1
|
Lozano-Elena F, Wendeborn S. The role and structure of molecular glues in plant signalling networks. Nat Rev Chem 2025:10.1038/s41570-025-00717-3. [PMID: 40355685 DOI: 10.1038/s41570-025-00717-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 05/14/2025]
Abstract
Protein-protein interactions are one of the pillars of all life processes. Many signalling molecules work by promoting and stabilizing these interactions. These molecular 'glues' bind simultaneously to two proteins inducing their interaction, which would be otherwise less favourable or non-favourable. Importantly, they can be harnessed for a clinical purpose, but, despite advances in medicine, the wealth of natural molecular glues in plants have only rarely been commercially utilized. These molecular glues may be plant-endogenous or plant-exogenous small molecules or peptides, and they may be involved in many different processes, such as growth promotion or stress response, opening new opportunities for crop protection, along with other applications. In this Review, we analyse the underlying structural motives and molecular interactions in detail, classifying the modes of actions based on their nature (small ligands versus peptides) and receptor classes. We discuss both natural metabolites and mimetics of such compounds, highlighting similarities and differences between signalling pathways and comparing them with relevant mechanisms in mammals.
Collapse
Affiliation(s)
- Fidel Lozano-Elena
- Institute for Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Sebastian Wendeborn
- Institute for Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland.
| |
Collapse
|
2
|
Singh MI, Rajendraprasad G, Katopodis V, Cui R, Barisic M, Bhowmick R, Hickson ID. Mechanistic insight into anaphase bridge signaling to the abscission checkpoint. EMBO J 2025:10.1038/s44318-025-00453-w. [PMID: 40355560 DOI: 10.1038/s44318-025-00453-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
During cytokinesis in human cells, a failure to resolve persistent DNA bridges that span the cell-division plane maintains the Aurora B-dependent abscission checkpoint in an active state. However, the molecular mechanism by which unresolved sister-chromatid bridging signals to this checkpoint is poorly defined. Here, we define an essential role for the Bloom's syndrome helicase, BLM, in signaling to the abscission-checkpoint machinery in response to replication stress through the conversion of dsDNA bridges into RPA-coated ssDNA. RPA then promotes ATR-CHK1 signaling to Aurora B, utilizing a kinase cascade shared with the S-phase checkpoint. BLM-deficient cells ultimately abandon cytokinesis in response to replication stress, which promotes binucleation and hence aneuploidy. Considering that aneuploidy is a hallmark of cancer, we propose that this role for BLM in cytokinesis is a plausible reason for cancer predisposition in Bloom's syndrome individuals. Consistent with this, BLM deficiency promotes anchorage-independent growth of non-cancer cells.
Collapse
Affiliation(s)
- Manika I Singh
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
- Centre for Genomic Medicine, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Girish Rajendraprasad
- Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen N, Denmark
| | - Vasileios Katopodis
- Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen N, Denmark
| | - Rui Cui
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Marin Barisic
- Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen N, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Rahul Bhowmick
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA.
| | - Ian D Hickson
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark.
| |
Collapse
|
3
|
Yaschenko AE, Alonso JM, Stepanova AN. Arabidopsis as a model for translational research. THE PLANT CELL 2025; 37:koae065. [PMID: 38411602 PMCID: PMC12082644 DOI: 10.1093/plcell/koae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/28/2024]
Abstract
Arabidopsis thaliana is currently the most-studied plant species on earth, with an unprecedented number of genetic, genomic, and molecular resources having been generated in this plant model. In the era of translating foundational discoveries to crops and beyond, we aimed to highlight the utility and challenges of using Arabidopsis as a reference for applied plant biology research, agricultural innovation, biotechnology, and medicine. We hope that this review will inspire the next generation of plant biologists to continue leveraging Arabidopsis as a robust and convenient experimental system to address fundamental and applied questions in biology. We aim to encourage laboratory and field scientists alike to take advantage of the vast Arabidopsis datasets, annotations, germplasm, constructs, methods, and molecular and computational tools in our pursuit to advance understanding of plant biology and help feed the world's growing population. We envision that the power of Arabidopsis-inspired biotechnologies and foundational discoveries will continue to fuel the development of resilient, high-yielding, nutritious plants for the betterment of plant and animal health and greater environmental sustainability.
Collapse
Affiliation(s)
- Anna E Yaschenko
- Department of Plant and Microbial Biology, Genetics and Genomics Academy, North Carolina State University, Raleigh, NC 27695, USA
| | - Jose M Alonso
- Department of Plant and Microbial Biology, Genetics and Genomics Academy, North Carolina State University, Raleigh, NC 27695, USA
| | - Anna N Stepanova
- Department of Plant and Microbial Biology, Genetics and Genomics Academy, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
4
|
Matsumoto S, Kogure Y, Ono S, Numata T, Endo T. Msp1 and Pex19-Pex3 cooperate to achieve correct localization of Pex15 to peroxisomes. FEBS J 2025. [PMID: 40344504 DOI: 10.1111/febs.70132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/14/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Yeast Msp1 is a dual-localized AAA-ATPase on the mitochondrial outer membrane (OM) and peroxisomal membrane. We previously showed that Msp1 transfers mistargeted tail-anchored (TA) proteins from mitochondria to the endoplasmic reticulum (ER) for degradation or delivery to their original destinations. However, the mechanism by which Msp1 in mitochondria and peroxisomes handles authentic peroxisomal TA proteins remains unclear. We show that newly synthesized Pex15 is targeted to peroxisomes primarily via the Pex19- and Pex3-dependent pathway. Mistargeted Pex15 on the mitochondrial OM is extracted by mitochondrial Msp1 and transferred to the ER via the guided-entry of TA proteins pathway for degradation or to peroxisomes via the Pex19-Pex3 pathway. Intriguingly, endogenous Pex15 localized in peroxisomes is also extracted from the membranes by peroxisomal Msp1 but returns to peroxisomes via the Pex19-Pex3 pathway. These results suggest that correct Pex15 localization to peroxisomes relies on not only the initial targeting by Pex19-Pex3 but also the constant re-routing by Msp1 and Pex19-Pex3.
Collapse
Affiliation(s)
- Shunsuke Matsumoto
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiki Kogure
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Suzuka Ono
- Faculty of Life Sciences, Kyoto Sangyo University, Japan
- Institute for Protein Dynamics, Kyoto Sangyo University, Japan
| | - Tomoyuki Numata
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiya Endo
- Faculty of Life Sciences, Kyoto Sangyo University, Japan
- Institute for Protein Dynamics, Kyoto Sangyo University, Japan
| |
Collapse
|
5
|
Butterfield GL, Reisman SJ, Iglesias N, Gersbach CA. Gene regulation technologies for gene and cell therapy. Mol Ther 2025; 33:2104-2122. [PMID: 40195118 DOI: 10.1016/j.ymthe.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
Gene therapy stands at the forefront of medical innovation, offering unique potential to treat the underlying causes of genetic disorders and broadly enable regenerative medicine. However, unregulated production of therapeutic genes can lead to decreased clinical utility due to various complications. Thus, many technologies for controlled gene expression are under development, including regulated transgenes, modulation of endogenous genes to leverage native biological regulation, mapping and repurposing of transcriptional regulatory networks, and engineered systems that dynamically react to cell state changes. Transformative therapies enabled by advances in tissue-specific promoters, inducible systems, and targeted delivery have already entered clinical testing and demonstrated significantly improved specificity and efficacy. This review highlights next-generation technologies under development to expand the reach of gene therapies by enabling precise modulation of gene expression. These technologies, including epigenome editing, antisense oligonucleotides, RNA editing, transcription factor-mediated reprogramming, and synthetic genetic circuits, have the potential to provide powerful control over cellular functions. Despite these remarkable achievements, challenges remain in optimizing delivery, minimizing off-target effects, and addressing regulatory hurdles. However, the ongoing integration of biological insights with engineering innovations promises to expand the potential for gene therapy, offering hope for treating not only rare genetic disorders but also complex multifactorial diseases.
Collapse
Affiliation(s)
- Gabriel L Butterfield
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Samuel J Reisman
- Department of Cell Biology, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Nahid Iglesias
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
6
|
Schweighofer J, Mulay B, Hoffmann I, Vogt D, Pesenti ME, Musacchio A. Interactions with multiple inner kinetochore proteins determine mitotic localization of FACT. J Cell Biol 2025; 224:e202412042. [PMID: 40094435 PMCID: PMC11912937 DOI: 10.1083/jcb.202412042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
The FAcilitates Chromatin Transcription (FACT) complex is a dimeric histone chaperone that operates on chromatin during transcription and replication. FACT also interacts with a specialized centromeric nucleosome containing the histone H3 variant centromere protein A (CENP-A) and with CENP-TW, two subunits of the constitutive centromere-associated network (CCAN), a 16-protein complex associated with CENP-A. The significance of these interactions remains elusive. Here, we show that FACT has multiple additional binding sites on CCAN. The interaction with CCAN is strongly stimulated by casein kinase II phosphorylation of FACT. Mitotic localization of FACT to kinetochores is strictly dependent on specific CCAN subcomplexes. Conversely, CENP-TW requires FACT for stable localization. Unexpectedly, we also find that DNA readily displaces FACT from CCAN, supporting the speculation that FACT becomes recruited through a pool of CCAN that is not stably integrated into chromatin. Collectively, our results point to a potential role of FACT in chaperoning CCAN during transcription or in the stabilization of CCAN at the centromere during the cell cycle.
Collapse
Affiliation(s)
- Julia Schweighofer
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Bhagyashree Mulay
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Ingrid Hoffmann
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Doro Vogt
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marion E. Pesenti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- Max Planck School Matter to Life, Heidelberg, Germany
| |
Collapse
|
7
|
Mahendrawada L, Warfield L, Donczew R, Hahn S. Low overlap of transcription factor DNA binding and regulatory targets. Nature 2025:10.1038/s41586-025-08916-0. [PMID: 40240607 DOI: 10.1038/s41586-025-08916-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/19/2025] [Indexed: 04/18/2025]
Abstract
DNA sequence-specific transcription factors (TFs) modulate transcription and chromatin architecture, acting from regulatory sites in enhancers and promoters of eukaryotic genes1,2. How multiple TFs cooperate to regulate individual genes is still unclear. In yeast, most TFs are thought to regulate transcription via binding to upstream activating sequences, which are situated within a few hundred base pairs upstream of the regulated gene3. Although this model has been validated for individual TFs and specific genes, it has not been tested in a systematic way. Here we integrated information on the binding and expression targets for the near-complete set of yeast TFs and show that, contrary to expectations, there are few TFs with dedicated activator or repressor roles, and that most TFs have a dual function. Although nearly all protein-coding genes are regulated by one or more TFs, our analysis revealed limited overlap between TF binding and gene regulation. Rapid depletion of many TFs also revealed many regulatory targets that were distant from detectable TF binding sites, suggesting unexpected regulatory mechanisms. Our study provides a comprehensive survey of TF functions and offers insights into interactions between the set of TFs expressed in a single cell type and how they contribute to the complex programme of gene regulation.
Collapse
Affiliation(s)
| | | | - Rafal Donczew
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Steven Hahn
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
8
|
Rojas-Pierce M, Bednarek SY. Manipulation of targeted protein degradation in plant biology. Biochem Soc Trans 2025; 53:BST20230939. [PMID: 40209052 DOI: 10.1042/bst20230939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
Inducible protein degradation systems are an important but untapped resource for the study of protein function in plant cells. Unlike mutagenesis or transcriptional control, regulated degradation of proteins of interest allows the study of the biological mechanisms of highly dynamic cellular processes involving essential proteins. While systems for targeted protein degradation are available for research and therapeutics in animals, there are currently limited options in plant biology. Targeted protein degradation systems rely on target ubiquitination by E3 ubiquitin ligases. Systems that are available or being developed in plants can be distinguished primarily by the type of E3 ubiquitin ligase involved, including those that utilize Cullin-RING ligases, bacterial novel E3 ligases, and N-end rule pathway E3 ligases, or they can be controlled by proteolysis targeting chimeras. Target protein ubiquitination leads to degradation by the proteasome or targeting to the vacuole, with both pathways being ubiquitous and important for the endogenous control of protein abundance in plants. Targeted proteolysis approaches for plants will likely be an important tool for basic research and to yield novel traits for crop biotechnology.
Collapse
Affiliation(s)
- Marcela Rojas-Pierce
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, U.S.A
| | | |
Collapse
|
9
|
Huang MY, Nalley MJ, Hecht P, Madhani HD. An auxin-inducible degron system for conditional mutation in the fungal meningitis pathogen Cryptococcus neoformans. G3 (BETHESDA, MD.) 2025:jkaf071. [PMID: 40194515 DOI: 10.1093/g3journal/jkaf071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/27/2025] [Indexed: 04/09/2025]
Abstract
Cryptococcus neoformans is the top-ranked W.H.O. fungal priority pathogen, but tools for generating conditional mutations are limited. Auxin-inducible degron systems permit rapid and effective cellular depletion of a tagged protein of interest upon adding a small molecule. These tools are invaluable, particularly for studying essential genes, which may play important roles in pathogen biology. AID2 is one such system that improves on previous strategies. This system achieves greater sensitivity and specificity through an auxin derivative, 5-Ph-IAA, alongside an OsTIR1F74G mutant. We adapted the AID2 system for C. neoformans by codon optimizing OsTIR1F74G and tested its use in multiple scenarios. We demonstrate that the C. neoformans optimized AID2 system enables effective degradation of proteins, including essential proteins, and can be used to help discriminate essential from non-essential genes. This tool enables the study of unexplored parts of the C. neoformans genome.
Collapse
Affiliation(s)
- Manning Y Huang
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Matthew J Nalley
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Patrick Hecht
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Hiten D Madhani
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
10
|
Parikh SJ, Terron HM, Burgard LA, Maranan DS, Butler DD, Wiseman A, LaFerla FM, Lane S, Leissring MA. Targeted Control of Gene Expression Using CRISPR-Associated Endoribonucleases. Cells 2025; 14:543. [PMID: 40214496 PMCID: PMC11988398 DOI: 10.3390/cells14070543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
CRISPR-associated endoribonucleases (Cas RNases) cleave single-stranded RNA in a highly sequence-specific manner by recognizing and binding to short RNA sequences known as direct repeats (DRs). Here, we investigate the potential of exploiting Cas RNases for the regulation of target genes with one or more DRs introduced into the 3' untranslated region, an approach we refer to as DREDGE (direct repeat-enabled downregulation of gene expression). The DNase-dead version of Cas12a (dCas12a) was identified as the most efficient among five different Cas RNases tested and was subsequently evaluated in doxycycline-regulatable systems targeting either stably expressed fluorescent proteins or an endogenous gene. DREDGE performed superbly in stable cell lines, resulting in up to 90% downregulation with rapid onset, notably in a fully reversible and highly selective manner. Successful control of an endogenous gene with DREDGE was demonstrated in two formats, including one wherein both the DR and the transgene driving expression of dCas12a were introduced in one step by CRISPR-Cas. Our results establish DREDGE as an effective method for regulating gene expression in a targeted, highly selective, and fully reversible manner, with several advantages over existing technologies.
Collapse
Affiliation(s)
- Sagar J. Parikh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Heather M. Terron
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Luke A. Burgard
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Derek S. Maranan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Dylan D. Butler
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Abigail Wiseman
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Shelley Lane
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Malcolm A. Leissring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Camlin NJ. Protein-targeting reverse genetic approaches: the future of oocyte and preimplantation embryo research. Mol Hum Reprod 2025; 31:gaaf008. [PMID: 40100642 PMCID: PMC12000532 DOI: 10.1093/molehr/gaaf008] [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: 11/22/2024] [Revised: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Reverse genetic approaches are the standard in molecular biology to determine a protein's function. Traditionally, nucleic acid targeting via gene knockout (DNA) and knockdown (RNA) has been the method of choice to remove proteins-of-interest. However, the nature of mammalian oocyte maturation and preimplantation embryo development can make nucleic acid-targeting approaches difficult. Gene knockout allows time for compensatory mechanisms and secondary phenotypes to develop which can make interpretation of a protein's function difficult. Furthermore, genes can be essential for animal and/or oocyte survival, and therefore, gene knockout is not always a viable approach to investigate oocyte maturation and preimplantation embryo development. Conversely, RNA-targeting approaches, i.e. RNA interference (RNAi) and morpholinos, rely on protein half-life and therefore are unable to knockdown every protein-of-interest. An increasing number of reverse genetic approaches that directly target proteins have been developed to overcome the limitations of nucleic acid-based approaches, including Trim-Away and auxin-inducible degradation. These protein-targeting approaches give researchers exquisite and fast control of protein loss. This review will discuss how Trim-Away and auxin-inducible degradation can overcome many of the challenges of nucleic acid-based reverse genetic approaches. Furthermore, it highlights the unique research opportunities these approaches afford, such as targeting post-translationally modified proteins.
Collapse
Affiliation(s)
- Nicole J Camlin
- Cell and Molecular Biology, School of Biological, Environmental and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, USA
| |
Collapse
|
12
|
Masłowska K, Wong R, Ulrich H, Pagès V. Post-replicative lesion processing limits DNA damage-induced mutagenesis. Nucleic Acids Res 2025; 53:gkaf198. [PMID: 40114379 PMCID: PMC11925729 DOI: 10.1093/nar/gkaf198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
DNA lesions are a threat to genome stability. To cope with them during DNA replication, cells have evolved lesion bypass mechanisms: Translesion Synthesis (TLS), which allows the cell to insert a nucleotide directly opposite the lesion, with the risk of introducing a mutation, and error-free damage avoidance (DA), which uses homologous recombination to retrieve the genetic information from the sister chromatid. In this study, we investigate the timing of lesion bypass in yeast and its implications for the accuracy of the process. Our findings reveal that DNA polymerase η can bypass common, UV-induced cyclobutane pyrimidine dimers at the fork, immediately after encountering the blocking lesion. In contrast, TLS at (6-4) photoproducts and bulky G-AAF adducts, mediated by Rev1 and Pol ζ, takes place behind the fork, at post-replicative gaps that are generated downstream of the lesion after repriming. We show that in this latter situation, TLS competes with the DA pathway, thus reducing overall mutagenicity of damage bypass. Additionally, our study demonstrates that Exo1 nuclease influences the balance between TLS and DA by modulating the size of the post-replicative gaps.
Collapse
Affiliation(s)
- Katarzyna H Masłowska
- Cancer Research Center of Marseille: Team DNA Damage and Genome Instability. CNRS, Aix Marseille University, Inserm, Institut Paoli-Calmettes, Marseille 13009, France
| | - Ronald P Wong
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany
| | - Helle D Ulrich
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany
| | - Vincent Pagès
- Cancer Research Center of Marseille: Team DNA Damage and Genome Instability. CNRS, Aix Marseille University, Inserm, Institut Paoli-Calmettes, Marseille 13009, France
| |
Collapse
|
13
|
Ge T, Brickner DG, Zehr K, VanBelzen DJ, Zhang W, Caffalette C, Moeller GC, Ungerleider S, Marcou N, Jacob A, Nguyen VQ, Chait B, Rout MP, Brickner JH. Exportin-1 functions as an adaptor for transcription factor-mediated docking of chromatin at the nuclear pore complex. Mol Cell 2025; 85:1101-1116.e8. [PMID: 40068679 PMCID: PMC11928253 DOI: 10.1016/j.molcel.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025]
Abstract
Nuclear pore proteins (nucleoporins [Nups]) physically interact with hundreds of chromosomal sites, impacting transcription. In yeast, transcription factors mediate interactions between Nups and enhancers and promoters. To define the molecular basis of this mechanism, we exploited a separation-of-function mutation in the Gcn4 transcription factor that blocks its interaction with the nuclear pore complex (NPC). This mutation reduces the interaction of Gcn4 with the highly conserved nuclear export factor Crm1/Xpo1. Crm1 and Nups co-occupy enhancers, and Crm1 inhibition blocks interaction of the nuclear pore protein Nup2 with the genome. In vivo, Crm1 interacts stably with the NPC and in vitro, Crm1 binds directly to both Gcn4 and Nup2. Importantly, the interaction between Crm1 and Gcn4 requires neither Ran-guanosine triphosphate (GTP) nor the nuclear export sequence binding site. Finally, Crm1 and Ran-GTP stimulate DNA binding by Gcn4, suggesting that allosteric coupling between Crm1-Ran-GTP binding and DNA binding facilitates the docking of transcription-factor-bound enhancers at the NPC.
Collapse
Affiliation(s)
- Tiffany Ge
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - Donna Garvey Brickner
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - Kara Zehr
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - D Jake VanBelzen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - Wenzhu Zhang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065, USA
| | - Christopher Caffalette
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Gavin C Moeller
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Sara Ungerleider
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - Nikita Marcou
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - Alexis Jacob
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA
| | - Vu Q Nguyen
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Brian Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Jason H Brickner
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60201, USA.
| |
Collapse
|
14
|
Latham AP, Zhang W, Tempkin JOB, Otsuka S, Ellenberg J, Sali A. Integrative spatiotemporal modeling of biomolecular processes: Application to the assembly of the nuclear pore complex. Proc Natl Acad Sci U S A 2025; 122:e2415674122. [PMID: 40085653 PMCID: PMC11929490 DOI: 10.1073/pnas.2415674122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Dynamic processes involving biomolecules are essential for the function of the cell. Here, we introduce an integrative method for computing models of these processes based on multiple heterogeneous sources of information, including time-resolved experimental data and physical models of dynamic processes. First, for each time point, a set of coarse models of compositional and structural heterogeneity is computed (heterogeneity models). Second, for each heterogeneity model, a set of static integrative structure models is computed (a snapshot model). Finally, these snapshot models are selected and connected into a series of trajectories that optimize the likelihood of both the snapshot models and transitions between them (a trajectory model). The method is demonstrated by application to the assembly process of the human nuclear pore complex in the context of the reforming nuclear envelope during mitotic cell division, based on live-cell correlated electron tomography, bulk fluorescence correlation spectroscopy-calibrated quantitative live imaging, and a structural model of the fully assembled nuclear pore complex. Modeling of the assembly process improves the model precision over static integrative structure modeling alone. The method is applicable to a wide range of time-dependent systems in cell biology and is available to the broader scientific community through an implementation in the open source Integrative Modeling Platform (IMP) software.
Collapse
Affiliation(s)
- Andrew P. Latham
- Department of Bioengineering and Therapeutic Sciences, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
- Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
| | - Wanlu Zhang
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg69117, Germany
| | - Jeremy O. B. Tempkin
- Department of Bioengineering and Therapeutic Sciences, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
- Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
| | - Shotaro Otsuka
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg69117, Germany
| | - Jan Ellenberg
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg69117, Germany
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
- Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
| |
Collapse
|
15
|
Jennrich J, Farkas Á, Urlaub H, Schwappach B, Bohnsack KE. The formation of chaperone-rich GET bodies depends on the tetratricopeptide repeat region of Sgt2 and is reversed by NADH. J Cell Sci 2025; 138:jcs263616. [PMID: 39976550 PMCID: PMC11959614 DOI: 10.1242/jcs.263616] [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: 10/18/2024] [Accepted: 01/30/2025] [Indexed: 03/21/2025] Open
Abstract
The guided-entry of tail-anchored proteins (GET) pathway is a post-translational targeting route to the endoplasmic reticulum (ER). Upon glucose withdrawal, the soluble GET proteins re-localize to dynamic cytosolic foci, here termed GET bodies. Our data reveal that the pre-targeting complex components, Sgt2 and the Get4-Get5 heterodimer, and the Get3 ATPase play important roles in the assembly of these structures in Saccharomyces cerevisiae. More specifically, the TPR region of Sgt2 is required as a GET body scaffold. Systematic compositional analyses of GET bodies reveal their chaperone-rich nature and the presence of numerous proteins involved in metabolic processes. Temporal analyses of GET body assembly demonstrate the sequential recruitment of different chaperones, and we discover the requirement of Sis1 and Sti1 for maintaining the dynamic properties of these structures. In vivo, NADH derived from the oxidation of ethanol to acetaldehyde can induce GET body disassembly in a reaction depending on the alcohol dehydrogenase Adh2 and in vitro, addition of NADH resolves GET bodies. This suggests a mechanistic basis for their formation and disassembly in response to the metabolic shift caused by glucose withdrawal and re-addition.
Collapse
Affiliation(s)
- Jonas Jennrich
- Department of Molecular Biology, University Medical Centre Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Ákos Farkas
- Department of Molecular Biology, University Medical Centre Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Henning Urlaub
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Faßberg 11, 37077 Göttingen, Germany
- Institute for Clinical Chemistry, University Medical Centre Göttingen, Robert-Koch-Straße 40, 35075 Göttingen, Germany
| | - Blanche Schwappach
- Department of Molecular Biology, University Medical Centre Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Katherine E. Bohnsack
- Department of Molecular Biology, University Medical Centre Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| |
Collapse
|
16
|
Zhang Y, Chen Y, Wu C, Cai Z, Yao W, Yang H, Song J, Xie X, Zhang L, Yi C. Establishment of a yeast essential protein conditional-degradation library and screening for autophagy-regulating genes. Autophagy 2025:1-13. [PMID: 39988731 DOI: 10.1080/15548627.2025.2469189] [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: 07/15/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved intracellular degradation pathway that relies on vacuoles or lysosomes. Over 40 ATG genes have been identified in yeast cells as participants in various types of autophagy, although these genes are non-essential. While some essential genes involved in autophagy have been identified using temperature-sensitive yeast strains, systematic research on essential genes in autophagy remains lacking. To address this, we established an essential protein conditional degradation library using the auxin-inducible degron (AID) system. By introducing the GFP-Atg8 plasmid, we identified 29 essential yeast genes involved in autophagy, 19 of which had not been previously recognized. In summary, the yeast essential protein conditional degradation library we constructed will serve as a valuable resource for systematically investigating the roles of essential genes in autophagy and other biological functions.Abbreviation: AID: auxin-inducible degron; ALP: alkaline phosphatase; ATG: autophagy related; CSG: constitutive slow growth; DAmP: Decreased Abundance by mRNA Perturbation; GFP: green fluorescent protein; MMS: methyl methanesulfonate; ORF: open reading frame; PAS: phagophore assembly site; PCR: polymerase chain reaction; SD-G: glucose starvation medium; SD-N: nitrogen starvation medium; TOR: target of rapamycin kinase; YGRC: yeast genetic resource center; YPD: yeast extract peptone dextrose.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yingcong Chen
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Choufei Wu
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Zhengyi Cai
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Weijing Yao
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huan Yang
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Song
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Xiankuan Xie
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University school of Medicine, Hangzhou, Zhejiang, China
| | - Liqin Zhang
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Cong Yi
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Liang Z, Huang J, Wang Y, Hua S, Jiang K. Diverse microtubule-binding repeats regulate TPX2 activities at distinct locations within the spindle. J Cell Biol 2025; 224:e202404025. [PMID: 39821262 PMCID: PMC11737348 DOI: 10.1083/jcb.202404025] [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: 04/05/2024] [Revised: 11/12/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
TPX2 is an elongated molecule containing multiple α-helical repeats. It stabilizes microtubules (MTs), promotes MT nucleation, and is essential for spindle assembly. However, the molecular basis of how TPX2 performs these functions remains elusive. Here, we systematically characterized the MT-binding activities of all TPX2 modules individually and in combinations and investigated their respective contributions both in vitro and in cells. We show that TPX2 contains α-helical repeats with opposite preferences for "extended" and "compacted" tubulin dimer spacing, and their distinct combinations produce divergent outcomes, making TPX2 activity highly robust yet tunable. Importantly, a repeat group at the C terminus, R8-9, is the key determinant of the TPX2 function. It stabilizes MTs by promoting rescues in vitro and is critical in spindle assembly. We propose a model where TPX2 activities are spatially regulated via its diverse MT-binding repeats to accommodate its varied functions in distinct locations within the spindle. Furthermore, we reveal a synergy between TPX2 and HURP in stabilizing spindle MTs.
Collapse
Affiliation(s)
- Zhuobi Liang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Junjie Huang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Yong Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Shasha Hua
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Kai Jiang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Scott TG, Guertin MJ. Rapid protein degradation systems to determine gene function in vivo. Lab Anim (NY) 2025; 54:66-67. [PMID: 40021933 PMCID: PMC11930339 DOI: 10.1038/s41684-025-01519-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
The functional characterization of proteins during temporally constrained periods of mammalian development and disease is largely limited by the inability to rapidly and reversibly perturb their function. A new study addresses this challenge by directly comparing two targeted protein degradation systems in mice. These systems enable precise temporal degradation of proteins, offering unprecedented opportunities to study dynamic biological processes. Several technical components remain to be optimized, but these technologies promise to provide novel insights in vivo.
Collapse
Affiliation(s)
- Thomas G Scott
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Michael J Guertin
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
| |
Collapse
|
19
|
Popchock AR, Hedouin S, Mao Y, Asbury CL, Stergachis AB, Biggins S. Stable centromere association of the yeast histone variant Cse4 requires its essential N-terminal domain. EMBO J 2025; 44:1488-1511. [PMID: 39809842 PMCID: PMC11876619 DOI: 10.1038/s44318-024-00345-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Chromosome segregation relies on kinetochores that assemble on specialized centromeric chromatin containing a histone H3 variant. In budding yeast, a single centromeric nucleosome containing Cse4 assembles at a sequence-defined 125 bp centromere. Yeast centromeric sequences are poor templates for nucleosome formation in vitro, suggesting the existence of mechanisms that specifically stabilize Cse4 nucleosomes in vivo. The extended Cse4 N-terminal tail binds to the chaperone Scm3, and a short essential region called END within the N-terminal tail binds the inner kinetochore complex Okp1/Ame1. To address the roles of these interactions, we utilized single-molecule fluorescence assays to monitor Cse4 during kinetochore assembly. We found that Okp1/Ame1 and Scm3 independently stabilize Cse4 at centromeres via their END interaction. Scm3 and Cse4 stability at the centromere are enhanced by Ipl1/Aurora B phosphorylation of the Cse4 END, identifying a previously unknown role for Ipl1 in ensuring Cse4 stability. Strikingly, a phosphomimetic mutation in the Cse4 END restores Cse4 recruitment in mutants defective in Okp1/Ame1 binding. Together, these data suggest that a key function of the essential Cse4 N-terminus is to ensure Cse4 localization at centromeres.
Collapse
Affiliation(s)
- Andrew R Popchock
- Howard Hughes Medical Institute, Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Sabrine Hedouin
- Howard Hughes Medical Institute, Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Yizi Mao
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Charles L Asbury
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Andrew B Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sue Biggins
- Howard Hughes Medical Institute, Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
| |
Collapse
|
20
|
Chen Y, Hu J, Zhao P, Fang J, Kuang Y, Liu Z, Dong S, Yao W, Ding Y, Wang X, Pan Y, Wu J, Zhao J, Yang J, Xu Z, Liu X, Zhang Y, Wu C, Zhang L, Fan M, Feng S, Hong Z, Yan Z, Xia H, Tang K, Yang B, Liu W, Sun Q, Mei K, Zou W, Huang Y, Feng D, Yi C. Rpl12 is a conserved ribophagy receptor. Nat Cell Biol 2025; 27:477-492. [PMID: 39934334 DOI: 10.1038/s41556-024-01598-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/12/2024] [Indexed: 02/13/2025]
Abstract
Ribophagy is a selective autophagic process that regulates ribosome turnover. Although NUFIP1 has been identified as a mammalian receptor for ribophagy, its homologues do not exist in yeast and nematodes. Here we demonstrate that Rpl12, a ribosomal large subunit protein, functions as a conserved ribophagy receptor in multiple organisms. Disruption of Rpl12-Atg8s binding leads to significant accumulation of ribosomal proteins and rRNA, while Atg1-mediated Rpl12 phosphorylation enhances its association with Atg11, thus triggering ribophagy during starvation. Ribophagy deficiency accelerates cell death induced by starvation and pathogen infection, leading to impaired growth and development and a shortened lifespan in both Caenorhabditis elegans and Drosophila melanogaster. Moreover, ribophagy deficiency results in motor impairments associated with ageing, while the overexpression of RPL12 significantly improves movement defects induced by starvation, ageing and Aβ accumulation in fly models. Our findings suggest that Rpl12 functions as a conserved ribophagy receptor vital for ribosome metabolism and cellular homeostasis.
Collapse
Affiliation(s)
- Yuting Chen
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jiaxin Hu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Pengwei Zhao
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Fang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingqi Kuang
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaojie Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuling Dong
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, China
| | - Weijing Yao
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanyuan Ding
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xinhui Wang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yibin Pan
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianbin Wu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingwei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Yang
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenzhong Xu
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodi Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yi Zhang
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Choufei Wu
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, China
| | - Liqin Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, China
| | - Mingzhu Fan
- Mass Spectrometry and Metabolomics Core Facility, Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, China
| | - Shan Feng
- Mass Spectrometry and Metabolomics Core Facility, Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, China
| | - Zhi Hong
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Zhangming Yan
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Hongguang Xia
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaiyue Tang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Bing Yang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Wei Liu
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kunrong Mei
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| | - Yunpeng Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Du Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Cong Yi
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
21
|
Klompstra TM, Yoon KJ, Koo BK. Evolution of organoid genetics. Eur J Cell Biol 2025; 104:151481. [PMID: 40056574 DOI: 10.1016/j.ejcb.2025.151481] [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: 11/30/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025] Open
Abstract
Organoids have revolutionized in vitro research by offering three-dimensional, multicellular systems that recapitulate the structure, function, and genetics of human tissues. Initially developed from both pluripotent stem cells (PSCs) and adult stem cells (AdSCs), organoids have expanded to model nearly every major human organ, significantly advancing developmental biology, disease modeling, and therapeutic screening. This review highlights the progression of organoid technologies, emphasizing the integration of genetic tools, including CRISPR-Cas9, prime editing, and lineage tracing. These advancements have facilitated precise modeling of human-specific pathologies and drug responses, often surpassing traditional 2D cultures and animal models in accuracy. Emerging technologies, such as organoid fusion, xenografting, and optogenetics, are expected to further enhance our understanding of cellular interactions and microenvironmental dynamics. As organoid complexity and genetic engineering methods continue to evolve, they will become increasingly indispensable for personalized medicine and translational research, bridging gaps between in vitro and in vivo systems.
Collapse
Affiliation(s)
- Thomas M Klompstra
- Center for Genome Engineering, Institute for Basic Sciences (IBS), Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Republic of Korea; Graduate School of Stem Cell and Regenerative Biology, KAIST, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, KAIST, Daejeon 34141, Republic of Korea
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Sciences (IBS), Republic of Korea; Graduate School of Stem Cell and Regenerative Biology, KAIST, Daejeon 34141, Republic of Korea; Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Republic of Korea.
| |
Collapse
|
22
|
Sladky VC, Strong MA, Tapias-Gomez D, Jewett CE, Drown CG, Scott PM, Holland AJ. Rapid and sustained degradation of the essential centrosome protein CEP192 in live mice using the AID2 system. SCIENCE ADVANCES 2025; 11:eadq2339. [PMID: 40020058 PMCID: PMC11870075 DOI: 10.1126/sciadv.adq2339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025]
Abstract
Studying essential genes required for dynamic processes in live mice is challenging as genetic perturbations are irreversible and limited by slow protein depletion kinetics. The auxin-inducible degron (AID) system is a powerful tool for analyzing inducible protein loss in vitro, but it is toxic to mice. Here, we use an optimized second-generation AID system to achieve the conditional and reversible loss of the essential centrosomal protein CEP192 in live mice. We show that the auxin derivative 5-phenyl-indole-3-acetic acid is well tolerated over 2 weeks and drives near-complete CEP192 degradation in less than 1 hour in vivo. CEP192 loss did not affect centriole duplication but decreased γ-tubulin recruitment to centrosomes impairing mitotic spindle assembly. Sustained CEP192 loss in vivo led to cell division failure and cell death in proliferative tissues. Thus, the second-generation AID system is well suited for rapid and/or sustained protein depletion in live mice to study essential functions in vivo.
Collapse
Affiliation(s)
- Valentina C. Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Margaret A. Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel Tapias-Gomez
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Cayla E. Jewett
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chelsea G. Drown
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Phillip M. Scott
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew J. Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
23
|
Murayama A, Matsui S, Abe T, Kanemaki MT, Kurosawa K, Hirota K, Ohta K, Seo H. Monoclonal antibody generation by controlled immunoglobulin gene rearrangements. Commun Biol 2025; 8:283. [PMID: 40011586 PMCID: PMC11865429 DOI: 10.1038/s42003-025-07690-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/06/2025] [Indexed: 02/28/2025] Open
Abstract
Monoclonal antibodies (mAbs) are essential for various applications including experimental reagents, diagnostics, and therapeutics. Thus, the platform technologies that stably generate antigen-specific mAbs are increasingly crucial. We previously developed a method to generate mAbs, termed the "ADLib system", utilizing the avian-derived B cell line DT40. Avian immunoglobulin (Ig) genes diversify principally through gene conversion-a kind of homologous recombination. The ADLib system isolates antigen-specific clones from libraries constructed using DT40 cells treated with Trichostatin A (TSA), a histone deacetylase inhibitor that enhances gene conversion frequencies. The obtained antigen-specific clones are cultured without TSA to minimize further diversification. However, low-frequency spontaneous gene conversion still occurs, potentially leading to gradual changes in the specificity of the clones. To address this, we engineered conditional mutants of activation-induced deaminase (AID), the initiator of gene conversion, using auxin-inducible degron system which enables targeted protein degradation via the auxin-dependent ubiquitin-proteasome pathway. The addition of the phytohormone auxin led to the degradation of degron-tagged AID proteins, effectively halting gene conversion. Subsequently, we carried out the ADLib system using these clones and successfully isolated antigen-specific mAbs. These suggest that our AID conditional mutants provide a powerful tool for generating and stabilizing antigen-specific clones isolated by the ADLib system.
Collapse
Affiliation(s)
- Akiho Murayama
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin Matsui
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Takuya Abe
- Department of Biochemistry, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Masato T Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Shizuoka, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Shizuoka, Japan
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kohei Kurosawa
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Department of Technology Development, Chiome Bioscience Inc, Tokyo, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Kunihiro Ohta
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidetaka Seo
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
24
|
Zellag RM, Poupart V, Negishi T, Labbé JC, Gerhold AR. The spatiotemporal distribution of LIN-5/NuMA regulates spindle orientation in the C. elegans germ line. Cell Rep 2025; 44:115296. [PMID: 39946234 DOI: 10.1016/j.celrep.2025.115296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/06/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Mitotic spindle orientation contributes to tissue organization and shape by setting the cell division plane. How spindle orientation is coupled to diverse tissue architectures is incompletely understood. The C. elegans gonad is a tube-shaped organ with germ cells forming a circumferential monolayer around a common cytoplasmic lumen. How this organization is maintained during development is unclear, as germ cells lack the canonical cell-cell junctions that ensure spindle orientation in other tissue types. Here, we show that the microtubule force generator dynein and its conserved regulator LIN-5/NuMA regulate germ cell spindle orientation and are required for germline tissue organization. We uncover a cyclic, polarized pattern of LIN-5/NuMA cortical localization that predicts centrosome positioning throughout the cell cycle, providing a means to align spindle orientation with the tissue plane. This work reveals a new mechanism by which oriented cell division can be achieved to maintain tissue organization during animal development.
Collapse
Affiliation(s)
- Réda M Zellag
- Institute for Research in Immunology and Cancer (IRIC), Montréal, QC H3C 3J7, Canada; Department of Pathology and Cell Biology, Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada; Department of Biology, McGill University, 1205 Avenue Docteur Penfield, Montréal, QC H2A 1B1, Canada
| | - Vincent Poupart
- Institute for Research in Immunology and Cancer (IRIC), Montréal, QC H3C 3J7, Canada
| | - Takefumi Negishi
- Multicellular Organization Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Mishima, Shizuoka 411-8540, Japan; Department of Genetics, School of Life Sciences, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Jean-Claude Labbé
- Institute for Research in Immunology and Cancer (IRIC), Montréal, QC H3C 3J7, Canada; Department of Pathology and Cell Biology, Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada.
| | - Abigail R Gerhold
- Department of Biology, McGill University, 1205 Avenue Docteur Penfield, Montréal, QC H2A 1B1, Canada.
| |
Collapse
|
25
|
Inglebert M, Dettwiler M, He C, Markkanen E, Opitz L, Naguleswaran A, Rottenberg S. Individualized Pooled CRISPR/Cas9 Screenings Identify CDK2 as a Druggable Vulnerability in a Canine Mammary Carcinoma Patient. Vet Sci 2025; 12:183. [PMID: 40005944 PMCID: PMC11861728 DOI: 10.3390/vetsci12020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
High-throughput omics approaches have long been used to uncover potential vulnerabilities in human personalized oncology but are often limited by the lack of functional validation. Therefore, we placed our emphasis on functional drug testing using patient-derived organoids (PDOs). However, PDOs generated from tumors mostly lack comparison with matching normal tissue, and the number of testable drugs is limited. Here, we demonstrate how matching the neoplastic and non-neoplastic mammary PDOs derived from the same dog can utilize targeted CRISPR/Cas9 screens to unveil cancer cell specific vulnerabilities. We performed two independent CRISPR/Cas9 dropout screens using sub-libraries targeting the epigenome (n = 1269) or druggable genes (n = 834) in paired PDOs derived from both carcinoma and normal mammary tissues from the same dog. A comparison of essential genes for tumor cells survival identified CDK2 as a functional vulnerability in canine mammary tumors (CMTs) that can be targeted with the PF3600 inhibitor. Additional potential targets were also uncovered, providing insights for personalized cancer treatments in dogs.
Collapse
Affiliation(s)
- Marine Inglebert
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.I.); (M.D.); (C.H.); (A.N.)
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Martina Dettwiler
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.I.); (M.D.); (C.H.); (A.N.)
- Vetscope Pathologie Dettwiler, Lörracherstrasse 50, 4125 Riehen, Switzerland
| | - Chang He
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.I.); (M.D.); (C.H.); (A.N.)
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8056 Zürich, Switzerland;
| | - Lennart Opitz
- Functional Genomics Center Zurich, University of Zürich and ETH, 8092 Zürich, Switzerland;
| | - Arunasalam Naguleswaran
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.I.); (M.D.); (C.H.); (A.N.)
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (M.I.); (M.D.); (C.H.); (A.N.)
- Bern Center for Precision Medicine, University of Bern, 3012 Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
26
|
Flashner S, Azizkhan-Clifford J. Emerging Roles for Transcription Factors During Mitosis. Cells 2025; 14:263. [PMID: 39996736 PMCID: PMC11853531 DOI: 10.3390/cells14040263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
The genome is dynamically reorganized, partitioned, and divided during mitosis. Despite their role in organizing interphase chromatin, transcription factors were largely believed to be mitotic spectators evicted from chromatin during mitosis, only able to reestablish their position on DNA upon entry into G1. However, a panoply of evidence now contradicts this early belief. Numerous transcription factors are now known to remain active during mitosis to achieve diverse purposes, including chromosome condensation, regulation of the centromere/kinetochore function, and control of centrosome homeostasis. Inactivation of transcription factors during mitosis results in chromosome segregation errors, key features of cancer. Moreover, active transcription and the production of centromere-derived transcripts during mitosis are also known to play key roles in maintaining chromosomal stability. Finally, many transcription factors are associated with chromosomal instability through poorly defined mechanisms. Herein, we will review the emerging roles of transcription factors and transcription during mitosis with a focus on their role in promoting the faithful segregation of sister chromatids.
Collapse
Affiliation(s)
| | - Jane Azizkhan-Clifford
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| |
Collapse
|
27
|
Matabishi-Bibi L, Goncalves C, Babour A. RNA exosome-driven RNA processing instructs the duration of the unfolded protein response. Nucleic Acids Res 2025; 53:gkaf088. [PMID: 39995043 PMCID: PMC11850225 DOI: 10.1093/nar/gkaf088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Upon stresses, cellular compartments initiate adaptive programs meant to restore homeostasis. Dedicated to the resolution of transient perturbations, these pathways are typically maintained at a basal level, activated upon stress, and critically downregulated upon reestablishment of cellular homeostasis. As such, prolonged activation of the unfolded protein response (UPR), a conserved adaptive transcriptional response to defective endoplasmic reticulum (ER) proteostasis, leads to cell death. Here, we elucidate an unanticipated role for the nuclear RNA exosome, an evolutionarily conserved ribonuclease complex that processes multiple classes of RNAs, in the control of UPR duration. Remarkably, the inactivation of Rrp6, an exclusively nuclear catalytic subunit of the RNA exosome, curtails UPR signaling, which is sufficient to promote the cell's resistance to ER stress. Mechanistically, accumulation of unprocessed RNA species diverts the processing machinery that maturates the messenger RNA encoding the master UPR regulator Hac1, thus restricting the UPR. Significantly, Rrp6 expression is naturally dampened upon ER stress, thereby participating in homeostatic UPR deactivation.
Collapse
Affiliation(s)
- Laura Matabishi-Bibi
- Université Paris Cité, INSERM U944 and CNRS 7212, Institut de Recherche Saint Louis, Hôpital Saint Louis, 75475 Paris Cedex 10, France
| | - Coralie Goncalves
- Université Paris Cité, CNRS 7592, Institut Jacques Monod, F-75013 Paris, France
| | - Anna Babour
- Université Paris Cité, INSERM U944 and CNRS 7212, Institut de Recherche Saint Louis, Hôpital Saint Louis, 75475 Paris Cedex 10, France
- Université Paris Cité, CNRS 7592, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
28
|
Aboreden NG, Zhao H, Shan F, Liu F, Zhang H, Blobel GA. Cis-regulatory chromatin contacts form de novo in the absence of loop extrusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632634. [PMID: 39975341 PMCID: PMC11838467 DOI: 10.1101/2025.01.12.632634] [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
NIPBL promotes chromatin loop extrusion by the cohesin complex until it stalls at convergently oriented CTCF sites, leading to the formation of structural loops. However, to what extent loop extrusion contributes to the establishment vs maintenance of cis-regulatory element (CRE) connectivity is poorly understood. Here, we explored the de novo establishment of chromatin folding patterns at the mitosis-to-G1-phase transition upon acute NIPBL loss. NIPBL depletion primarily impaired the formation of cohesion-mediated structural loops with NIPBL dependence being proportional to loop length. In contrast, the majority of CRE loops were established independently of loop extrusion regardless of length. However, NIPBL depletion slowed the re-formation of CRE loops with weak enhancers. Transcription of genes at NIPBL-independent loop anchors was activated normally in the absence of NIPBL. In sum, establishment of most regulatory contacts and gene transcription following mitotic exit is independent of loop extrusion.
Collapse
Affiliation(s)
- Nicholas G. Aboreden
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Han Zhao
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Fengnian Shan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
- South China University of Technology, Guangzhou, China
| | - Fuhai Liu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Haoyue Zhang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Gerd A. Blobel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
29
|
Shade O, Ryan A, Belsito G, Deiters A. Investigating protein degradability through site-specific ubiquitin ligase recruitment. RSC Chem Biol 2025; 6:240-248. [PMID: 39711601 PMCID: PMC11657224 DOI: 10.1039/d4cb00273c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024] Open
Abstract
We report targeted protein degradation through the site-specific recruitment of native ubiquitin ligases to a protein of interest via conjugation of E3 ligase ligands. Direct comparison of degradation ability of proteins displaying the corresponding bioconjugation handle at different regions of protein surfaces was explored. We demonstrate the benefit of proximal lysine residues and investigate flexibility in linker length for the design of optimal degraders. Two proteins without known small molecule ligands, EGFP and DUSP6, were differentially degraded when modified at different locations on their protein surfaces. Further, the cereblon-mediated degradation of the known PROTAC target ERRα was improved through the recruitment of the E3 ligase to regions different from the known ligand binding site. This new methodology will provide insight into overall protein degradability, even in the absence of a known small molecule ligand and inform the process of new ligand and PROTAC development to achieve optimal protein degradation. Furthermore, this approach represents a new, small molecule-based conditional OFF switch of protein function with complete genetic specificity. Importantly, the protein of interest is only modified with a minimal surface modification (<200 Da) and does not require any protein domain fusions.
Collapse
Affiliation(s)
- Olivia Shade
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Amy Ryan
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Gabriella Belsito
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| |
Collapse
|
30
|
Gameiro E, Juárez-Núñez KA, Fung JJ, Shankar S, Luke B, Khmelinskii A. Genome-wide conditional degron libraries for functional genomics. J Cell Biol 2025; 224:e202409007. [PMID: 39692735 DOI: 10.1083/jcb.202409007] [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: 09/03/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Functional genomics with libraries of knockout alleles is limited to non-essential genes and convoluted by the potential accumulation of suppressor mutations in knockout backgrounds, which can lead to erroneous functional annotations. To address these limitations, we constructed genome-wide libraries of conditional alleles based on the auxin-inducible degron (AID) system for inducible degradation of AID-tagged proteins in the budding yeast Saccharomyces cerevisiae. First, we determined that N-terminal tagging is at least twice as likely to inadvertently impair protein function across the proteome. We thus constructed two libraries with over 5,600 essential and non-essential proteins fused at the C-terminus with an AID tag and an optional fluorescent protein. Approximately 90% of AID-tagged proteins were degraded in the presence of the auxin analog 5-Ph-IAA, with initial protein abundance and tag accessibility as limiting factors. Genome-wide screens for DNA damage response factors revealed a role for the glucose signaling factor GSF2 in resistance to hydroxyurea, highlighting how the AID libraries extend the yeast genetics toolbox.
Collapse
Affiliation(s)
| | | | | | | | - Brian Luke
- Institute of Molecular Biology , Mainz, Germany
- Johannes Gutenberg University Mainz, Institute for Developmental Neurology , Mainz, Germany
| | - Anton Khmelinskii
- Institute of Molecular Biology , Mainz, Germany
- Institute for Quantitative and Computational Biosciences, Johannes Gutenberg University Mainz , Mainz, Germany
| |
Collapse
|
31
|
Adli M, Przybyla L, Burdett T, Burridge PW, Cacheiro P, Chang HY, Engreitz JM, Gilbert LA, Greenleaf WJ, Hsu L, Huangfu D, Hung LH, Kundaje A, Li S, Parkinson H, Qiu X, Robson P, Schürer SC, Shojaie A, Skarnes WC, Smedley D, Studer L, Sun W, Vidović D, Vierbuchen T, White BS, Yeung KY, Yue F, Zhou T. MorPhiC Consortium: towards functional characterization of all human genes. Nature 2025; 638:351-359. [PMID: 39939790 DOI: 10.1038/s41586-024-08243-w] [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: 10/31/2023] [Accepted: 10/17/2024] [Indexed: 02/14/2025]
Abstract
Recent advances in functional genomics and human cellular models have substantially enhanced our understanding of the structure and regulation of the human genome. However, our grasp of the molecular functions of human genes remains incomplete and biased towards specific gene classes. The Molecular Phenotypes of Null Alleles in Cells (MorPhiC) Consortium aims to address this gap by creating a comprehensive catalogue of the molecular and cellular phenotypes associated with null alleles of all human genes using in vitro multicellular systems. In this Perspective, we present the strategic vision of the MorPhiC Consortium and discuss various strategies for generating null alleles, as well as the challenges involved. We describe the cellular models and scalable phenotypic readouts that will be used in the consortium's initial phase, focusing on 1,000 protein-coding genes. The resulting molecular and cellular data will be compiled into a catalogue of null-allele phenotypes. The methodologies developed in this phase will establish best practices for extending these approaches to all human protein-coding genes. The resources generated-including engineered cell lines, plasmids, phenotypic data, genomic information and computational tools-will be made available to the broader research community to facilitate deeper insights into human gene functions.
Collapse
Affiliation(s)
- Mazhar Adli
- Robert H. Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Laralynne Przybyla
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Tony Burdett
- Omics Section, European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, UK
| | - Paul W Burridge
- Department of Pharmacology, Center for Pharmacogenomics, Northwestern University, Feinberg School of Medicine, Evanston, IL, USA
| | - Pilar Cacheiro
- William Harvey Research Institute, Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London, UK
| | - Howard Y Chang
- Department of Dermatology, Stanford University, Stanford, CA, USA
| | - Jesse M Engreitz
- Department of Genetics, Stanford University, Stanford, CA, USA
- Basic Science and Engineering (BASE) Initiative, Stanford University, Stanford, CA, USA
| | - Luke A Gilbert
- Department of Urology, University of California, San Francisco, CA, USA
| | | | - Li Hsu
- Department of Biostatistics, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Ling-Hong Hung
- School of Engineering and Technology, University of Washington Tacoma, Tacoma, WA, USA
| | - Anshul Kundaje
- Departments of Genetics and Computer Science, Stanford University, Stanford, CA, USA
| | - Sheng Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Helen Parkinson
- Knowledge Management Section, European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, UK
| | - Xiaojie Qiu
- Basic Science and Engineering (BASE) Initiative, Stanford University, Stanford, CA, USA
- Departments of Genetics and Computer Science, Stanford University, Stanford, CA, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Stephan C Schürer
- Molecular and Cellular Pharmacology; Sylvester Comprehensive Cancer Center, University of Miami, Coral Gables, FL, USA
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | - Damian Smedley
- William Harvey Research Institute, Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London, UK
| | - Lorenz Studer
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Wei Sun
- Department of Biostatistics, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dušica Vidović
- Molecular and Cellular Pharmacology; Sylvester Comprehensive Cancer Center, University of Miami, Coral Gables, FL, USA
| | - Thomas Vierbuchen
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Brian S White
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ka Yee Yeung
- School of Engineering and Technology, University of Washington Tacoma, Tacoma, WA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Ting Zhou
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| |
Collapse
|
32
|
Saca VR, Huber T, Sakmar TP. G protein-coupled receptor-targeted proteolysis-targeting chimeras in cancer therapeutics. Mol Pharmacol 2025; 107:100013. [PMID: 40023512 DOI: 10.1016/j.molpha.2024.100013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/05/2024] [Indexed: 03/04/2025] Open
Abstract
G protein-coupled receptors (GPCRs) comprise a family of heptahelical membrane proteins that mediate intracellular and intercellular transmembrane signaling. Defects in GPCR signaling pathways are implicated in the pathophysiology of many diseases, including cardiovascular disease, endocrinopathies, immune disorders, and cancer. Although GPCRs are attractive drug targets, only a small number of Food and Drug Administration-approved anticancer therapeutics target GPCRs. Targeted protein degradation (TPD) technology allows for the direct modulation of the cellular expression level of a protein of interest. TPD methods such as proteolysis-targeting chimeras (PROTACs) use the ubiquitin-proteasome system to degrade a protein of interest selectively. Although the PROTAC system has not been widely applied to GPCRs and other membrane proteins, there is evidence that PROTACs or other TPD methods could be applied to the GPCRome. Current GPCR PROTACs show the feasibility of using PROTACs to degrade GPCRs; however, the degradation mechanism for some of these GPCR PROTACs is uncertain. Additional studies aimed at elucidating the degradation mechanism of GPCRs with PROTACs are necessary. Discovery of new allosteric intracellular small molecule binders of GPCRs will be required for the development of intracellularly oriented PROTACs. Promising early results in targeted degradation of GPCRs suggest that TPD drug discovery platforms will be useful in developing PROTACs targeting pathological GPCRs. SIGNIFICANCE STATEMENT: Aberrant signaling of G protein-coupled receptors (GPCRs) can contribute to the pathophysiology of cancer. Although GPCRs are generally highly attractive drug targets, many individual GPCRs are currently undrugged using traditional drug discovery approaches. Targeted protein degradation technologies, such as proteolysis-targeting chimeras, provide a new approach to drug discovery for targeting previously undruggable GPCRs relevant to the molecular pathophysiology of cancer.
Collapse
Affiliation(s)
- Victoria R Saca
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York; Tri-Institutional PhD Program in Chemical Biology, New York, New York
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York.
| |
Collapse
|
33
|
Bunner S, Prince K, Pujadas Liwag EM, Eskndir N, Srikrishna K, Amonu McCarthy A, Kuklinski A, Jackson O, Pellegrino P, Jagtap S, Eweka I, Lawlor C, Eastin E, Yas G, Aiello J, LaPointe N, Schramm von Blucher I, Hardy J, Chen J, Figueroa S, Backman V, Janssen A, Packard M, Dorfman K, Almassalha L, Bahiru MS, Stephens AD. Decreased DNA density is a better indicator of a nuclear bleb than lamin B loss. J Cell Sci 2025; 138:jcs262082. [PMID: 39501901 PMCID: PMC11883270 DOI: 10.1242/jcs.262082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/30/2024] [Indexed: 11/13/2024] Open
Abstract
Nuclear blebs are herniations of the nucleus that occur in diseased nuclei and cause nuclear rupture leading to cellular dysfunction. Chromatin and lamins are two of the major structural components of the nucleus that maintain its shape and function, but their relative roles in nuclear blebbing remain elusive. To determine the composition of nuclear blebs, we compared the immunofluorescence intensity of DNA and lamin B in the main nucleus body to that in the nuclear bleb across cell types and perturbations. DNA density in the nuclear bleb was consistently decreased to about half that of the nuclear body whereas lamin B levels in the nuclear bleb varied widely. Partial wave spectroscopic (PWS) microscopy recapitulated the significantly decreased likelihood of high-density domains in the nuclear bleb versus body, and that it was independent of lamin B level. Time-lapse imaging into immunofluorescence revealed that decreased DNA density marked all nuclear blebs whereas decreased lamin B1 levels only occurred in blebs that had recently ruptured. Thus, decreased DNA density is a better marker of a nuclear bleb than lamin B level.
Collapse
Affiliation(s)
- Samantha Bunner
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Kelsey Prince
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Emily M. Pujadas Liwag
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- IBIS Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Nebiyat Eskndir
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Karan Srikrishna
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | | - Anna Kuklinski
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Olivia Jackson
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Pedro Pellegrino
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Shrushti Jagtap
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Imuetiyan Eweka
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Colman Lawlor
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Emma Eastin
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Griffin Yas
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Julianna Aiello
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Nathan LaPointe
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | | - Jillian Hardy
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jason Chen
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Schuyler Figueroa
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Anne Janssen
- School of Biological Sciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Mary Packard
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Katherine Dorfman
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Luay Almassalha
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Michael Seifu Bahiru
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, MA 01003, USA
| | - Andrew D. Stephens
- Biology Department, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
34
|
Hata R, Sugawara A, Fukuda M. Rab10 function in tubular endosome formation requires the N-terminal K3 residue and is disrupted by N-terminal tagging. J Cell Sci 2025; 138:JCS263649. [PMID: 39783278 DOI: 10.1242/jcs.263649] [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: 10/29/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Various N-terminal tags have often been used to identify the functions and localization of Rab small GTPases, but their impact on Rab proteins themselves has been poorly investigated. Here, we used a knockout (KO)-rescue approach to systematically evaluate the effect of N-terminal tagging of two Rabs, Rab10 and Rab27A, on RAB10-KO HeLa cells and Rab27A-deficient melanocytes (melan-ash cells), respectively. The results showed that all of the N-terminal-tagged Rab27A proteins mediated actin-based melanosome transport in the melan-ash cells, but none of the N-terminal-tagged Rab10 proteins fully rescued the defect in tubular endosome formation in RAB10-KO cells. Although the N-terminal-tagged Rab10 proteins had the ability to localize tubular endosomes in wild-type HeLa cells, they sometimes exhibited a dominant-negative effect on tubular endosome formation. We also found that a conserved lysine residue at amino acid position 3 (K3) in the Rab10 proteins of different species is required for tubular endosome formation. Thus, it will be important to determine whether other Rab isoforms with N-terminal tags behave similarly to their corresponding untagged isoforms by performing appropriate KO-rescue experiments in future studies.
Collapse
Affiliation(s)
- Rinka Hata
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Akira Sugawara
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
35
|
Trakroo D, Agarwal P, Alekar A, Ghosh SK. Nonessential kinetochore proteins contribute to meiotic chromosome condensation through polo-like kinase. Mol Biol Cell 2025; 36:ar14. [PMID: 39705398 PMCID: PMC11809314 DOI: 10.1091/mbc.e24-08-0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/12/2024] [Accepted: 12/10/2024] [Indexed: 12/22/2024] Open
Abstract
Chromosome condensation plays a pivotal role during faithful chromosome segregation, hence, understanding the factors that drive condensation is crucial to get mechanistic insight into chromosome segregation. Previously, we showed that in budding yeast, the absence of the nonessential kinetochore proteins affects chromatin-condensin association in meiosis but not in mitosis. A differential organization of the kinetochores, that we and others observed earlier during mitosis and meiosis may contribute to the meiotic-specific role. Here, with our in-depth investigation using in vivo chromosome condensation assays in cells lacking a nonessential kinetochore protein, Ctf19, we establish that these proteins have roles in achieving a higher meiotic condensation without influencing much of the mitotic condensation. We further observed an accumulation of the polo-like kinase Cdc5 owing to its higher protein stability in ctf19Δ meiotic cells. High Cdc5 activity causes hyperphosphorylation of the condensin resulting in its reduced stability and concomitant decreased association with the chromatin. Overall, our findings highlight the role of Ctf19 in promoting meiotic chromosome condensation by influencing the activity of Cdc5 and thereby affecting the stability and association of condensin with the chromatin.
Collapse
Affiliation(s)
- Deepika Trakroo
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai-400076, India
| | - Prakhar Agarwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai-400076, India
| | - Anushka Alekar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai-400076, India
| | - Santanu Kumar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai-400076, India
| |
Collapse
|
36
|
Luo M, Zhu S, Dang H, Wen Q, Niu R, Long J, Wang Z, Tong Y, Ning Y, Yuan M, Xu G. Genetically-encoded targeted protein degradation technology to remove endogenous condensation-prone proteins and improve crop performance. Nat Commun 2025; 16:1159. [PMID: 39880812 PMCID: PMC11779824 DOI: 10.1038/s41467-025-56570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Effective modulation of gene expression in plants is achievable through tools like CRISPR and RNA interference, yet methods for directly modifying endogenous proteins remain lacking. Here, we identify the E3 ubiquitin ligase E3TCD1 and develope a Targeted Condensation-prone-protein Degradation (TCD) strategy. The X-E3TCD1 fusion protein acts as a genetically engineered degrader, selectively targeting endogenous proteins prone to condensation. For example, a transgenic E3TCD1 fusion with Teosinte branched 1 (TB1) degrades the native TB1 protein, resulting in increased tiller numbers in rice. Additionally, conditional degradation of the negative defense regulator Early Flowering 3 via a pathogen-responsive ProTBF1-uORFsTBF1 cassette enhances rice blast resistance without affecting flowering time in the absence of pathogen. Unlike prevailing targeted protein degradation strategies, the TCD system does not rely on small molecules, antibodies, or genetic knock-in fusion tags, demonstrating its promise as a transgene-based approach for optimizing crop performance.
Collapse
Affiliation(s)
- Ming Luo
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Sitao Zhu
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Hua Dang
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Qing Wen
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Ruixia Niu
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Jiawei Long
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Zhao Wang
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Yongjia Tong
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
| | - Yuese Ning
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Meng Yuan
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- National Key Laboratory of Crop Genetic Improvement, National Centre of Plant Gene Research (Wuhan), Huazhong Agricultural University, Wuhan, Hubei, China
| | - Guoyong Xu
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China.
- Hubei Hongshan Laboratory, Wuhan, Hubei, China.
- RNA Institute, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
37
|
Li WS, Carter LM, Almassalha LM, Gong R, Pujadas-Liwag EM, Kuo T, MacQuarrie KL, Carignano M, Dunton C, Dravid V, Kanemaki MT, Szleifer I, Backman V. Mature chromatin packing domains persist after RAD21 depletion in 3D. SCIENCE ADVANCES 2025; 11:eadp0855. [PMID: 39854464 PMCID: PMC11759041 DOI: 10.1126/sciadv.adp0855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025]
Abstract
Understanding chromatin organization requires integrating measurements of genome connectivity and physical structure. It is well established that cohesin is essential for TAD and loop connectivity features in Hi-C, but the corresponding change in physical structure has not been studied using electron microscopy. Pairing chromatin scanning transmission electron tomography with multiomic analysis and single-molecule localization microscopy, we study the role of cohesin in regulating the conformationally defined chromatin nanoscopic packing domains. Our results indicate that packing domains are not physical manifestation of TADs. Using electron microscopy, we found that only 20% of packing domains are lost upon RAD21 depletion. The effect of RAD21 depletion is restricted to small, poorly packed (nascent) packing domains. In addition, we present evidence that cohesin-mediated loop extrusion generates nascent domains that undergo maturation through nucleosome posttranslational modifications. Our results demonstrate that a 3D genomic structure, composed of packing domains, is generated through cohesin activity and nucleosome modifications.
Collapse
Affiliation(s)
- Wing Shun Li
- Applied Physics Program, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Lucas M. Carter
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- IBIS Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Luay Matthew Almassalha
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Gastroenterology and Hepatology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Ruyi Gong
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Emily M. Pujadas-Liwag
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- IBIS Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Tiffany Kuo
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- IBIS Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Kyle L. MacQuarrie
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Marcelo Carignano
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Cody Dunton
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Vinayak Dravid
- Applied Physics Program, Northwestern University, Evanston, IL 60208, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
- Northwestern University Atomic and Nanoscale Characterization Experimental (NUANCE) Center, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology (IIN), Northwestern University, Evanston, IL 60208, USA
| | - Masato T. Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Yata 1111, Mishima, Shizuoka 411-8540, Japan
- Department of Biological Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Igal Szleifer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
38
|
Morabito RD, Tatarakis D, Swick R, Stettnisch S, Schilling TF, Horsfield JA, Martin BL. The ratio of Wnt signaling activity to Sox2 transcription factor levels predicts neuromesodermal fate potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633481. [PMID: 39868081 PMCID: PMC11761523 DOI: 10.1101/2025.01.16.633481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Neuromesodermal progenitors (NMPs) are a vertebrate cell type that contribute descendants to both the spinal cord and the mesoderm. The undifferentiated bipotential NMP state is maintained when both Wnt signaling is active and Sox2 is present. We used transgenic reporter lines to live-image both Wnt activity and Sox2 levels in NMPs and observed a unique cellular ratio in NMPs compared to NMP-derived mesoderm or neural tissue. We used this unique signature to identify the previously unknown anatomical position of a progenitor population that gives rise to the midline tissues of the floor plate of the spinal cord and the mesodermal notochord. Thus, quantification of the active Wnt signaling to Sox2 ratio can be used to predict and identify cells with neuromesodermal potential. We also developed the auxin inducible degron 2 system for use in zebrafish to test the temporal role that Sox2 plays during midline formation. We found ectopic Sox2 in the presence of Wnt activity holds cells in the undifferentiated floor plate/notochord progenitor state, and that degradation of the ectopic Sox2 is required for cells to adopt a notochord fate.
Collapse
Affiliation(s)
- Robert D. Morabito
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - David Tatarakis
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Ryan Swick
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - Samantha Stettnisch
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Julia A. Horsfield
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Auckland, New Zealand
- Genetics Otago Research Centre, University of Otago, Dunedin, New Zealand
| | - Benjamin L. Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| |
Collapse
|
39
|
Aboreden NG, Lam JC, Goel VY, Wang S, Wang X, Midla SC, Quijano A, Keller CA, Giardine BM, Hardison RC, Zhang H, Hansen AS, Blobel GA. LDB1 establishes multi-enhancer networks to regulate gene expression. Mol Cell 2025; 85:376-393.e9. [PMID: 39721581 PMCID: PMC11741933 DOI: 10.1016/j.molcel.2024.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/17/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024]
Abstract
How specific enhancer-promoter pairing is established remains mostly unclear. Besides the CTCF/cohesin machinery, few nuclear factors have been studied for a direct role in physically connecting regulatory elements. Using a murine erythroid cell model, we show via acute degradation experiments that LDB1 directly and broadly promotes connectivity among regulatory elements. Most LDB1-mediated contacts, even those spanning hundreds of kb, can form in the absence of CTCF, cohesin, or YY1 as determined using multiple degron systems. Moreover, an engineered LDB1-driven chromatin loop is cohesin independent. Cohesin-driven loop extrusion does not stall at LDB1-occupied sites but aids the formation of a subset of LDB1-anchored loops. Leveraging the dynamic reorganization of nuclear architecture during the transition from mitosis to G1 phase, we observe that loop formation and de novo LDB1 occupancy correlate and can occur independently of structural loops. Tri-C and Region Capture Micro-C reveal that LDB1 organizes multi-enhancer networks to activate transcription. These findings establish LDB1 as a driver of spatial connectivity.
Collapse
Affiliation(s)
- Nicholas G Aboreden
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jessica C Lam
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Viraat Y Goel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Siqing Wang
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xiaokang Wang
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Susannah C Midla
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alma Quijano
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cheryl A Keller
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Belinda M Giardine
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Ross C Hardison
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Haoyue Zhang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Anders S Hansen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Gerd A Blobel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Spiridon-Bodi M, Ros-Carrero C, Igual JC, Gomar-Alba M. Dual regulation of the levels and function of Start transcriptional repressors drives G1 arrest in response to cell wall stress. Cell Commun Signal 2025; 23:31. [PMID: 39819572 PMCID: PMC11737188 DOI: 10.1186/s12964-025-02027-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Many different stress signaling pathways converge in a common response: slowdown or arrest cell cycle in the G1 phase. The G1/S transition (called Start in budding yeast) is a key checkpoint controlled by positive and negative regulators. Among them, Whi7 and Whi5 are transcriptional repressors of the G1/S transcriptional program, yeast functional homologs of the Retinoblastoma family proteins in mammalian cells. Under standard conditions, Whi7 plays a lesser role than Whi5 in Start inhibition. However, under cell wall stress, Whi7 is induced and plays a more important role in G1/S control. In this work, we investigated the functional hallmarks of Whi7 and Whi5, which determine their strength as Start inhibitors under cell wall stress. METHODS The response of Saccharomyces cerevisiae to Calcofluor White was investigated to characterize the regulation and function of Whi7 and Whi5 under cell wall stress. To control their protein levels, we used dose-dependent β-estradiol-induced expression and auxin-induced degron protein fusions. We also performed Chromatin Immunoprecipitation assays to investigate Whi7 and Whi5 association with Start promoters and scored cell cycle arrest and re-entry using cell microscopy assays. RESULTS We found that cell wall stress promoted the specific upregulation of the Whi7 Start repressor. First, although cell wall stress increases Whi7 protein levels, this is not the only determinant behind the Whi7 function in promoting G1 arrest. Indeed, artificial induction of Whi5 at the same protein level resulted in a lower G1 block. Second, under cell wall stress, Whi7 was specifically recruited to SBF-target promoters, independent of the increase in its protein levels or cell cycle stage. Finally, we found that Whi7 protein instability further increased during cell wall stress and that Whi7 degradation triggered advanced cell cycle re-entry. CONCLUSIONS Here, we show that cell wall stress signaling specifically enhances Whi7 function as a Start transcriptional repressor. Importantly, we identified new Whi7-specific regulatory mechanisms that do not operate in the Whi5 repressor. Our results indicate that cells may benefit from stress-specific repressors to ensure the stress-induced G1 arrest and that Whi7 rapid degradation may be particularly important to resume cell cycle upon adaptation.
Collapse
Affiliation(s)
- Mihai Spiridon-Bodi
- Institut de Biotecnologia i Biomedicina (BIOTECMED) and Departament de Bioquímica i Biologia Molecular, Universitat de València, Burjassot, 46100, Spain
| | - Cristina Ros-Carrero
- Institut de Biotecnologia i Biomedicina (BIOTECMED) and Departament de Bioquímica i Biologia Molecular, Universitat de València, Burjassot, 46100, Spain
| | - J Carlos Igual
- Institut de Biotecnologia i Biomedicina (BIOTECMED) and Departament de Bioquímica i Biologia Molecular, Universitat de València, Burjassot, 46100, Spain
| | - Mercè Gomar-Alba
- Institut de Biotecnologia i Biomedicina (BIOTECMED) and Departament de Bioquímica i Biologia Molecular, Universitat de València, Burjassot, 46100, Spain.
| |
Collapse
|
41
|
Dong J, Sathyan K, Scott T, Mukherjee R, Guertin M. ZNF143 binds DNA and stimulates transcription initiation to activate and repress direct target genes. Nucleic Acids Res 2025; 53:gkae1182. [PMID: 39676670 PMCID: PMC11754675 DOI: 10.1093/nar/gkae1182] [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: 05/29/2024] [Revised: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024] Open
Abstract
Transcription factors bind to sequence motifs and act as activators or repressors. Transcription factors interface with a constellation of accessory cofactors to regulate distinct mechanistic steps to regulate transcription. We rapidly degraded the essential and pervasively expressed transcription factor ZNF143 to determine its function in the transcription cycle. ZNF143 facilitates RNA polymerase initiation and activates gene expression. ZNF143 binds the promoter of nearly all its activated target genes. ZNF143 also binds near the site of genic transcription initiation to directly repress a subset of genes. Although ZNF143 stimulates initiation at ZNF143-repressed genes (i.e. those that increase transcription upon ZNF143 depletion), the molecular context of binding leads to cis repression. ZNF143 competes with other more efficient activators for promoter access, physically occludes transcription initiation sites and promoter-proximal sequence elements, and acts as a molecular roadblock to RNA polymerases during early elongation. The term context specific is often invoked to describe transcription factors that have both activation and repression functions. We define the context and molecular mechanisms of ZNF143-mediated cis activation and repression.
Collapse
Affiliation(s)
- Jinhong Dong
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Ave, Farmington, Connecticut 06030, USA
| | - Kizhakke Mattada Sathyan
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Ave, Farmington, Connecticut 06030, USA
| | - Thomas G Scott
- Department of Biochemistry and Molecular Genetics, University of Virginia, 1340 Jefferson Park Ave, Charlottesville, Virginia 22903, USA
| | - Rudradeep Mukherjee
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Ave, Farmington, Connecticut 06030, USA
| | - Michael J Guertin
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Ave, Farmington, Connecticut 06030, USA
- Department of Genetics and Genome Sciences, University of Connecticut, 400 Farmington Ave, Farmington, Connecticut 06030, USA
| |
Collapse
|
42
|
Almassalha LM, Carignano M, Liwag EP, Li WS, Gong R, Acosta N, Dunton CL, Gonzalez PC, Carter LM, Kakkaramadam R, Kröger M, MacQuarrie KL, Frederick J, Ye IC, Su P, Kuo T, Medina KI, Pritchard JA, Skol A, Nap R, Kanemaki M, Dravid V, Szleifer I, Backman V. Chromatin conformation, gene transcription, and nucleosome remodeling as an emergent system. SCIENCE ADVANCES 2025; 11:eadq6652. [PMID: 39792661 PMCID: PMC11721585 DOI: 10.1126/sciadv.adq6652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025]
Abstract
In single cells, variably sized nanoscale chromatin structures are observed, but it is unknown whether these form a cohesive framework that regulates RNA transcription. Here, we demonstrate that the human genome is an emergent, self-assembling, reinforcement learning system. Conformationally defined heterogeneous, nanoscopic packing domains form by the interplay of transcription, nucleosome remodeling, and loop extrusion. We show that packing domains are not topologically associated domains. Instead, packing domains exist across a structure-function life cycle that couples heterochromatin and transcription in situ, explaining how heterochromatin enzyme inhibition can produce a paradoxical decrease in transcription by destabilizing domain cores. Applied to development and aging, we show the pairing of heterochromatin and transcription at myogenic genes that could be disrupted by nuclear swelling. In sum, packing domains represent a foundation to explore the interactions of chromatin and transcription at the single-cell level in human health.
Collapse
Affiliation(s)
- Luay M. Almassalha
- Department of Gastroenterology and Hepatology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Marcelo Carignano
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Emily Pujadas Liwag
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Wing Shun Li
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Applied Physics Program, Northwestern University, Evanston, IL 60208, USA
| | - Ruyi Gong
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Nicolas Acosta
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Cody L. Dunton
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Paola Carrillo Gonzalez
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Lucas M. Carter
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Rivaan Kakkaramadam
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Martin Kröger
- Magnetism and Interface Physics and Computational Polymer Physics, Department of Materials, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Kyle L. MacQuarrie
- Stanley Manne Children’s Research Institute, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jane Frederick
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - I Chae Ye
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Patrick Su
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Tiffany Kuo
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Karla I. Medina
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL 60208, USA
| | - Josh A Pritchard
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Andrew Skol
- Stanley Manne Children’s Research Institute, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Rikkert Nap
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Masato Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Yata 1111, Mishima, Shizuoka 411-8540, Japan
- Department of Biological Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Vinayak Dravid
- Applied Physics Program, Northwestern University, Evanston, IL 60208, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
- Northwestern University Atomic and Nanoscale Characterization Experimental (NUANCE) Center, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology (IIN), Northwestern University, Evanston, IL 60208, USA
| | - Igal Szleifer
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Vadim Backman
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
43
|
Ostrowski MS, Yang MG, McNally CP, Abdulhay NJ, Wang S, Renduchintala K, Irkliyenko I, Biran A, Chew BTL, Midha AD, Wong EV, Sandoval J, Jain IH, Groth A, Nora EP, Goodarzi H, Ramani V. The single-molecule accessibility landscape of newly replicated mammalian chromatin. Cell 2025; 188:237-252.e19. [PMID: 39549698 DOI: 10.1016/j.cell.2024.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 07/15/2024] [Accepted: 10/21/2024] [Indexed: 11/18/2024]
Abstract
We present replication-aware single-molecule accessibility mapping (RASAM), a method to nondestructively measure replication status and protein-DNA interactions on chromatin genome-wide. Using RASAM, we uncover a genome-wide state of single-molecule "hyperaccessibility" post-replication that resolves over several hours. Combining RASAM with cellular models for rapid protein degradation, we demonstrate that histone chaperone CAF-1 reduces nascent chromatin accessibility by filling single-molecular "gaps" and generating closely spaced dinucleosomes on replicated DNA. At cis-regulatory elements, we observe unique modes by which nascent chromatin hyperaccessibility resolves: at CCCTC-binding factor (CTCF)-binding sites, CTCF and nucleosomes compete, reducing CTCF occupancy and motif accessibility post-replication; at active transcription start sites, high chromatin accessibility is maintained, implying rapid re-establishment of nucleosome-free regions. Our study introduces a new paradigm for studying replicated chromatin fiber organization. More broadly, we uncover a unique organization of newly replicated chromatin that must be reset by active processes, providing a substrate for epigenetic reprogramming.
Collapse
Affiliation(s)
- Megan S Ostrowski
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA
| | - Marty G Yang
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA
| | - Colin P McNally
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA; UCSF Department of Biochemistry & Biophysics, San Francisco, CA 94158, USA
| | - Nour J Abdulhay
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA; UCSF Department of Biochemistry & Biophysics, San Francisco, CA 94158, USA
| | - Simai Wang
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA
| | | | - Iryna Irkliyenko
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA
| | - Alva Biran
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Brandon T L Chew
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Ayush D Midha
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Emily V Wong
- UCSF Department of Biochemistry & Biophysics, San Francisco, CA 94158, USA
| | - Jonathan Sandoval
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA
| | - Isha H Jain
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Anja Groth
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Cellular and Molecular Medicine (ICMM), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elphège P Nora
- UCSF Department of Biochemistry & Biophysics, San Francisco, CA 94158, USA; Cardiovascular Research Institute, UCSF, San Francisco, CA 94158, USA; Chan-Zuckerberg BioHub, San Francisco, CA 94158, USA
| | - Hani Goodarzi
- UCSF Department of Biochemistry & Biophysics, San Francisco, CA 94158, USA; Chan-Zuckerberg BioHub, San Francisco, CA 94158, USA; Helen Diller Cancer Research Center, UCSF, San Francisco, CA 94158, USA; Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA 94158, USA
| | - Vijay Ramani
- Gladstone Institute for Data Science & Biotechnology, San Francisco, CA 94158, USA; UCSF Department of Biochemistry & Biophysics, San Francisco, CA 94158, USA; Helen Diller Cancer Research Center, UCSF, San Francisco, CA 94158, USA; Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA 94158, USA.
| |
Collapse
|
44
|
Rentsch D, Bergs A, Shao J, Elvers N, Ruse C, Seidenthal M, Aoki I, Gottschalk A. Tools and methods for cell ablation and cell inhibition in Caenorhabditis elegans. Genetics 2025; 229:1-48. [PMID: 39110015 PMCID: PMC11708922 DOI: 10.1093/genetics/iyae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 01/11/2025] Open
Abstract
To understand the function of cells such as neurons within an organism, it can be instrumental to inhibit cellular function, or to remove the cell (type) from the organism, and thus to observe the consequences on organismic and/or circuit function and animal behavior. A range of approaches and tools were developed and used over the past few decades that act either constitutively or acutely and reversibly, in systemic or local fashion. These approaches make use of either drugs or genetically encoded tools. Also, there are acutely acting inhibitory tools that require an exogenous trigger like light. Here, we give an overview of such methods developed and used in the nematode Caenorhabditis elegans.
Collapse
Affiliation(s)
- Dennis Rentsch
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Amelie Bergs
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Jiajie Shao
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Nora Elvers
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Christiane Ruse
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Marius Seidenthal
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Ichiro Aoki
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| |
Collapse
|
45
|
Varandas KC, Hodges BM, Lubeck L, Farinas A, Liang Y, Lu Y, Shaham S. Glia detect and transiently protect against dendrite substructure disruption in C. elegans. Nat Commun 2025; 16:79. [PMID: 39747235 PMCID: PMC11696001 DOI: 10.1038/s41467-024-55674-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Glia assess axon structure to modulate myelination and axon repair. Whether glia similarly detect dendrites and their substructures is not well understood. Here we show that glia monitor the integrity of dendrite substructures and transiently protect them against perturbations. We demonstrate that disruption of C. elegans sensory neuron dendrite cilia elicits acute glial responses, including increased accumulation of glia-derived extracellular matrix around cilia, changes in gene expression, and alteration of secreted protein repertoire. DGS-1, a 7-transmembrane domain neuronal protein, and FIG-1, a multifunctional thrombospondin-domain glial protein, are required for glial detection of cilia integrity, physically interact, and exhibit mutually-dependent localization to and around cilia, respectively. Glial responses to dendrite cilia disruption transiently protect against damage. Thus, our studies uncover a homeostatic, protective, dendrite-glia signaling interaction regulating dendrite substructure integrity.
Collapse
Affiliation(s)
- Katherine C Varandas
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA
| | - Brianna M Hodges
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA
| | - Lauren Lubeck
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA
- Graduate Program in Biology, Hopkins Marine Station of Stanford University, Pacific Grove, CA, USA
| | - Amelia Farinas
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA
- Graduate Program in Neuroscience, Stanford University, Stanford, CA, USA
| | - Yupu Liang
- CCTS Research Bioinformatics, The Rockefeller University, New York, NY, USA
- Bioinformatics Data Engineering, Alexion Pharmaceuticals, Boston, MA, USA
| | - Yun Lu
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
46
|
Mohajan S, Rubio LS, Gross DS. Nuclear basket proteins Mlp1 and Nup2 drive heat shock-induced 3D genome restructuring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.01.631024. [PMID: 39803495 PMCID: PMC11722380 DOI: 10.1101/2025.01.01.631024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The nuclear pore complex (NPC), a multisubunit complex located within the nuclear envelope, regulates RNA export and the import and export of proteins. Here we address the role of the NPC in driving thermal stress-induced 3D genome repositioning of Heat Shock Responsive (HSR) genes in yeast. We found that two nuclear basket proteins, Mlp1 and Nup2, although dispensable for NPC integrity, are required for driving HSR genes into coalesced chromatin clusters, consistent with their strong, heat shock-dependent recruitment to HSR gene regulatory and coding regions. HSR gene clustering occurs predominantly within the nucleoplasm and is independent of the essential scaffold-associated proteins Nup1 and Nup145. Notably, double depletion of Mlp1 and Nup2 has little effect on the formation of Heat Shock Factor 1 (Hsf1)-containing transcriptional condensates, Hsf1 and Pol II recruitment to HSR genes, or HSR mRNA abundance. Our results define a 3D genome restructuring role for nuclear basket proteins extrinsic to the NPC and downstream of HSR gene activation.
Collapse
Affiliation(s)
- Suman Mohajan
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| | - Linda S. Rubio
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| | - David S. Gross
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| |
Collapse
|
47
|
Hervé S, Scelfo A, Bersano Marchisio G, Grison M, Vaidžiulytė K, Dumont M, Angrisani A, Keikhosravi A, Pegoraro G, Deygas M, P F Nader G, Macé AS, Gentili M, Williart A, Manel N, Piel M, Miroshnikova YA, Fachinetti D. Chromosome mis-segregation triggers cell cycle arrest through a mechanosensitive nuclear envelope checkpoint. Nat Cell Biol 2025; 27:73-86. [PMID: 39779939 PMCID: PMC11735390 DOI: 10.1038/s41556-024-01565-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025]
Abstract
Errors during cell division lead to aneuploidy, which is associated with genomic instability and cell transformation. In response to aneuploidy, cells activate the tumour suppressor p53 to elicit a surveillance mechanism that halts proliferation and promotes senescence. The molecular sensors that trigger this checkpoint are unclear. Here, using a tunable system of chromosome mis-segregation, we show that mitotic errors trigger nuclear deformation, nuclear softening, and lamin and heterochromatin alterations, leading to rapid p53/p21 activation upon mitotic exit in response to changes in nuclear mechanics. We identify mTORC2 and ATR as nuclear deformation sensors upstream of p53/p21 activation. While triggering mitotic arrest, the chromosome mis-segregation-induced alterations of nuclear envelope mechanics provide a fitness advantage for aneuploid cells by promoting nuclear deformation resilience and enhancing pro-invasive capabilities. Collectively, this work identifies a nuclear mechanical checkpoint triggered by altered chromatin organization that probably plays a critical role in cellular transformation and cancer progression.
Collapse
Affiliation(s)
- Solène Hervé
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Scelfo
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | | | - Marine Grison
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | - Kotryna Vaidžiulytė
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Marie Dumont
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | - Annapaola Angrisani
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | - Adib Keikhosravi
- High-Throughput Imaging Facility, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gianluca Pegoraro
- High-Throughput Imaging Facility, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mathieu Deygas
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Guilherme P F Nader
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anne-Sophie Macé
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
- CNRS UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, PSL Research University, Paris, France
| | - Matteo Gentili
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Alice Williart
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Nicolas Manel
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Matthieu Piel
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Yekaterina A Miroshnikova
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Daniele Fachinetti
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France.
| |
Collapse
|
48
|
Kong W, Hara M, Tokunaga Y, Okumura K, Hirano Y, Miao J, Takenoshita Y, Hashimoto M, Sasaki H, Fujimori T, Wakabayashi Y, Fukagawa T. CENP-C-Mis12 complex establishes a regulatory loop through Aurora B for chromosome segregation. Life Sci Alliance 2025; 8:e202402927. [PMID: 39433344 PMCID: PMC11494776 DOI: 10.26508/lsa.202402927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Establishing the correct kinetochore-microtubule attachment is crucial for faithful chromosome segregation. The kinetochore has various regulatory mechanisms for establishing correct bipolar attachment. However, how the regulations are coupled is not fully understood. Here, we demonstrate a regulatory loop between the kinetochore protein CENP-C and Aurora B kinase, which is critical for the error correction of kinetochore-microtubule attachment. This regulatory loop is mediated through the binding of CENP-C to the outer kinetochore Mis12 complex (Mis12C). Although the Mis12C-binding region of CENP-C is dispensable for mouse development and proliferation in human RPE-1 cells, those cells lacking this region display increased mitotic defects. The CENP-C-Mis12C interaction facilitates the centromeric recruitment of Aurora B and the mitotic error correction in human cells. Given that Aurora B reinforces the CENP-C-Mis12C interaction, our findings reveal a positive regulatory loop between Aurora B recruitment and the CENP-C-Mis12C interaction, which ensures chromosome biorientation for accurate chromosome segregation.
Collapse
Affiliation(s)
- Weixia Kong
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Masatoshi Hara
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yurika Tokunaga
- Division of Experimental Animal Research, Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Kazuhiro Okumura
- Division of Experimental Animal Research, Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Jiahang Miao
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | | - Masakazu Hashimoto
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Cell Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hiroshi Sasaki
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Yuichi Wakabayashi
- Division of Experimental Animal Research, Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
49
|
Formichetti S, Sadowska A, Ascolani M, Hansen J, Ganter K, Lancrin C, Humphreys N, Boulard M. Genetic gradual reduction of OGT activity unveils the essential role of O-GlcNAc in the mouse embryo. PLoS Genet 2025; 21:e1011507. [PMID: 39787076 PMCID: PMC11717234 DOI: 10.1371/journal.pgen.1011507] [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: 08/14/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
The reversible glycosylation of nuclear and cytoplasmic proteins (O-GlcNAcylation) is catalyzed by a single enzyme, namely O-GlcNAc transferase (OGT). The mammalian Ogt gene is X-linked, and it is essential for embryonic development and for the viability of proliferating cells. We perturbed OGT's function in vivo by creating a murine allelic series of four single amino acid substitutions, reducing OGT's catalytic activity to a range of degrees. The severity of the embryonic lethality was proportional to the extent of impairment of OGT's catalysis, demonstrating that the O-GlcNAc modification itself is required for early development. We identified hypomorphic Ogt alleles that perturb O-GlcNAc homeostasis while being compatible with embryogenesis. The analysis of the transcriptomes of the mutant embryos at different developmental stages suggested a sexually-dimorphic developmental delay caused by the decrease in O-GlcNAc. Furthermore, a mild reduction of OGT's enzymatic activity was sufficient to loosen the silencing of endogenous retroviruses in vivo.
Collapse
Affiliation(s)
- Sara Formichetti
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Germany
| | - Agnieszka Sadowska
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| | - Michela Ascolani
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| | - Julia Hansen
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| | - Kerstin Ganter
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| | - Christophe Lancrin
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| | - Neil Humphreys
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| | - Mathieu Boulard
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Italy
| |
Collapse
|
50
|
Foretek D, Gabriel M, Morillon A. Analysis of Cytoplasmic RNA Decay Targets Using the Auxin Degron System. Methods Mol Biol 2025; 2863:321-338. [PMID: 39535718 DOI: 10.1007/978-1-0716-4176-7_19] [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: 11/16/2024]
Abstract
RNA degradation in mammalian cells is performed by multiple enzymes and cofactors making it difficult to identify the specific impact of each of them separately. The auxin-inducible degron system enables direct depletion of a protein of interest limiting the time of depletion and thus reducing secondary effects due to cell adaptation. In this chapter, using XRN1 as an example of cytoplasmic RNA decay enzyme, we describe a combination of methods to introduce the auxin-inducible degron by CRISPR-Cas9, together with downstream analyses of RNA levels after protein depletion.
Collapse
Affiliation(s)
- Dominika Foretek
- ncRNA, Epigenetic and Genome Fluidity, Institut Curie, PSL University, Sorbonne Université, CNRS UMR3244, Paris, France
| | - Marc Gabriel
- ncRNA, Epigenetic and Genome Fluidity, Institut Curie, PSL University, Sorbonne Université, CNRS UMR3244, Paris, France
| | - Antonin Morillon
- ncRNA, Epigenetic and Genome Fluidity, Institut Curie, PSL University, Sorbonne Université, CNRS UMR3244, Paris, France.
| |
Collapse
|