, but the extent to which BubR1 contributes to CENP-E localization at kinetochores has remained controversial. Here we define the molecular determinants that specify the interaction between BubR1 and CENP-E. The basic C-terminal helix of BubR1 is necessary but not sufficient for CENP-E interaction, and a minimal key acidic patch on the kinetochore-targeting domain of CENP-E is also essential. We then demonstrate that BubR1 is required for the recruitment of CENP-E to kinetochores to facilitate chromosome alignment. This BubR1-CENP-E axis is critical for alignment of chromosomes that have failed to congress through other pathways and recapitulates the major known function of CENP-E. Overall, our studies define the molecular basis and the function for CENP-E recruitment to BubR1 at kinetochores during mammalian mitosis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Thibault Legal
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Agata Gluszek-Kustusz
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Elizabeth A Blackburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Christos Spanos
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Juri Rappsilber
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
- Chair of Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin 10623, Germany
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| |
Collapse
|
36
|
Jain Goyal M, Zhao X, Bozhinova M, Andrade-López K, de Heus C, Schulze-Dramac S, Müller-McNicoll M, Klumperman J, Béthune J. A paralog-specific role of COPI vesicles in the neuronal differentiation of mouse pluripotent cells. Life Sci Alliance 2020; 3:3/9/e202000714. [PMID: 32665377 PMCID: PMC7368096 DOI: 10.26508/lsa.202000714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 12/03/2022] Open
Abstract
The paralogous COPI coat subunit γ1-COP plays a unique role in promoting neurite outgrowth during the neuronal differentiation of mouse pluripotent cells. Coat protein complex I (COPI)–coated vesicles mediate membrane trafficking between Golgi cisternae as well as retrieval of proteins from the Golgi to the endoplasmic reticulum. There are several flavors of the COPI coat defined by paralogous subunits of the protein complex coatomer. However, whether paralogous COPI proteins have specific functions is currently unknown. Here, we show that the paralogous coatomer subunits γ1-COP and γ2-COP are differentially expressed during the neuronal differentiation of mouse pluripotent cells. Moreover, through a combination of genome editing experiments, we demonstrate that whereas γ-COP paralogs are largely functionally redundant, γ1-COP specifically promotes neurite outgrowth. Our work stresses a role of the COPI pathway in neuronal polarization and provides evidence for distinct functions for coatomer paralogous subunits in this process.
Collapse
Affiliation(s)
- Manu Jain Goyal
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Xiyan Zhao
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Mariya Bozhinova
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Karla Andrade-López
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Cecilia de Heus
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sandra Schulze-Dramac
- RNA Regulation Group, Cluster of Excellence "Macromolecular Complexes," Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Michaela Müller-McNicoll
- RNA Regulation Group, Cluster of Excellence "Macromolecular Complexes," Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Julien Béthune
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany .,Heidelberg University Biochemistry Center, Heidelberg, Germany
| |
Collapse
|
37
|
Kabrawala S, Zimmer MD, Campellone KG. WHIMP links the actin nucleation machinery to Src-family kinase signaling during protrusion and motility. PLoS Genet 2020; 16:e1008694. [PMID: 32196488 PMCID: PMC7112243 DOI: 10.1371/journal.pgen.1008694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/01/2020] [Accepted: 02/22/2020] [Indexed: 12/27/2022] Open
Abstract
Cell motility is governed by cooperation between the Arp2/3 complex and nucleation-promoting factors from the Wiskott-Aldrich Syndrome Protein (WASP) family, which together assemble actin filament networks to drive membrane protrusion. Here we identify WHIMP (WAVE Homology In Membrane Protrusions) as a new member of the WASP family. The Whimp gene is encoded on the X chromosome of a subset of mammals, including mice. Murine WHIMP promotes Arp2/3-dependent actin assembly, but is less potent than other nucleation factors. Nevertheless, WHIMP-mediated Arp2/3 activation enhances both plasma membrane ruffling and wound healing migration, whereas WHIMP depletion impairs protrusion and slows motility. WHIMP expression also increases Src-family kinase activity, and WHIMP-induced ruffles contain the additional nucleation-promoting factors WAVE1, WAVE2, and N-WASP, but not JMY or WASH. Perturbing the function of Src-family kinases, WAVE proteins, or Arp2/3 complex inhibits WHIMP-driven ruffling. These results suggest that WHIMP-associated actin assembly plays a direct role in membrane protrusion, but also results in feedback control of tyrosine kinase signaling to modulate the activation of multiple WASP-family members. The actin cytoskeleton is a collection of protein polymers that assemble and disassemble within cells at specific times and locations. Sophisticated cytoskeletal regulators called nucleation-promoting factors ensure that actin polymerizes when and where it is needed, and many of these factors are members of the Wiskott-Aldrich Syndrome Protein (WASP) family. Several of the 8 known WASP-family proteins function in cell motility, but how the different factors collaborate with one another is not well understood. In this study, we identified WHIMP, a new WASP-family member that is encoded on the X chromosome of a variety of mammals. In mouse cells, WHIMP enhances cell motility by assembling actin filaments that push the plasma membrane forward. Unexpectedly, WHIMP also activates tyrosine kinases, enzymes that stimulate multiple WASP-family members during motility. Our results open new avenues of research into how nucleation factors cooperate during movement and how the molecular activities that underlie motility differ in distinct cell types and organisms.
Collapse
Affiliation(s)
- Shail Kabrawala
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Margaret D. Zimmer
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Kenneth G. Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
38
|
Pizon V, Gaudin N, Poteau M, Cifuentes-Diaz C, Demdou R, Heyer V, Reina San Martin B, Azimzadeh J. hVFL3/CCDC61 is a component of mother centriole subdistal appendages required for centrosome cohesion and positioning. Biol Cell 2019; 112:22-37. [PMID: 31789463 DOI: 10.1111/boc.201900038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The centrosome regulates cell spatial organisation by controlling the architecture of the microtubule (MT) cytoskeleton. Conversely, the position of the centrosome within the cell depends on cytoskeletal networks it helps organizing. In mammalian cells, centrosome positioning involves a population of MT stably anchored at centrioles, the core components of the centrosome. An MT-anchoring complex containing the proteins ninein and Cep170 is enriched at subdistal appendages (SAP) that decorate the older centriole (called mother centriole) and at centriole proximal ends. Here, we studied the role played at the centrosome by hVFL3/CCDC61, the human ortholog of proteins required for anchoring distinct sets of cytoskeletal fibres to centrioles in unicellular eukaryotes. RESULTS We show that hVFL3 co-localises at SAP and at centriole proximal ends with components of the MT-anchoring complex, and physically interacts with Cep170. Depletion of hVFL3 increased the distance between mother and daughter centrioles without affecting the assembly of a filamentous linker that tethers the centrioles and contains the proteins rootletin and C-Nap1. When the linker was disrupted by inactivating C-Nap1, hVFL3-depletion exacerbated centriole splitting, a phenotype also observed following depletion of other SAP components. This supported that hVFL3 is required for SAP function, which we further established by showing that centrosome positioning is perturbed in hVFL3-depleted interphase cells. Finally, we found that hVFL3 is an MT-binding protein. CONCLUSIONS AND SIGNIFICANCE Together, our results support that hVFL3 is required for anchoring MT at SAP during interphase and ensuring proper centrosome cohesion and positioning. The role of the VFL3 family of proteins thus appears to have been conserved in evolution despite the great variation in the shape of centriole appendages in different eukaryotic species.
Collapse
Affiliation(s)
- Véronique Pizon
- Université de Paris, Institut Jacques Monod, 75013, Paris, France
| | - Noémie Gaudin
- Université de Paris, Institut Jacques Monod, 75013, Paris, France
| | - Marion Poteau
- Institut Gustave Roussy, CNRS UMR 8200/Université Paris-Sud, 94 805, Villejuif, France
| | | | - Roland Demdou
- Université de Paris, Institut Jacques Monod, 75013, Paris, France
| | - Vincent Heyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Bernardo Reina San Martin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
39
|
Bonner MK, Haase J, Swinderman J, Halas H, Miller Jenkins LM, Kelly AE. Enrichment of Aurora B kinase at the inner kinetochore controls outer kinetochore assembly. J Cell Biol 2019; 218:3237-3257. [PMID: 31527147 PMCID: PMC6781445 DOI: 10.1083/jcb.201901004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/19/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022] Open
Abstract
Outer kinetochore assembly enables chromosome attachment to microtubules and spindle assembly checkpoint (SAC) signaling in mitosis. Aurora B kinase controls kinetochore assembly by phosphorylating the Mis12 complex (Mis12C) subunit Dsn1. Current models propose Dsn1 phosphorylation relieves autoinhibition, allowing Mis12C binding to inner kinetochore component CENP-C. Using Xenopus laevis egg extracts and biochemical reconstitution, we found that autoinhibition of the Mis12C by Dsn1 impedes its phosphorylation by Aurora B. Our data indicate that the INCENP central region increases Dsn1 phosphorylation by enriching Aurora B at inner kinetochores, close to CENP-C. Furthermore, centromere-bound CENP-C does not exchange in mitosis, and CENP-C binding to the Mis12C dramatically increases Dsn1 phosphorylation by Aurora B. We propose that the coincidence of Aurora B and CENP-C at inner kinetochores ensures the fidelity of kinetochore assembly. We also found that the central region is required for the SAC beyond its role in kinetochore assembly, suggesting that kinetochore enrichment of Aurora B promotes the phosphorylation of other kinetochore substrates.
Collapse
Affiliation(s)
- Mary Kate Bonner
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Julian Haase
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jason Swinderman
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hyunmi Halas
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alexander E Kelly
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
40
|
Reja SI, Minoshima M, Hori Y, Kikuchi K. Development of an effective protein-labeling system based on smart fluorogenic probes. J Biol Inorg Chem 2019; 24:443-455. [PMID: 31152238 DOI: 10.1007/s00775-019-01669-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 12/23/2022]
Abstract
Proteins are an important component of living systems and play a crucial role in various physiological functions. Fluorescence imaging of proteins is a powerful tool for monitoring protein dynamics. Fluorescent protein (FP)-based labeling methods are frequently used to monitor the movement and interaction of cellular proteins. However, alternative methods have also been developed that allow the use of synthetic fluorescent probes to target a protein of interest (POI). Synthetic fluorescent probes have various advantages over FP-based labeling methods. They are smaller in size than the fluorescent proteins, offer a wide variety of colors and have improved photochemical properties. There are various chemical recognition-based labeling techniques that can be used for labeling a POI with a synthetic probe. In this review, we focus on the development of protein-labeling systems, particularly the SNAP-tag, BL-tag, and PYP-tag systems, and understanding the fluorescence behavior of the fluorescently labeled target protein in these systems. We also discuss the smart fluorogenic probes for these protein-labeling systems and their applications. The fluorogenic protein labeling will be a useful tool to investigate complex biological phenomena in future work on cell biology.
Collapse
Affiliation(s)
- Shahi Imam Reja
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masafumi Minoshima
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Hori
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazuya Kikuchi
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan.
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
41
|
Knocking in Multifunctional Gene Tags into SMC Complex Subunits Using Gene Editing. Methods Mol Biol 2019. [PMID: 31147912 DOI: 10.1007/978-1-4939-9520-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Condensin, a highly conserved pentameric chromosome complex, is required for the correct organization and folding of the genome. Here, we highlight how to knock protein tags into endogenous loci to faithfully study the condensin complex in vertebrates and dissect its multiple functions. These include using the streptavidin binding peptide (SBP) to create the first genome-wide map of condensin and perform varied applications in proteomics and enzymology of the complex. The revolution in gene editing using CRISPR/Cas9 has made it possible to insert tags into endogenous loci with relative ease, allowing physiological and fully functional tagged protein to be analyzed biochemically (affinity tags), microscopically (fluorescent tags) or both purified and localized (multifunctional tags). In this chapter, we detail how to engineer vertebrate cells using CRISPR/Cas9 to provide researchers powerful tools to obtain greater precision than ever to understand how the complex interacts and behaves in cells.
Collapse
|
42
|
Lin CH, Wang Z, Duque-Afonso J, Wong SHK, Demeter J, Loktev AV, Somervaille TCP, Jackson PK, Cleary ML. Oligomeric self-association contributes to E2A-PBX1-mediated oncogenesis. Sci Rep 2019; 9:4915. [PMID: 30894657 PMCID: PMC6426973 DOI: 10.1038/s41598-019-41393-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
Abstract
The PBX1 homeodomain transcription factor is converted by t(1;19) chromosomal translocations in acute leukemia into the chimeric E2A-PBX1 oncoprotein. Fusion with E2A confers potent transcriptional activation and constitutive nuclear localization, bypassing the need for dimerization with protein partners that normally stabilize and regulate import of PBX1 into the nucleus, but the mechanisms underlying its oncogenic activation are incompletely defined. We demonstrate here that E2A-PBX1 self-associates through the PBX1 PBC-B domain of the chimeric protein to form higher-order oligomers in t(1;19) human leukemia cells, and that this property is required for oncogenic activity. Structural and functional studies indicate that self-association facilitates the binding of E2A-PBX1 to DNA. Mutants unable to self-associate are transformation defective, however their oncogenic activity is rescued by the synthetic oligomerization domain of FKBP, which confers conditional transformation properties on E2A-PBX1. In contrast to self-association, PBX1 protein domains that mediate interactions with HOX DNA-binding partners are dispensable. These studies suggest that oligomeric self-association may compensate for the inability of monomeric E2A-PBX1 to stably bind DNA and circumvents protein interactions that otherwise modulate PBX1 stability, nuclear localization, DNA binding, and transcriptional activity. The unique dependence on self-association for E2A-PBX1 oncogenic activity suggests potential approaches for mechanism-based targeted therapies.
Collapse
MESH Headings
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Carcinogenesis/genetics
- Cell Line, Tumor
- Chromosomes, Human, Pair 1/chemistry
- Chromosomes, Human, Pair 19/chemistry
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Gene Expression Regulation, Neoplastic
- HEK293 Cells
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Pre-B-Cell Leukemia Transcription Factor 1/genetics
- Pre-B-Cell Leukemia Transcription Factor 1/metabolism
- Protein Binding
- Protein Multimerization
- Protein Stability
- Tacrolimus Binding Proteins/genetics
- Tacrolimus Binding Proteins/metabolism
- Transcription, Genetic
- Translocation, Genetic
Collapse
Affiliation(s)
- Chiou-Hong Lin
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Zhong Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Sun Yat-Sen University, School of Pharmaceutical Sciences, Guangzhou, 510006, China
| | - Jesús Duque-Afonso
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Hematology and Oncology, University Medical Center Freiburg, Freiburg, Germany
| | - Stephen Hon-Kit Wong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Janos Demeter
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alexander V Loktev
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tim C P Somervaille
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4GJ, UK
| | - Peter K Jackson
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael L Cleary
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
43
|
Remnant L, Booth DG, Vargiu G, Spanos C, Kerr ARW, Earnshaw WC. In vitro BioID: mapping the CENP-A microenvironment with high temporal and spatial resolution. Mol Biol Cell 2019; 30:1314-1325. [PMID: 30892990 PMCID: PMC6724601 DOI: 10.1091/mbc.e18-12-0799] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The centromere is located at the primary constriction of condensed chromosomes where it acts as a platform regulating chromosome segregation. The histone H3 variant CENP-A is the foundation for kinetochore formation. CENP-A directs the formation of a highly dynamic molecular neighborhood whose temporal characterization during mitosis remains a challenge due to limitations in available techniques. BioID is a method that exploits a “promiscuous” biotin ligase (BirA118R or BirA*) to identify proteins within close proximity to a fusion protein of interest. As originally described, cells expressing BirA* fusions were exposed to high biotin concentrations for 24 h during which the ligase transferred activated biotin (BioAmp) to other proteins within the immediate vicinity. The protein neighborhood could then be characterized by streptavidin-based purification and mass spectrometry. Here we describe a further development to this technique, allowing CENP-A interactors to be characterized within only a few minutes, in an in vitro reaction in lysed cells whose physiological progression is “frozen.” This approach, termed in vitro BioID (ivBioID), has the potential to study the molecular neighborhood of any structural protein whose interactions change either during the cell cycle or in response to other changes in cell physiology.
Collapse
Affiliation(s)
- Lucy Remnant
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology and
| | - Daniel G Booth
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology and.,Centre for Brain Discovery Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Giulia Vargiu
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology and
| | - Christos Spanos
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology and
| | - Alastair R W Kerr
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology and
| | | |
Collapse
|
44
|
Pergu R, Dagar S, Kumar H, Kumar R, Bhattacharya J, Mylavarapu SVS. The chaperone ERp29 is required for tunneling nanotube formation by stabilizing MSec. J Biol Chem 2019; 294:7177-7193. [PMID: 30877198 DOI: 10.1074/jbc.ra118.005659] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/14/2019] [Indexed: 01/23/2023] Open
Abstract
Tunneling nanotubes (TNTs) are membrane conduits that mediate long-distance intercellular cross-talk in several organisms and play vital roles during development, pathogenic transmission, and cancer metastasis. However, the molecular mechanisms of TNT formation and function remain poorly understood. The protein MSec (also known as TNFα-induced protein 2 (TNFAIP2) and B94) is essential for TNT formation in multiple cell types. Here, using affinity protein purification, mass spectrometric identification, and confocal immunofluorescence microscopy assays, we found that MSec interacts with the endoplasmic reticulum (ER) chaperone ERp29. siRNA-mediated ERp29 depletion in mammalian cells significantly reduces TNT formation, whereas its overexpression induces TNT formation, but in a strictly MSec-dependent manner. ERp29 stabilized MSec protein levels, but not its mRNA levels, and the chaperone activity of ERp29 was required for maintaining MSec protein stability. Subcellular ER fractionation and subsequent limited proteolytic treatment suggested that MSec is associated with the outer surface of the ER. The ERp29-MSec interaction appeared to require the presence of other bridging protein(s), perhaps triggered by post-translational modification of ERp29. Our study implicates MSec as a target of ERp29 and reveals an indispensable role for the ER in TNT formation, suggesting new modalities for regulating TNT numbers in cells and tissues.
Collapse
Affiliation(s)
- Rajaiah Pergu
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and.,the Manipal Academy of Higher Education, Manipal Karnataka 576104, and
| | - Sunayana Dagar
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and.,the Kalinga Institute of Industrial Technology, Bhubaneswar Odisha 751024, India
| | - Harsh Kumar
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and.,the Manipal Academy of Higher Education, Manipal Karnataka 576104, and
| | - Rajesh Kumar
- the HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad Haryana 121001
| | - Jayanta Bhattacharya
- the HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad Haryana 121001
| | - Sivaram V S Mylavarapu
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and .,the Manipal Academy of Higher Education, Manipal Karnataka 576104, and.,the Kalinga Institute of Industrial Technology, Bhubaneswar Odisha 751024, India
| |
Collapse
|
45
|
Remmelzwaal S, Boxem M. Protein interactome mapping in Caenorhabditis elegans. CURRENT OPINION IN SYSTEMS BIOLOGY 2019; 13:1-9. [PMID: 32984658 PMCID: PMC7493430 DOI: 10.1016/j.coisb.2018.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The systematic identification of all protein-protein interactions that take place in an organism (the 'interactome') is an important goal in modern biology. The nematode Caenorhabditis elegans was one of the first multicellular models for which a proteome-wide interactome mapping project was initiated. Most Caenorhabditis elegans interactome mapping efforts have utilized the yeast two-hybrid system, yielding an extensive binary interactome, while recent developments in mass spectrometry-based approaches hold great potential for further improving our understanding of protein interactome networks in a multicellular context. For example, methods like co-fractionation, proximity labeling, and tissue-specific protein purification not only identify protein-protein interactions, but have the potential to provide crucial insight into when and where interactions take place. Here we review current standards and recent improvements in protein interaction mapping in C. elegans.
Collapse
Affiliation(s)
- Sanne Remmelzwaal
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Mike Boxem
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| |
Collapse
|
46
|
Chen C, Whitney IP, Banerjee A, Sacristan C, Sekhri P, Kern DM, Fontan A, Kops GJPL, Tyson JJ, Cheeseman IM, Joglekar AP. Ectopic Activation of the Spindle Assembly Checkpoint Signaling Cascade Reveals Its Biochemical Design. Curr Biol 2018; 29:104-119.e10. [PMID: 30595520 DOI: 10.1016/j.cub.2018.11.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 11/27/2022]
Abstract
Switch-like activation of the spindle assembly checkpoint (SAC) is critical for accurate chromosome segregation and for cell division in a timely manner. To determine the mechanisms that achieve this, we engineered an ectopic, kinetochore-independent SAC activator: the "eSAC." The eSAC stimulates SAC signaling by artificially dimerizing Mps1 kinase domain and a cytosolic KNL1 phosphodomain, the kinetochore signaling scaffold. By exploiting variable eSAC expression in a cell population, we defined the dependence of the eSAC-induced mitotic delay on eSAC concentration in a cell to reveal the dose-response behavior of the core signaling cascade of the SAC. These quantitative analyses and subsequent mathematical modeling of the dose-response data uncover two crucial properties of the core SAC signaling cascade: (1) a cellular limit on the maximum anaphase-inhibitory signal that the cascade can generate due to the limited supply of SAC proteins and (2) the ability of the KNL1 phosphodomain to produce the anaphase-inhibitory signal synergistically, when it recruits multiple SAC proteins simultaneously. We propose that these properties together achieve inverse, non-linear scaling between the signal output per kinetochore and the number of signaling kinetochores. When the number of kinetochores is low, synergistic signaling by KNL1 enables each kinetochore to produce a disproportionately strong signal output. However, when many kinetochores signal concurrently, they compete for a limited supply of SAC proteins. This frustrates synergistic signaling and lowers their signal output. Thus, the signaling activity of unattached kinetochores will adapt to the changing number of signaling kinetochores to enable the SAC to approximate switch-like behavior.
Collapse
Affiliation(s)
- Chu Chen
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ian P Whitney
- Whitehead Institute for Biomedical Research and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Anand Banerjee
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Carlos Sacristan
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences), and Molecular Cancer Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Palak Sekhri
- Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David M Kern
- Whitehead Institute for Biomedical Research and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Adrienne Fontan
- Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Geert J P L Kops
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences), and Molecular Cancer Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Ajit P Joglekar
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA; Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
47
|
Monda JK, Cheeseman IM. Dynamic regulation of dynein localization revealed by small molecule inhibitors of ubiquitination enzymes. Open Biol 2018; 8:rsob.180095. [PMID: 30257893 PMCID: PMC6170511 DOI: 10.1098/rsob.180095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/29/2018] [Indexed: 11/27/2022] Open
Abstract
Cytoplasmic dynein is a minus-end-directed microtubule-based motor that acts at diverse subcellular sites. During mitosis, dynein localizes simultaneously to the mitotic spindle, spindle poles, kinetochores and the cell cortex. However, it is unclear what controls the relative targeting of dynein to these locations. As dynein is heavily post-translationally modified, we sought to test a role for these modifications in regulating dynein localization. We find that dynein rapidly and strongly accumulates at mitotic spindle poles following treatment with NSC697923, a small molecule that inhibits the ubiquitin E2 enzyme, Ubc13, or treatment with PYR-41, a ubiquitin E1 inhibitor. Subsets of dynein regulators such as Lis1, ZW10 and Spindly accumulate at the spindle poles, whereas others do not, suggesting that NSC697923 differentially affects specific dynein populations. We additionally find that dynein relocalization induced by NSC697923 or PYR-41 can be suppressed by simultaneous treatment with the non-selective deubiquitinase inhibitor, PR-619. However, we did not observe altered dynein localization following treatment with the selective E1 inhibitor, TAK-243. Although it is possible that off-target effects of NSC697923 and PYR-41 are responsible for the observed changes in dynein localization, the rapid relocalization upon drug treatment highlights the highly dynamic nature of dynein regulation during mitosis.
Collapse
Affiliation(s)
- Julie K Monda
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
48
|
Delcourt V, Brunelle M, Roy AV, Jacques JF, Salzet M, Fournier I, Roucou X. The Protein Coded by a Short Open Reading Frame, Not by the Annotated Coding Sequence, Is the Main Gene Product of the Dual-Coding Gene MIEF1. Mol Cell Proteomics 2018; 17:2402-2411. [PMID: 30181344 PMCID: PMC6283296 DOI: 10.1074/mcp.ra118.000593] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 07/19/2018] [Indexed: 12/18/2022] Open
Abstract
Proteogenomics and ribosome profiling concurrently show that genes may code for both a large and one or more small proteins translated from annotated coding sequences (CDSs) and unannotated alternative open reading frames (named alternative ORFs or altORFs), respectively, but the stoichiometry between large and small proteins translated from a same gene is unknown. MIEF1, a gene recently identified as a dual-coding gene, harbors a CDS and a newly annotated and actively translated altORF located in the 5′UTR. Here, we use absolute quantification with stable isotope-labeled peptides and parallel reaction monitoring to determine levels of both proteins in two human cells lines and in human colon. We report that the main MIEF1 translational product is not the canonical 463 amino acid MiD51 protein but the small 70 amino acid alternative MiD51 protein (altMiD51). These results demonstrate the inadequacy of the single CDS concept and provide a strong argument for incorporating altORFs and small proteins in functional annotations.
Collapse
Affiliation(s)
- Vivian Delcourt
- Département de Biochimie, Université de Sherbrooke, Québec, Canada; Univ. Lille, INSERM U1192, Laboratoire Protéomique, Réponse Inflammatoire and Spectrométrie de Masse (PRISM) F-59000 Lille, France; PROTEO, Québec Network for Research on Protein Function, Structure, and Engineering, Québec, Canada
| | - Mylène Brunelle
- Département de Biochimie, Université de Sherbrooke, Québec, Canada; PROTEO, Québec Network for Research on Protein Function, Structure, and Engineering, Québec, Canada
| | - Annie V Roy
- Département de Biochimie, Université de Sherbrooke, Québec, Canada; PROTEO, Québec Network for Research on Protein Function, Structure, and Engineering, Québec, Canada
| | - Jean-François Jacques
- Département de Biochimie, Université de Sherbrooke, Québec, Canada; PROTEO, Québec Network for Research on Protein Function, Structure, and Engineering, Québec, Canada
| | - Michel Salzet
- Univ. Lille, INSERM U1192, Laboratoire Protéomique, Réponse Inflammatoire and Spectrométrie de Masse (PRISM) F-59000 Lille, France
| | - Isabelle Fournier
- Univ. Lille, INSERM U1192, Laboratoire Protéomique, Réponse Inflammatoire and Spectrométrie de Masse (PRISM) F-59000 Lille, France
| | - Xavier Roucou
- Département de Biochimie, Université de Sherbrooke, Québec, Canada; PROTEO, Québec Network for Research on Protein Function, Structure, and Engineering, Québec, Canada.
| |
Collapse
|
49
|
Pires HR, Boxem M. Mapping the Polarity Interactome. J Mol Biol 2018; 430:3521-3544. [DOI: 10.1016/j.jmb.2017.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
|
50
|
Ivanov MP, Ladurner R, Poser I, Beveridge R, Rampler E, Hudecz O, Novatchkova M, Hériché JK, Wutz G, van der Lelij P, Kreidl E, Hutchins JR, Axelsson-Ekker H, Ellenberg J, Hyman AA, Mechtler K, Peters JM. The replicative helicase MCM recruits cohesin acetyltransferase ESCO2 to mediate centromeric sister chromatid cohesion. EMBO J 2018; 37:e97150. [PMID: 29930102 PMCID: PMC6068434 DOI: 10.15252/embj.201797150] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 02/27/2018] [Accepted: 04/09/2018] [Indexed: 11/09/2022] Open
Abstract
Chromosome segregation depends on sister chromatid cohesion which is established by cohesin during DNA replication. Cohesive cohesin complexes become acetylated to prevent their precocious release by WAPL before cells have reached mitosis. To obtain insight into how DNA replication, cohesion establishment and cohesin acetylation are coordinated, we analysed the interaction partners of 55 human proteins implicated in these processes by mass spectrometry. This proteomic screen revealed that on chromatin the cohesin acetyltransferase ESCO2 associates with the MCM2-7 subcomplex of the replicative Cdc45-MCM-GINS helicase. The analysis of ESCO2 mutants defective in MCM binding indicates that these interactions are required for proper recruitment of ESCO2 to chromatin, cohesin acetylation during DNA replication, and centromeric cohesion. We propose that MCM binding enables ESCO2 to travel with replisomes to acetylate cohesive cohesin complexes in the vicinity of replication forks so that these complexes can be protected from precocious release by WAPL Our results also indicate that ESCO1 and ESCO2 have distinct functions in maintaining cohesion between chromosome arms and centromeres, respectively.
Collapse
Affiliation(s)
| | - Rene Ladurner
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Evelyn Rampler
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Otto Hudecz
- Institute of Molecular Biotechnology, Vienna, Austria
| | | | | | - Gordana Wutz
- Research Institute of Molecular Pathology, Vienna, Austria
| | | | - Emanuel Kreidl
- Research Institute of Molecular Pathology, Vienna, Austria
| | | | | | - Jan Ellenberg
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Karl Mechtler
- Research Institute of Molecular Pathology, Vienna, Austria
- Institute of Molecular Biotechnology, Vienna, Austria
| | | |
Collapse
|