1
|
Lebedev M, Chan FY, Rackles E, Bellessem J, Mikeladze-Dvali T, Xavier Carvalho A, Zanin E. Anillin mediates unilateral furrowing during cytokinesis by limiting RhoA binding to its effectors. J Cell Biol 2025; 224:e202405182. [PMID: 40261302 PMCID: PMC12013513 DOI: 10.1083/jcb.202405182] [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: 05/31/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
During unilateral furrow ingression, one side of the cytokinetic ring (leading edge) ingresses before the opposite side (lagging edge). Anillin mediates unilateral furrowing during cytokinesis in the one-cell C. elegans zygote by limiting myosin II accumulation in the ring. Here, we address the role of anillin in this process and show that anillin inhibits not only the accumulation of myosin II but also of other RhoA effectors by binding and blocking the RhoA effector site. The interaction between the anillin's RhoA-binding domain (RBD) and active RhoA is enhanced by the disordered linker region and differentially regulated at the leading and lagging edge, which together results in asymmetric RhoA signaling and accumulation of myosin II. In summary, we discover a RhoA GEF- and GAP-independent mechanism, where RhoA activity is limited by anillin binding to the RhoA effector site. Spatial fine-tuning of anillin's inhibitory role on RhoA signaling enables unilateral furrow ingression and contributes to animal development.
Collapse
Affiliation(s)
- Mikhail Lebedev
- Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fung-Yi Chan
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Elisabeth Rackles
- Department Biology II, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jennifer Bellessem
- Department Biology II, Ludwig-Maximilians University Munich, Munich, Germany
| | | | - Ana Xavier Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Esther Zanin
- Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department Biology II, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
2
|
Puerta D, Rivera-Martín S, Fragoso-Luna A, Strome S, Crittenden SL, Kimble J, Pérez-Martín J. Notch controls APC/C FZR-1 to enable accumulation of chromatin regulators in germline stem cells from Caenorhabditis elegans. SCIENCE ADVANCES 2025; 11:eadu8572. [PMID: 40446035 PMCID: PMC12124362 DOI: 10.1126/sciadv.adu8572] [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: 11/24/2024] [Accepted: 04/25/2025] [Indexed: 06/02/2025]
Abstract
Originally known for its function in the cell cycle, the anaphase-promoting complex/cyclosome (APC/C) also plays a crucial role in regulating differentiation and maintaining cell identity. However, the mechanisms by which APC/C mediates developmental processes are not fully understood. In this study, we show that APC/C and its activator FZR-1 regulate the chromatin regulators MES-4 and MES-3. These proteins are part of histone methylation complexes essential for maintaining germline stem cell (GSC) identity in the germ line of Caenorhabditis elegans. APC/CFZR-1 facilitates the degradation of MES-4 and MES-3 when GSCs transition toward differentiating into oocytes. The activity of APC/CFZR-1 is restricted by the Notch signaling pathway provided by the distal tip cell, which is responsible for maintaining the stemness of the GSC pool. This negative regulation enables the accumulation of MES-3 and MES-4 in GSCs, offering an additional component by which niche activity modulates the C. elegans germ line.
Collapse
Affiliation(s)
- David Puerta
- Instituto de Biomedicina de Valencia (CSIC), Valencia, Spain
- Instituto de Biología Funcional y Genómica (CSIC), Salamanca, Spain
| | | | | | - Susan Strome
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Sarah L Crittenden
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - José Pérez-Martín
- Instituto de Biomedicina de Valencia (CSIC), Valencia, Spain
- Instituto de Biología Funcional y Genómica (CSIC), Salamanca, Spain
| |
Collapse
|
3
|
Warren EC, Brown AEX, Sarkisyan KS. Screening conditions and constructs for attempted genetic transformation of C. elegans by Agrobacterium. PLoS One 2025; 20:e0325060. [PMID: 40424298 PMCID: PMC12111652 DOI: 10.1371/journal.pone.0325060] [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: 09/04/2024] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Manipulating gene expression within a model organism is important for reverse genetic experimentation, and while techniques to generate transgenic C. elegans are available, they are optimised for creating individual lines. The ability to create libraries of genetically modified animals using C. elegans as a model would make new types of experiments possible and would speed up studies of animal physiology. Here, we describe a range of constructs designed to establish a high-throughput method of C. elegans transformation mediated by gene transfer from Agrobacterium. We demonstrate that C. elegans are able to survive on Agrobacterium as a sole food source, and screen conditions for Agrobacterium-mediated transformation in this organism. While we do not achieve routine gene transfer from Agrobacterium to C. elegans, we suggest that this technique has potential following further optimization. The success of the approach would enable rapid and high-throughput transformation of C. elegans, providing an improvement on currently available methods. Here we provide details of optimization conditions tested, and a useful resource of T-binary constructs for use by the scientific community.
Collapse
Affiliation(s)
- Eleanor C. Warren
- MRC Laboratory of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Andre E. X. Brown
- MRC Laboratory of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Karen S. Sarkisyan
- MRC Laboratory of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
4
|
Knoebel E, Brinck A, Nonet ML. Parameters that influence bipartite reporter system expression in Caenorhabditis elegans. Genetics 2025:iyaf076. [PMID: 40341369 DOI: 10.1093/genetics/iyaf076] [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: 02/29/2024] [Accepted: 04/08/2025] [Indexed: 05/10/2025] Open
Abstract
The development of bipartite reporter systems in Caenorhabditis elegans has lagged by more than a decade behind its adoption in Drosophila, the other invertebrate model commonly used to dissect biological mechanisms. Here, we characterize many parameters that influence expression in recently developed C. elegans bipartite systems. We examine how DNA binding site number and spacing influence expression and characterize how these expression parameters vary in distinct tissue types. Furthermore, we examine how both basal promoters and 3' UTR influence the specificity and level of expression. These studies provide both a framework for the rational design of driver and reporter transgenes and molecular and genetic tools for the creation, characterization, and optimization of bipartite system components for expression in other cell types.
Collapse
Affiliation(s)
- Emma Knoebel
- Department of Neuroscience, Washington University Medical School, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Anna Brinck
- Department of Neuroscience, Washington University Medical School, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael L Nonet
- Department of Neuroscience, Washington University Medical School, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Rose AB, Baer A, Shaker I, Monroe JG, Korf I, Rose LS. Introns increase gene expression in Caenorhabditis elegans by a mechanism that must be at least partly different than in plants. Sci Rep 2025; 15:15862. [PMID: 40328889 PMCID: PMC12055998 DOI: 10.1038/s41598-025-99739-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
The wide diversity of organisms in which introns stimulate gene expression suggests that this is an ancient phenomenon. However, the mechanisms through which introns boost expression remain poorly understood, and the degree the which the action of introns is evolutionarily conserved is unknown. Here we compared the effect on expression of introns at different positions and tested ten different introns at the same location in a reporter gene in single-copy transgenic nematodes. The introns boosted expression most when near the start of the gene, as previously observed in several organisms. All ten introns tested at the same position increased mRNA accumulation 10- to 17-fold, in contrast to plants where introns vary widely in their effect on expression and relatively few increase mRNA levels 10-fold or more. These results suggest that some aspects of the mechanisms through which introns boost expression are fundamentally different in nematodes and plants.
Collapse
Affiliation(s)
- Alan B Rose
- Department of Molecular and Cellular Biology, University of California, Davis, 95616, USA.
| | - Aaron Baer
- Department of Molecular and Cellular Biology, University of California, Davis, 95616, USA
| | - Isaac Shaker
- Department of Molecular and Cellular Biology, University of California, Davis, 95616, USA
| | - J Grey Monroe
- Department of Plant Sciences, University of California, Davis, 95616, USA
| | - Ian Korf
- Department of Molecular and Cellular Biology, University of California, Davis, 95616, USA
- Genome Center, University of California, Davis, 95616, USA
| | - Lesilee S Rose
- Department of Molecular and Cellular Biology, University of California, Davis, 95616, USA
| |
Collapse
|
6
|
Engelfriet ML, Guo Y, Arnold A, Valen E, Ciosk R. Reprograming gene expression in 'hibernating' C. elegans involves the IRE-1/XBP-1 pathway. eLife 2025; 13:RP101186. [PMID: 40326887 PMCID: PMC12055002 DOI: 10.7554/elife.101186] [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] [Indexed: 05/07/2025] Open
Abstract
In nature, many animals respond to cold by entering hibernation, while in clinical settings, controlled cooling is used in transplantation and emergency medicine. However, the molecular mechanisms that enable cells to survive severe cold are still not fully understood. One key aspect of cold adaptation is the global downregulation of protein synthesis. Studying it in the nematode Caenorhabditis elegans, we find that the translation of most mRNAs continues in the cold, albeit at a slower rate, and propose that cold-specific gene expression is regulated primarily at the transcription level. Supporting this idea, we found that the transcription of certain cold-induced genes is linked to the activation of unfolded protein response (UPR) through the conserved IRE-1/XBP-1 signaling pathway. Our findings suggest that this pathway is triggered by cold-induced perturbations in proteins and lipids within the endoplasmic reticulum, and that its activation is beneficial for cold survival.
Collapse
Affiliation(s)
- Melanie Lianne Engelfriet
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| | - Yanwu Guo
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| | - Andreas Arnold
- Division of Molecular Neuroscience, Department of Biomedicine, University of BaselBaselSwitzerland
- University Psychiatric Clinics, University of BaselBaselSwitzerland
| | - Eivind Valen
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| | - Rafal Ciosk
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| |
Collapse
|
7
|
Rivera DE, Poirier K, Moore S, Nicolle O, Morgan E, Longares JF, Singh A, Michaux G, Félix MA, Luallen RJ. Dynamics of gut colonization by commensal and pathogenic bacteria that attach to the intestinal epithelium. NPJ Biofilms Microbiomes 2025; 11:70. [PMID: 40319018 PMCID: PMC12049552 DOI: 10.1038/s41522-025-00696-9] [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: 10/28/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Bacterial adherence to the intestinal epithelium plays a role in niche establishment in the gut lumen. Through sampling natural populations of Caenorhabditis, we discovered several bacterial species that adhere to the intestinal epithelium via polar, intimate association, best described as attachment. These bacteria had varying effects on host fitness and physiology, with one species having negative effects, and the others exhibiting neutral effects. These bacteria can actively divide in the gut lumen, either replicating throughout the gut simultaneously or anteroposteriorly. In competition assays, animals pre-colonized with an attaching commensal bacteria reduced colonization by the pathogenic bacteria, but this effect was not seen when animals were colonized by both species simultaneously. Regardless of the colonization paradigm, populations exposed to both bacteria showed a near-identical mitigation of the pathogenic effects. Altogether, these strains illustrate the capacity of microbiome bacteria to attach, replicate, and establish a niche across the entire intestinal lumen.
Collapse
Affiliation(s)
- Dalaena E Rivera
- Department of Biology, San Diego State University, San Diego, USA
| | - Kayla Poirier
- Department of Biology, San Diego State University, San Diego, USA
| | - Samuel Moore
- Department of Biology, San Diego State University, San Diego, USA
| | - Ophélie Nicolle
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) -UMR 6290, F-35000, Rennes, France
| | - Emily Morgan
- Department of Biology, San Diego State University, San Diego, USA
| | | | - Anupama Singh
- Department of Biology, San Diego State University, San Diego, USA
| | - Grégoire Michaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) -UMR 6290, F-35000, Rennes, France
| | - Marie-Anne Félix
- Institut de Biologie de l'École Normale Supérieure, Centre National de la Recherche Scientifique, INSERM, École Normale Supérieure, Paris Sciences et Lettres, Paris, France.
| | - Robert J Luallen
- Department of Biology, San Diego State University, San Diego, USA.
| |
Collapse
|
8
|
Mul W, Mitra A, Prevo B, Peterman EJG. DYF-5 regulates intraflagellar transport by affecting train turnaround. Mol Biol Cell 2025; 36:ar53. [PMID: 40072497 DOI: 10.1091/mbc.e24-08-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Intraflagellar transport (IFT) coordinates the transport of cargo in cilia and is essential for ciliary function. CILK1 has been identified as a key regulator of IFT. The mechanism by which it acts has, however, remained unclear. In this study, we use fluorescence imaging and single-molecule tracking in the phasmid cilia of live Caenorhabditis elegans to study the effect of the CILK1 homologue DYF-5 on the dynamics of the IFT. We show that in the absence of DYF-5, IFT components accumulate at the ciliary tip. Kinesin-II is no longer restricted to the proximal segment of the cilium but is present throughout the cilium, while its velocity is different from that of OSM-3. The frequency of IFT trains is reduced and in particular retrograde trains were rarely observed. In the absence of DYF-5, retrograde transport is vastly reduced, resulting in the accumulation of IFT components at the tip and depletion at the base. The latter results in impeded anterograde train assembly, resulting in fewer trains with irregular composition. Our results show that DYF-5 plays a key role in regulating the turnarounds of IFT trains at the ciliary tip.
Collapse
Affiliation(s)
- Wouter Mul
- Department of Physics and Astronomy, and LaserLaB, Vrije Universiteit Amsterdam, The Netherlands 1081HV
| | - Aniruddha Mitra
- Department of Physics and Astronomy, and LaserLaB, Vrije Universiteit Amsterdam, The Netherlands 1081HV
| | - Bram Prevo
- Department of Physics and Astronomy, and LaserLaB, Vrije Universiteit Amsterdam, The Netherlands 1081HV
| | - Erwin J G Peterman
- Department of Physics and Astronomy, and LaserLaB, Vrije Universiteit Amsterdam, The Netherlands 1081HV
| |
Collapse
|
9
|
Martins CS, Iv F, Suman SK, Panagiotou TC, Sidor C, Ruso-López M, Plancke CN, Omi S, Pagès R, Gomes M, Llewellyn A, Bandi SR, Ramond L, Arbizzani F, Rimoli CV, Schnorrer F, Robin F, Wilde A, LeGoff L, Pedelacq JD, Jégou A, Cabantous S, Rincon SA, Chandre C, Brasselet S, Mavrakis M. Genetically encoded reporters of actin filament organization in living cells and tissues. Cell 2025; 188:2540-2559.e27. [PMID: 40179884 DOI: 10.1016/j.cell.2025.03.003] [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: 03/04/2024] [Revised: 12/09/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
The cytoskeletal protein actin is crucial for cell shape and integrity throughout eukaryotes. Actin filaments perform essential biological functions, including muscle contraction, cell division, and tissue morphogenesis. These diverse activities are achieved through the ability of actin filaments to be arranged into precise architectures. Much progress has been made in defining the proteome of the actin cytoskeleton, but a detailed appreciation of the dynamic organizational state of the actin filaments themselves has been hindered by available tools. Fluorescence polarization microscopy is uniquely placed for measuring actin filament organization by exploiting the sensitivity of polarized light excitation to the orientation of fluorophores attached to actin filaments. By engineering fusions of five widely used actin localization reporters to fluorescent proteins with constrained mobility, we have succeeded in developing genetically encoded, green- and red-fluorescent-protein-based reporters for non-invasive, quantitative measurements of actin filament organization in living cells and tissues by fluorescence polarization microscopy.
Collapse
Affiliation(s)
- Carla Silva Martins
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - François Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Shashi Kumar Suman
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Thomas C Panagiotou
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1M1, Canada
| | - Clara Sidor
- Aix Marseille Univ, CNRS, IBDM, Turing Centre for Living Systems, 13009 Marseille, France
| | - María Ruso-López
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca 37007, Spain
| | - Camille N Plancke
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Rebecca Pagès
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Maxime Gomes
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Alexander Llewellyn
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Sourish Reddy Bandi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Laurie Ramond
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | | | - Caio Vaz Rimoli
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Frank Schnorrer
- Aix Marseille Univ, CNRS, IBDM, Turing Centre for Living Systems, 13009 Marseille, France
| | - François Robin
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Andrew Wilde
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1M1, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1M1, Canada
| | - Loïc LeGoff
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Jean-Denis Pedelacq
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Stéphanie Cabantous
- Centre de Recherche en Cancérologie de Toulouse (CRCT), Inserm, Université Paul Sabatier - Toulouse III, CNRS, 31037 Toulouse, France
| | - Sergio A Rincon
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca 37007, Spain
| | | | - Sophie Brasselet
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France.
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France.
| |
Collapse
|
10
|
Quintin S, Saad MI, Amann G, Reymann AC. In vivo detection of ALFA-tagged proteins in C. elegans with a transgenic fluorescent nanobody. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001542. [PMID: 40321834 PMCID: PMC12048842 DOI: 10.17912/micropub.biology.001542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025]
Abstract
To track tagged endogenous proteins in vivo , we created a C. elegans strain expressing a fluorescently-labelled nanobody directed against the ALFA-tag epitope. The strain, which expresses an anti-ALFA nanobody fused to mKate2, is healthy and allows clear detection of the ALFA-tagged junction protein DLG-1 at all stages. This method is adapted for live imaging, circumvents the need of immuno-histochemistry, and opens perspective to study protein function in vivo . The future detection of sensitive proteins can therefore be envisaged in nematodes by using transgenic nanobodies, or chromobodies, in combination with ALFA-tagging by CRISPR.
Collapse
Affiliation(s)
- Sophie Quintin
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
- CNRS, UMR 7104, F-67400 Illkirch, France
- Inserm, UMR-S 1258, F-67400 Illkirch, France
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67400 Illkirch, France
| | - Maria Izabella Saad
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
- CNRS, UMR 7104, F-67400 Illkirch, France
- Inserm, UMR-S 1258, F-67400 Illkirch, France
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67400 Illkirch, France
| | - Grégory Amann
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
- CNRS, UMR 7104, F-67400 Illkirch, France
- Inserm, UMR-S 1258, F-67400 Illkirch, France
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67400 Illkirch, France
| | - Anne-Cécile Reymann
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
- CNRS, UMR 7104, F-67400 Illkirch, France
- Inserm, UMR-S 1258, F-67400 Illkirch, France
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67400 Illkirch, France
| |
Collapse
|
11
|
Carrick BH, Crittenden SL, Linsley M, Dos Santos SJC, Wickens M, Kimble J. The PUF RNA-binding protein, FBF-2, maintains stem cells without binding to RNA. RNA (NEW YORK, N.Y.) 2025; 31:623-632. [PMID: 39984282 PMCID: PMC12001965 DOI: 10.1261/rna.080307.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
Like all canonical PUF proteins, Caenorhabditis elegans FBF-2 binds to specific RNAs via tripartite recognition motifs. Here, we report that an FBF-2 mutant protein that cannot bind to RNA is nonetheless biologically active and maintains stem cells. This unexpected result challenges the conventional wisdom that RBPs must bind to RNAs to achieve biological activity. Also unexpectedly, FBF-2 interactions with partner proteins can compensate for the loss of RNA binding. FBF-2 only loses biological activity when its RNA-binding and partner interactions are both defective. These findings highlight the complementary contributions of RNA-binding and protein partner interactions to the activity of an RNA-binding protein.
Collapse
Affiliation(s)
- Brian H Carrick
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Sarah L Crittenden
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - MaryGrace Linsley
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | - Marvin Wickens
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
12
|
Yang B, Galletta B, Rusan N, McJunkin K. An intrinsically disordered region of Drosha selectively promotes miRNA biogenesis, independent of tissue-specific Microprocessor condensates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.10.648254. [PMID: 40291697 PMCID: PMC12027344 DOI: 10.1101/2025.04.10.648254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Precise control of miRNA biogenesis is of extreme importance, since mis-regulation of miRNAs underlies or exacerbates many disease states. The Microprocessor complex, composed of DROSHA and DGCR8, carries out the first cleavage step in canonical miRNA biogenesis. Despite recent advances in understanding the molecular mechanism of Microprocessor, the N-terminal region of DROSHA is less characterized due its high intrinsic disorder. Here we demonstrate that Microprocessor forms condensates with properties consistent with liquid-liquid phase separation (LLPS) in select tissues in C. elegans . While DRSH-1/Drosha recruitment to granules is only partially dependent on its intrinsically disordered regions (IDRs), one of these N-terminal IDRs is crucial for biogenesis of a subset of miRNAs and normal development. A cis region of an IDR-dependent miRNA confers IDR-dependence to another miRNA, suggesting that the IDR recognizes sequences or structures in the miRNA primary transcript. Future studies will further elucidate the specificity of this interaction and the putative role of Microprocessor condensates.
Collapse
|
13
|
Mitra A, Gioukakis E, Mul W, Peterman EJG. Delivery of intraflagellar transport proteins to the ciliary base and assembly into trains. SCIENCE ADVANCES 2025; 11:eadr1716. [PMID: 40184459 PMCID: PMC11970479 DOI: 10.1126/sciadv.adr1716] [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: 06/18/2024] [Accepted: 03/03/2025] [Indexed: 04/06/2025]
Abstract
Anterograde intraflagellar transport (IFT) trains, composed of IFT-B, IFT-A, and BBSome subcomplexes, are responsible for transporting ciliary proteins into the cilium. How IFT subcomplexes reach the ciliary base and assemble into IFT trains is poorly understood. Here, we perform quantitative single-molecule imaging in Caenorhabditis elegans chemosensory cilia to uncover how IFT subcomplexes arrive at the base, organize in IFT trains, and enter the cilium. We find that BBSomes reach the base via diffusion where they either associate with assembling IFT trains or with the membrane surrounding the base. In contrast, IFT-B and IFT-A reach the base via directed transport most likely on vesicles that stop at distinct locations near the base. Individual subcomplexes detach from the vesicles into a diffusive pool and associate to assembling trains. Our results show that IFT-B is first incorporated into IFT trains, followed by IFT-A, and finally BBSomes, indicating that the assembly of IFT trains is a highly regulated, step-wise process.
Collapse
Affiliation(s)
| | - Evangelos Gioukakis
- Department of Physics and Astronomy and LaserLaB, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wouter Mul
- Department of Physics and Astronomy and LaserLaB, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Erwin J. G. Peterman
- Department of Physics and Astronomy and LaserLaB, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
Jin Q, Feng X, Hong M, Wang K, Chen X, Cheng J, Kuang Y, Si X, Xu M, Huang X, Guang S, Zhu C. Peri-centrosomal localization of small interfering RNAs in C. elegans. SCIENCE CHINA. LIFE SCIENCES 2025; 68:895-911. [PMID: 39825209 DOI: 10.1007/s11427-024-2818-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/16/2024] [Indexed: 01/20/2025]
Abstract
The centrosome is the microtubule-organizing center and a crucial part of cell division. Centrosomal RNAs (cnRNAs) have been reported to enable precise spatiotemporal control of gene expression during cell division in many species. Whether and how cnRNAs exist in C. elegans are unclear. Here, using the nuclear RNAi Argonaute protein NRDE-3 as a reporter, we observed potential peri-centrosome localized small interfering (si)RNAs in C. elegans. NRDE-3 was previously shown to associate with pre-mRNAs and pre-rRNAs via a process involving the presence of complementary siRNAs. We generated a GFP-NRDE-3 knock-in transgene through CRISPR/Cas9 technology and observed that NRDE-3 formed peri-centrosomal foci neighboring the tubulin protein TBB-2, other centriole proteins and pericentriolar material (PCM) components in C. elegans embryos. The peri-centrosomal accumulation of NRDE-3 depends on RNA-dependent RNA polymerase (RdRP)-synthesized 22G siRNAs and the PAZ domain of NRDE-3, which is essential for siRNA binding. Mutation of eri-1, ergo-1, or drh-3 significantly increased the percentage of pericentrosome-enriched NRDE-3. At the metaphase of the cell cycle, NRDE-3 was enriched in both the peri-centrosomal region and the spindle. Moreover, the integrity of centriole proteins and pericentriolar material (PCM) components is also required for the peri-centrosomal accumulation of NRDE-3. Therefore, we concluded that siRNAs could accumulate in the pericentrosomal region in C. elegans and suggested that the peri-centrosomal region may also be a platform for RNAi-mediated gene regulation.
Collapse
Affiliation(s)
- Qile Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Xuezhu Feng
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Minjie Hong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Ke Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Xiangyang Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Jiewei Cheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Yan Kuang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Xiaoyue Si
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Mingjing Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China
| | - Xinya Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China.
| | - Shouhong Guang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China.
| | - Chengming Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
15
|
Sun Y, Zhou J, Debnath A, Xie B, Wang Z, Jin Y. Multiple regulators constrain the abundance of Caenorhabditis elegans DLK-1 in ciliated sensory neurons. G3 (BETHESDA, MD.) 2025; 15:jkaf004. [PMID: 39854273 PMCID: PMC11917482 DOI: 10.1093/g3journal/jkaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025]
Abstract
The conserved MAP3K DLKs are widely known for their functions in synapse formation, axonal regeneration and degeneration, and neuronal survival, notably under traumatic injury and chronic disease conditions. In contrast, their roles in other neuronal compartments are much less explored. Through an unbiased forward genetic screening in C. elegans for altered patterns of GFP-tagged DLK-1 expressed from the endogenous locus, we have recently uncovered a mechanism by which the abundance of DLK-1 is tightly regulated by intraflagellar transport in ciliated sensory neurons. Here, we report additional mutants identified from the genetic screen. Most mutants exhibit increased accumulation of GFP::DLK-1 in sensory endings, and the levels of misaccumulated GFP::DLK-1 are exacerbated by loss of function in cebp-1, the b-Zip transcription factor acting downstream of DLK-1. We identify several new mutations in genes encoding proteins functioning in intraflagellar transport and cilia assembly, in components of BBSome, MAPK-15, and DYF-5 kinases. We report a novel mutation in the chaperone HSP90 that causes misaccumulation of GFP::DLK-1 and up-regulation of CEBP-1 selectively in ciliated sensory neurons. We also find that the guanylate cyclase ODR-1 constrains GFP::DLK-1 abundance throughout cilia and dendrites of AWC neurons. Moreover, in odr-1 mutants, AWC cilia display distorted morphology, which is ameliorated by loss of function in dlk-1 or cebp-1. These data expand the landscape of DLK-1 signaling in ciliated sensory neurons and underscore a high degree of cell- and neurite- specific regulation.
Collapse
Affiliation(s)
- Yue Sun
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Junxiang Zhou
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Arunima Debnath
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Bokun Xie
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhiping Wang
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
16
|
Liontis T, Senchuk MM, Zhu S, Jacob-Tomas S, Anglas U, Traa A, Soo SK, Van Raamsdonk JM. Intestine-specific disruption of mitochondrial superoxide dismutase extends longevity. Free Radic Biol Med 2025; 229:195-205. [PMID: 39827921 DOI: 10.1016/j.freeradbiomed.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/01/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen containing molecules that are generated by normal metabolism. While ROS can cause damage to the building blocks that make up cells, these molecules can also act as intracellular signals that promote longevity. The levels of ROS within the cell can be regulated by antioxidant enzymes, such as superoxide dismutase (SOD), which converts superoxide to hydrogen peroxide. Interestingly, our previous work has shown that disruption of the mitochondrial SOD gene sod-2 results in increased lifespan, suggesting that elevating levels of mitochondrial superoxide can promote longevity. To explore the molecular mechanisms involved, we determined the tissues in which disruption of sod-2 is necessary for lifespan extension and the tissues in which disruption of sod-2 is sufficient to extend lifespan. We found that tissue-specific restoration of SOD-2 expression in worms lacking SOD-2 could partially revert changes in fertility, embryonic lethality and resistance to stress, but did not inhibit the effects of sod-2 deletion on lifespan. Knocking down sod-2 expression using RNA interference specifically in the intestine, but not other tissues, was sufficient to extend longevity. Intestine-specific knockdown of sod-2 also increased resistance to heat stress while decreasing resistance to oxidative stress. Combined, these results indicate that disruption of sod-2 in neurons, intestine, germline, or muscle is not required for lifespan extension, but that decreasing sod-2 expression in just the intestine extends lifespan. This work defines the conditions required for disruption of mitochondrial superoxide dismutase to increase longevity.
Collapse
Affiliation(s)
- Thomas Liontis
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Megan M Senchuk
- Laboratory of Aging and Neurodegenerative Disease (LAND), Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Shusen Zhu
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Suleima Jacob-Tomas
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Ulrich Anglas
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Annika Traa
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Sonja K Soo
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jeremy M Van Raamsdonk
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Laboratory of Aging and Neurodegenerative Disease (LAND), Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA; Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
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
|
18
|
Schwartz HT, Sternberg PW. A sequencing-based screening method identifies regulators of EGFR signaling from nonviable mutants in Caenorhabditis elegans. Sci Signal 2025; 18:eadp9377. [PMID: 39999212 DOI: 10.1126/scisignal.adp9377] [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: 04/18/2024] [Revised: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Suppressor screens can identify genetic modifiers of biochemical pathways but generally require that the suppressed mutant be viable and fertile. We developed a screening method that obviated this requirement and enabled the identification of mutations that partially suppressed the early developmental arrest and lethality caused by loss of the epidermal growth factor (EGF) receptor ortholog LET-23 in Caenorhabditis elegans. We chemically mutagenized animals carrying the loss-of-function allele let-23(sy15), recovered let-23(sy15) homozygotes that escaped early developmental arrest but were nevertheless inviable, and sequenced their genomes. Testing of candidate causal mutations identified 11 genes that, when mutated, mitigated the early lethality caused by loss of EGF signaling. These included genes encoding homologs of the small guanosine triphosphatase (GTPase) Ras (let-60), which is a downstream effector of LET-23, and of regulators of the small GTPase Rho, including the homolog of the phosphotyrosine-binding protein TENSIN (tns-1). We also recovered suppressing mutations in genes encoding nuclear proteins that protect against DNA damage, including the homolog of MutS homolog 4 (him-14). Genetic experiments were consistent with the repression of Rho activity or the activation of the DNA damage response compensating for the loss of EGF signaling. This sequencing-based, whole-animal screening method may be adapted to other organisms to enable the identification of mutations for which the phenotype does not allow the recovery of viable animals.
Collapse
Affiliation(s)
- Hillel T Schwartz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Paul W Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
19
|
Garcia-Sanchez JA, Bonnet E, Loubatier C, Doye A, Paillier G, Segui F, Larbret F, Chaintreuil P, Batistic L, Torre C, Deckert M, Polanowska J, Munro P, Boyer L, Visvikis O. Evolutionary conserved regulation of TFEB stability by the E3 ubiquitin ligase WWP2 modulates response to stress in vivo. iScience 2025; 28:111838. [PMID: 39995862 PMCID: PMC11848471 DOI: 10.1016/j.isci.2025.111838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Transcription factor EB (TFEB) is a key transcription factor that orchestrates the cellular response to stress. Dysregulation of TFEB is associated with a range of human diseases, and understanding the regulatory mechanisms of TFEB is crucial for identifying potential drug targets. In this study, we used Caenorhabditis elegans to screen for E3 ubiquitin ligases regulating the activity of TFEB's homolog, HLH-30, upon pathogenic infection. We identified WWP-1 as a regulator of HLH-30-dependent immune response controlling HLH-30 stability to mediate host defense in vivo. We found that HLH-30 interacts with WWP-1, supporting a model of WWP-1 directly regulating HLH-30. Furthermore, we found that WWP-1's human homolog WWP2 binds TFEB, directly induces TFEB ubiquitination and stabilizes TFEB. Finally, we found that WWP2 is required for TFEB-dependent host response in human monocytes-derived macrophages upon infection. Overall, our work has identified an evolutionarily conserved regulation of TFEB by WWP2 and highlighted its role in modulating stress response.
Collapse
Affiliation(s)
| | - Estelle Bonnet
- Université Côte d’Azur, INSERM, C3M, Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur, Centre Scientifique de Monaco, Monaco, Monaco
| | | | - Anne Doye
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | - Fabien Segui
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | | | | | - Cédric Torre
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | | | | | | | | |
Collapse
|
20
|
Borchers C, Osburn K, Roh HC, Aoki ST. In vivo pulse-chase in C. elegans reveals intestinal histone turnover changes upon starvation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638128. [PMID: 39990428 PMCID: PMC11844474 DOI: 10.1101/2025.02.13.638128] [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: 02/25/2025]
Abstract
The ability to study protein dynamics and function in the authentic context of a multicellular organism is paramount to better understand biological phenomena in animal health and disease. Pulse-chase of self-labeling fusion protein tags provide the opportunity to label proteins of interest and track those proteins over time. There are currently several challenges associated with performing in vivo protein pulse-chase in animals, such as cost, reproducibility, and accurate detection methods. The C. elegans model organism has attributes that alleviate many of these challenges. This work tests the feasibility of applying the Halo modified enzyme (HaloTag) for in vivo protein pulse-chase in C. elegans. HaloTag intestinal histone reporters were created in the worm and used to demonstrate that reporter protein could be efficiently pulse-labeled by soaking animals in ligand. Labeled protein stability could be monitored over time by fluorescent confocal microscopy. Further investigation revealed reporter protein stability was dependent on the animal's nutritional state. Chromatin Immunoprecipitation and sequencing (ChIP-seq) of the reporters showed incorporation in chromatin with little change hours into starvation, implying a lack of chromatin regulation at the time point tested. Collectively, this work presents a straightforward method to label and track proteins of interest in C. elegans that can address a multitude of biological questions surrounding protein stability and dynamics in this animal model.
Collapse
Affiliation(s)
- Christopher Borchers
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
- Indiana BioMedical Gateway (IBMG) Program; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| | - Kara Osburn
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| | - Hyun Cheol Roh
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| | - Scott T. Aoki
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| |
Collapse
|
21
|
Neves AR, Čavka I, Rausch T, Köhler S. Crossovers are regulated by a conserved and disordered synaptonemal complex domain. Nucleic Acids Res 2025; 53:gkaf095. [PMID: 39964475 PMCID: PMC11833701 DOI: 10.1093/nar/gkaf095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
During meiosis, the number and distribution of crossovers (COs) must be precisely regulated through CO assurance and interference to prevent chromosome missegregation and genomic instability in the progeny. Here we show that this regulation of COs depends on a disordered and conserved domain within the synaptonemal complex (SC). This domain is located at the C-terminus of the central element protein SYP-4 in Caenorhabditis elegans. While not necessary for synapsis, the C-terminus of SYP-4 is crucial for both CO assurance and interference. Although the SYP-4 C-terminus contains many potential phosphorylation sites, we found that phosphorylation is not the primary regulator of CO events. Instead, we discovered that nine conserved phenylalanines are required to recruit a pro-CO factor predicted to be an E3 ligase and regulate the physical properties of the SC. We propose that this conserved and disordered domain plays a crucial role in maintaining the SC in a state that allows transmitting signals to regulate CO formation. While the underlying mechanisms remain to be fully understood, our findings align with existing models suggesting that the SC plays a critical role in determining the number and distribution of COs along chromosomes, thereby safeguarding the genome for future generations.
Collapse
Affiliation(s)
- Ana Rita Rodrigues Neves
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, 69117 Heidelberg University, Heidelberg, Germany
| | - Ivana Čavka
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, 69117 Heidelberg University, Heidelberg, Germany
| | - Tobias Rausch
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- GeneCore, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Simone Köhler
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| |
Collapse
|
22
|
Alves Domingos H, Green M, Ouzounidis VR, Finlayson C, Prevo B, Cheerambathur DK. The kinetochore protein KNL-1 regulates the actin cytoskeleton to control dendrite branching. J Cell Biol 2025; 224:e202311147. [PMID: 39625434 PMCID: PMC11613958 DOI: 10.1083/jcb.202311147] [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: 11/23/2023] [Revised: 07/23/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The function of the nervous system is intimately tied to its complex and highly interconnected architecture. Precise control of dendritic branching in individual neurons is central to building the complex structure of the nervous system. Here, we show that the kinetochore protein KNL-1 and its associated KMN (Knl1/Mis12/Ndc80 complex) network partners, typically known for their role in chromosome-microtubule coupling during mitosis, control dendrite branching in the Caenorhabditis elegans mechanosensory PVD neuron. KNL-1 restrains excess dendritic branching and promotes contact-dependent repulsion events, ensuring robust sensory behavior and preventing premature neurodegeneration. Unexpectedly, KNL-1 loss resulted in significant alterations of the actin cytoskeleton alongside changes in microtubule dynamics within dendrites. We show that KNL-1 modulates F-actin dynamics to generate proper dendrite architecture and that its N-terminus can initiate F-actin assembly. These findings reveal that the postmitotic neuronal KMN network acts to shape the developing nervous system by regulating the actin cytoskeleton and provide new insight into the mechanisms controlling dendrite architecture.
Collapse
Affiliation(s)
- Henrique Alves Domingos
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Mattie Green
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Vasileios R. Ouzounidis
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Cameron Finlayson
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Bram Prevo
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Dhanya K. Cheerambathur
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
23
|
Sorensen Turpin CG, Sloan D, LaForest M, Klebanow L, Mitchell D, Severson AF, Bembenek JN. Securin regulates the spatiotemporal dynamics of separase. J Cell Biol 2025; 224:e202312099. [PMID: 39556062 PMCID: PMC11574863 DOI: 10.1083/jcb.202312099] [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: 12/20/2023] [Revised: 07/25/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
Separase regulates multiple aspects of the metaphase-to-anaphase transition. Separase cleaves cohesin to allow chromosome segregation and localizes to vesicles to promote exocytosis. The anaphase-promoting complex/cyclosome (APC/C) activates separase by ubiquitinating its inhibitory chaperone, securin, triggering its degradation. How this pathway controls the exocytic function of separase is unknown. During meiosis I, securin is degraded over several minutes, while separase rapidly relocalizes from kinetochore structures at the spindle and cortex to sites of action on chromosomes and vesicles at anaphase onset. The loss of cohesin coincides with the relocalization of separase to the chromosome midbivalent at anaphase onset. APC/C depletion prevents separase relocalization, while securin depletion causes precocious separase relocalization. Expression of non-degradable securin inhibits chromosome segregation, exocytosis, and separase localization to vesicles but not to the anaphase spindle. We conclude that APC/C-mediated securin degradation controls separase localization. This spatiotemporal regulation will impact the effective local concentration of separase for more precise targeting of substrates in anaphase.
Collapse
Affiliation(s)
- Christopher G. Sorensen Turpin
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dillon Sloan
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Marian LaForest
- Columbia University, Herbert Irving Comprehensive Cancer Center, NYC, New York, NY, USA
| | | | - Diana Mitchell
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Aaron F. Severson
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH, USA
| | - Joshua N. Bembenek
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
24
|
Lee HJ, Liang J, Chaudhary S, Moon S, Yu Z, Wu T, Liu H, Choi MK, Zhang Y, Lu H. Automated cell annotation in multi-cell images using an improved CRF_ID algorithm. eLife 2025; 12:RP89050. [PMID: 39853076 PMCID: PMC11759411 DOI: 10.7554/elife.89050] [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] [Indexed: 01/26/2025] Open
Abstract
Cell identification is an important yet difficult process in data analysis of biological images. Previously, we developed an automated cell identification method called CRF_ID and demonstrated its high performance in Caenorhabditis elegans whole-brain images (Chaudhary et al., 2021). However, because the method was optimized for whole-brain imaging, comparable performance could not be guaranteed for application in commonly used C. elegans multi-cell images that display a subpopulation of cells. Here, we present an advancement, CRF_ID 2.0, that expands the generalizability of the method to multi-cell imaging beyond whole-brain imaging. To illustrate the application of the advance, we show the characterization of CRF_ID 2.0 in multi-cell imaging and cell-specific gene expression analysis in C. elegans. This work demonstrates that high-accuracy automated cell annotation in multi-cell imaging can expedite cell identification and reduce its subjectivity in C. elegans and potentially other biological images of various origins.
Collapse
Affiliation(s)
- Hyun Jee Lee
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Jingting Liang
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Shivesh Chaudhary
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Sihoon Moon
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Zikai Yu
- Interdisciplinary BioEngineering Program, Georgia Institute of TechnologyAtlantaUnited States
| | - Taihong Wu
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - He Liu
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Myung-Kyu Choi
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
- Interdisciplinary BioEngineering Program, Georgia Institute of TechnologyAtlantaUnited States
| |
Collapse
|
25
|
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
|
26
|
Khan H, Huang X, Raj V, Wang H. A versatile site-directed gene trap strategy to manipulate gene activity and control gene expression in Caenorhabditis elegans. PLoS Genet 2025; 21:e1011541. [PMID: 39841730 PMCID: PMC11753634 DOI: 10.1371/journal.pgen.1011541] [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: 08/30/2024] [Accepted: 12/15/2024] [Indexed: 01/24/2025] Open
Abstract
The ability to manipulate gene activity and control transgene expression is essential to study gene function. While several genetic tools for modifying genes or controlling expression separately are available for Caenorhabditis elegans, there are no genetic approaches to generate mutations that simultaneously disrupt gene function and provide genetic access to the cells expressing the disrupted gene. To achieve this, we developed a versatile gene trap strategy based on cGAL, a GAL4-UAS bipartite expression system for C. elegans. We designed a cGAL gene trap cassette and used CRISPR/Cas9 to insert it into the target gene, creating a bicistronic operon that simultaneously expresses a truncated endogenous protein and the cGAL driver in the cells expressing the target gene. We demonstrate that our cGAL gene trap strategy robustly generated loss-of-function alleles. Combining the cGAL gene trap lines with different UAS effector strains allowed us to rescue the loss-of-function phenotype, observe the gene expression pattern, and manipulate cell activity spatiotemporally. We show that, by recombinase-mediated cassette exchange (RMCE) via microinjection or genetic crossing, the cGAL gene trap lines can be further engineered in vivo to easily swap cGAL with other bipartite expression systems' drivers, including QF/QF2, Tet-On/Tet-Off, and LexA, to generate new gene trap lines with different drivers at the same genomic locus. These drivers can be combined with their corresponding effectors for orthogonal transgenic control. Thus, our cGAL-based gene trap is versatile and represents a powerful genetic tool for gene function analysis in C. elegans, which will ultimately provide new insights into how genes in the genome control the biology of an organism.
Collapse
Affiliation(s)
- Haania Khan
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xinyu Huang
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Vishnu Raj
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Han Wang
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
27
|
Kim J, Dutta N, Vega M, Bong A, Averbuhk M, Barahona RA, Alcala A, Holmes JT, Garcia G, Higuchi-Sanabria R. Cross comparison of imaging strategies of mitochondria in C. elegans during aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.24.630282. [PMID: 39763886 PMCID: PMC11703187 DOI: 10.1101/2024.12.24.630282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Mitochondria are double membrane-bound organelles with pleiotropic roles in the cell, including energy production through aerobic respiration, calcium signaling, metabolism, proliferation, immune signaling, and apoptosis. Dysfunction of mitochondria is associated with numerous physiological consequences and drives various diseases, and is one of twelve biological hallmarks of aging, linked to aging pathology. There are many distinct changes that occur to the mitochondria during aging including changes in mitochondrial morphology, which can be used as a robust and simple readout of mitochondrial quality and function. Although mitochondrial morphology alone cannot be used to conclude the quality of mitochondria, it is highly correlated with mitochondrial function whereby mitochondria exhibit increased fragmentation with age in multiple cell types of the nematode C. elegans. Thus, C. elegans serve as a robust model for rapidly measuring mitochondrial morphology changes during aging. To standardize imaging methods for mitochondrial morphology in C. elegans, we provide a detailed comparative characterization of several transgenic constructs, highlighting benefits and caveats for aging biology studies.
Collapse
Affiliation(s)
- Juri Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Naibedya Dutta
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Matthew Vega
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Andrew Bong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Maxim Averbuhk
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Rebecca Aviles Barahona
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Athena Alcala
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Jacob T. Holmes
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Gilberto Garcia
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Ryo Higuchi-Sanabria
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
28
|
Almeida AC, Rocha H, Raas MWD, Witte H, Sommer RJ, Snel B, Kops GJPL, Gassmann R, Maiato H. An evolutionary perspective on the relationship between kinetochore size and CENP-E dependence for chromosome alignment. J Cell Sci 2024; 137:jcs263466. [PMID: 39698944 PMCID: PMC11827601 DOI: 10.1242/jcs.263466] [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: 08/02/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Chromosome alignment during mitosis can occur as a consequence of bi-orientation or is assisted by the CENP-E (kinesin-7) motor at kinetochores. We previously found that Indian muntjac chromosomes with larger kinetochores bi-orient more efficiently and are biased to align in a CENP-E-independent manner, suggesting that CENP-E dependence for chromosome alignment negatively correlates with kinetochore size. Here, we used targeted phylogenetic profiling of CENP-E in monocentric (localized centromeres) and holocentric (centromeres spanning the entire chromosome length) clades to test this hypothesis at an evolutionary scale. We found that, despite being present in common ancestors, CENP-E was lost more frequently in taxa with holocentric chromosomes, such as Hemiptera and Nematoda. Functional experiments in two nematodes with holocentric chromosomes in which a CENP-E ortholog is absent (Caenorhabditis elegans) or present (Pristionchus pacificus) revealed that targeted expression of human CENP-E to C. elegans kinetochores partially rescued chromosome alignment defects associated with attenuated polar-ejection forces, whereas CENP-E inactivation in P. pacificus had no detrimental effects on mitosis and viability. These data showcase the dispensability of CENP-E for mitotic chromosome alignment in species with larger kinetochores.
Collapse
Affiliation(s)
- Ana C. Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
| | - Helder Rocha
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
- Escola Superior de Saúde, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 420-075 Porto, Portugal
| | - Maximilian W. D. Raas
- Oncode Institute, Hubrecht Institute – KNAW, and University Medical Center Utrecht, 3584 CT, Utrecht, Netherlands
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, 384 CH Utrecht, the Netherlands
| | - Hanh Witte
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology, Max-Planck-Ring 9, 776 Tuebingen, Germany
| | - Ralf J. Sommer
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology, Max-Planck-Ring 9, 776 Tuebingen, Germany
| | - Berend Snel
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, 384 CH Utrecht, the Netherlands
| | - Geert J. P. L. Kops
- Oncode Institute, Hubrecht Institute – KNAW, and University Medical Center Utrecht, 3584 CT, Utrecht, Netherlands
| | - Reto Gassmann
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
| | - Helder Maiato
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 400-135 Porto, Portugal
- Cell Division Group, Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 420-319 Porto, Portugal
| |
Collapse
|
29
|
Spangler RK, Braun K, Ashley GE, van der Does M, Wruck D, Coronado AR, Matthew Ragle J, Iesmantavicius V, Morales Moya LJ, Jonnalagadda K, Partch CL, Großhans H, Ward JD. A conserved chronobiological complex times C. elegans development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593322. [PMID: 38766223 PMCID: PMC11100808 DOI: 10.1101/2024.05.09.593322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The mammalian PAS-domain protein PERIOD (PER) and its C. elegans orthologue LIN-42 have been proposed to constitute an evolutionary link between two distinct, circadian and developmental, timing systems. However, while the function of PER in animal circadian rhythms is well understood molecularly and mechanistically, this is not true for LIN-42's function in timing rhythmic development. Here, using targeted deletions, we find that the LIN-42 PAS domains are dispensable for the protein's function in timing molts. Instead, we observe arrhythmic molts upon deletion of a distinct sequence element, conserved with PER. We show that this element, designated CK1δ-binding domain (CK1BD), mediates stable binding to KIN-20, the C. elegans CK1δ/ε orthologue. We demonstrate that CK1δ phosphorylates LIN-42 and define two conserved helical motifs in the CK1BD, CK1BD-A and CK1BD-B, that have distinct roles in controlling CK1δ-binding and kinase activity in vitro. KIN-20 and the LIN-42 CK1BD are required for proper molting timing in vivo, and loss of LIN-42 binding changes KIN-20 subcellular localization. The interactions mirror the central role of a stable circadian PER-CK1 complex in setting a robust ~24-hour period. Hence, our results establish LIN-42/PER - KIN-20/CK1δ/ε as a functionally conserved signaling module of two distinct chronobiological systems.
Collapse
Affiliation(s)
- Rebecca K Spangler
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kathrin Braun
- Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
| | - Guinevere E Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Marit van der Does
- Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Daniel Wruck
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andrea Ramos Coronado
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | | | | | - Keya Jonnalagadda
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
- Center for Circadian Biology, University of California-San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California-Santa Cruz, Santa Cruz 95064, USA
| | - Helge Großhans
- Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
30
|
Dinneen E, Silva-García CG. Universal Single Copy Knock-In System in Caenorhabditis elegans : One Plasmid to Target All Chromosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627295. [PMID: 39713286 PMCID: PMC11661065 DOI: 10.1101/2024.12.06.627295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Successful transgenesis in model organisms has dramatically helped us understand gene function, regulation, genetic networks, and potential applications. Here, we introduce the universal single-copy knock-in system (Universal SKI System or U-SKI), designed for inserting any transgene by CRISPR/Cas9 in the Caenorhabditis elegans genome. The Universal SKI System takes advantage of a plasmid (pSKI), which can also be used for extrachromosomal arrays, to facilitate the insertion of a transgene at specific safe harbor loci on each autosomal chromosome. The pSKI plasmid contains multiple restriction sites for easy cloning and serves as a CRISPR/Cas9-based insertion repair template because it has two synthetic and long homology arms that recombine with the SKI cassettes. This system also uses a single crRNA guide, which acts as a Co-CRISPR enrichment marker. Overall, the Universal SKI System is highly flexible; with the same Universal SKI cassette on each autosome, researchers can select the insertion site and streamline tracking while reducing the complexity of expressing single-copy transgenes in C. elegans .
Collapse
|
31
|
Popiel EM, Ahluwalia R, Schuetz S, Yu B, Derry WB. MRCK-1 activates non-muscle myosin for outgrowth of a unicellular tube in Caenorhabditis elegans. Development 2024; 151:dev202772. [PMID: 39494605 PMCID: PMC11634028 DOI: 10.1242/dev.202772] [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: 02/06/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
The formation and patterning of unicellular biological tubes is essential for metazoan development. It is well established that vascular tubes and neurons use similar guidance cues to direct their development, but the downstream mechanisms that promote the outgrowth of biological tubes are not well characterized. We show that the conserved kinase MRCK-1 and its substrate the regulatory light chain of non-muscle myosin, MLC-4, are required for outgrowth of the unicellular excretory canal in C. elegans. Ablation of MRCK-1 or MLC-4 in the canal causes severe truncations with unlumenized projections of the basal membrane. Structure-function analysis of MRCK-1 indicates that the kinase domain, but not the small GTPase-binding CRIB domain, is required for canal outgrowth. Expression of a phosphomimetic form of MLC-4 rescues canal truncations in mrck-1 mutants and shows enrichment at the growing canal tip. Moreover, our work reveals a previously unreported function for non-muscle myosin downstream of MRCK-1 in excretory canal outgrowth that may be conserved in the development of seamless tubes in other organisms.
Collapse
Affiliation(s)
- Evelyn M. Popiel
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1X5, Canada
| | - Rhea Ahluwalia
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Ontario Institute for Cancer Research, 661 University Avenue, Toronto, ON M5G 0A3, Canada
| | - Stefan Schuetz
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1X5, Canada
| | - Bin Yu
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - W. Brent Derry
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1X5, Canada
| |
Collapse
|
32
|
Chen Y, Jiang Y, Sarvanantharajah N, Apirakkan O, Yang M, Milcova A, Topinka J, Abbate V, Arlt VM, Stürzenbaum SR. Genome-modified Caenorhabditis elegans expressing the human cytochrome P450 (CYP1A1 and CYP1A2) pathway: An experimental model for environmental carcinogenesis and pharmacological research. ENVIRONMENT INTERNATIONAL 2024; 194:109187. [PMID: 39671827 DOI: 10.1016/j.envint.2024.109187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), including the Group 1 human carcinogen benzo[a]pyrene (BaP), are produced by the incomplete combustion of organic matter and thus are present in tobacco smoke, charbroiled food and diesel exhaust. The nematode Caenorhabditis elegans is an established model organism, however it lacks the genetic components of the classical mammalian cytochrome P450 (CYP)-mediated BaP-diol-epoxide metabolism pathway. We therefore introduced human CYP1A1 or CYP1A2 together with human epoxide hydrolase (EPHX) into the worm genome by Mos1-mediated Single Copy Insertion (MosSCI) and evaluated their response to BaP exposure via toxicological endpoints. Compared to wild-type control, CYP-humanised worms were characterised by an increase in pharyngeal pumping rate and a decrease in volumetric surface area. Furthermore, BaP exposure reduced reproductive performance, as reflected in smaller brood size, which coincided with the downregulation of the nematode-specific major sperm protein as determined by transcriptomics (RNAseq). BaP-mediated reproductive toxicity was exacerbated in CYP-humanised worms at higher exposure levels. Collagen-related genes were downregulated in BaP-exposed animals, which correlate with the reduction in volumetric size. Whole genome DNA sequencing revealed a higher frequency of T > G (A > C) base substitution mutations in worms expressing human CYP1A1;EPHX which aligned with an increase in DNA adducts identified via an ELISA method (but not classical 32P-postlabelling). Overall, the CYP-humanised worms provided new insights into the value of genome-optimised invertebrate models by identifying the benefits and limitations within the context of the (3Rs) concept which aims to replace, reduce and refine the use of animals in research.
Collapse
Affiliation(s)
- Yuzhi Chen
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Yang Jiang
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Utrecht, Netherlands
| | - Nirujah Sarvanantharajah
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Orapan Apirakkan
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mengqi Yang
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Alena Milcova
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Jan Topinka
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK; Toxicology Department, GAB Consulting GmbH, 69126 Heidelberg, Germany
| | - Stephen R Stürzenbaum
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
33
|
Kiontke K, Fernandez P, Woronik A, Fitch DHA. Morphologically defined substages of tail morphogenesis in C. elegans males. Dev Dyn 2024; 253:1147-1164. [PMID: 38924277 PMCID: PMC11611696 DOI: 10.1002/dvdy.721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/01/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Sex-specific morphogenesis occurs in Caenorhabditis elegans in the vulva of the hermaphrodite and in the male tail during the last larval stage. Temporal progression of vulva morphogenesis has been described in fine detail. However, a similar precise description of male tail morphogenesis was lacking. RESULTS We here describe morphogenesis of the male tail at time points matching vulva development with special focus on morphogenesis of the tail tip. Using fluorescent reporters, we follow changes in cell shapes, cell fusions, nuclear migration, modifications in the basement membrane, and formation of a new apical extracellular matrix at the end of the tail. CONCLUSION Our analysis answers two open questions about tail tip morphogenesis (TTM) by showing that one of the four tail tip cells, hyp11, remains largely separate, while the other cells fully fuse with each other and with two additional tail cells to form a ventral tail syncytium. This merger of cells begins at the apical surface early during TTM but is only completed toward the end of the process. This work provides a framework for future investigations of cell biological factors that drive male tail morphogenesis.
Collapse
Affiliation(s)
- Karin Kiontke
- Department of Biology, New York University, New York, New York, USA
| | | | | | - David H A Fitch
- Department of Biology, New York University, New York, New York, USA
| |
Collapse
|
34
|
Turmel-Couture S, Martel PO, Beaulieu L, Lechasseur X, Fotso Dzuna LV, Narbonne P. Bidirectional transfer of a small membrane-impermeable molecule between the Caenorhabditis elegans intestine and germline. J Biol Chem 2024; 300:107963. [PMID: 39510179 DOI: 10.1016/j.jbc.2024.107963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
The extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) is a positive regulator of cell proliferation often upregulated in cancer. Its Caenorhabditis elegans ortholog MPK-1 stimulates germline stem cell (GSC) proliferation nonautonomously from the intestine or somatic gonad. How MPK-1 can perform this task from either of these two tissues however remains unclear. We reasoned that somatic MPK-1 activity could lead to the generation of proproliferative small molecules that could transfer from the intestine and/or somatic gonad to the germline. Here, in support of this hypothesis, we demonstrate that a significant fraction of the small membrane-impermeable fluorescent molecule, 5-carboxyfluorescein, transfers to the germline after its microinjection in the animal's intestine. The larger part of this transfer targets oocytes and requires the germline receptor mediated endocytosis 2 (RME-2) yolk receptor. A minor quantity of the dye is however distributed independently from RME-2 and more widely in the animal, including the distal germline, gonadal sheath, coelomocytes, and hypodermis. We further show that the intestine-to-germline transfer efficiency of this RME-2 independent fraction does not vary together with GSC proliferation rates or MPK-1 activity. Therefore, if germline proliferation was influenced by small membrane-impermeable molecules generated in the intestine, it is unlikely that proliferation would be regulated at the level of molecule transfer rate. Finally, we show that conversely, a similar fraction of germline injected 5-carboxyfluorescein transfers to the intestine, demonstrating transfer bidirectionality. Altogether, our results establish the possibility of an intestine-to-germline signaling axis mediated by small membrane-impermeable molecules that could promote GSC proliferation cell nonautonomously downstream of MPK-1 activity.
Collapse
Affiliation(s)
- Sarah Turmel-Couture
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Pier-Olivier Martel
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Lucie Beaulieu
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Xavier Lechasseur
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | | | - Patrick Narbonne
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada.
| |
Collapse
|
35
|
Bellutti L, Macaisne N, El Mossadeq L, Ganeswaran T, Canman JC, Dumont J. Regulation of outer kinetochore assembly during meiosis I and II by CENP-A and KNL-2/M18BP1 in C. elegans oocytes. Curr Biol 2024; 34:4853-4868.e6. [PMID: 39353426 PMCID: PMC11537844 DOI: 10.1016/j.cub.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/24/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024]
Abstract
During cell division, chromosomes build kinetochores that attach to spindle microtubules. Kinetochores usually form at the centromeres, which contain CENP-A nucleosomes. The outer kinetochore, which is the core attachment site for microtubules, is composed of the KMN network (Knl1c, Mis12c, and Ndc80c complexes) and is recruited downstream of CENP-A and its partner CENP-C. In C. elegans oocytes, kinetochores have been suggested to form independently of CENP-A nucleosomes. Yet kinetochore formation requires CENP-C, which acts in parallel to the nucleoporin MEL-28ELYS. Here, we used a combination of RNAi and Degron-based depletion of CENP-A (or downstream CENP-C) to demonstrate that both proteins are in fact responsible for a portion of outer kinetochore assembly during meiosis I and are essential for accurate chromosome segregation. The remaining part requires the coordinated action of KNL-2 (ortholog of human M18BP1) and of the nucleoporin MEL-28ELYS. Accordingly, co-depletion of CENP-A (or CENP-C) and KNL-2M18BP1 (or MEL-28ELYS) prevented outer kinetochore assembly in oocytes during meiosis I. We further found that KNL-2M18BP1 and MEL-28ELYS are interdependent for kinetochore localization. Using engineered mutants, we demonstrated that KNL-2M18BP1 recruits MEL-28ELYS at meiotic kinetochores through a specific N-terminal domain, independently of its canonical CENP-A loading factor activity. Finally, we found that meiosis II outer kinetochore assembly was solely dependent on the canonical CENP-A/CENP-C pathway. Thus, like in most cells, outer kinetochore assembly in C. elegans oocytes depends on centromeric chromatin. However, during meiosis I, an additional KNL-2M18BP1 and MEL-28ELYS pathway acts in a non-redundant manner and in parallel to canonical centromeric chromatin.
Collapse
Affiliation(s)
- Laura Bellutti
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Nicolas Macaisne
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Layla El Mossadeq
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | | | - Julie C Canman
- Columbia University, Irving Medical Center, Department of Pathology and Cell Biology, New York, NY 10032, USA
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France.
| |
Collapse
|
36
|
Lara-Gonzalez P, Variyar S, Moghareh S, Nguyen ACN, Kizhedathu A, Budrewicz J, Schlientz A, Varshney N, Bellaart A, Oegema K, Bardwell L, Desai A. Cyclin B3 is a dominant fast-acting cyclin that drives rapid early embryonic mitoses. J Cell Biol 2024; 223:e202308034. [PMID: 39105756 PMCID: PMC11303871 DOI: 10.1083/jcb.202308034] [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: 08/03/2023] [Revised: 04/27/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024] Open
Abstract
Mitosis in early embryos often proceeds at a rapid pace, but how this pace is achieved is not understood. Here, we show that cyclin B3 is the dominant driver of rapid embryonic mitoses in the C. elegans embryo. Cyclins B1 and B2 support slow mitosis (NEBD to anaphase ∼600 s), but the presence of cyclin B3 dominantly drives the approximately threefold faster mitosis observed in wildtype. Multiple mitotic events are slowed down in cyclin B1 and B2-driven mitosis, and cyclin B3-associated Cdk1 H1 kinase activity is ∼25-fold more active than cyclin B1-associated Cdk1. Addition of cyclin B1 to fast cyclin B3-only mitosis introduces an ∼60-s delay between completion of chromosome alignment and anaphase onset; this delay, which is important for segregation fidelity, is dependent on inhibitory phosphorylation of the anaphase activator Cdc20. Thus, cyclin B3 dominance, coupled to a cyclin B1-dependent delay that acts via Cdc20 phosphorylation, sets the rapid pace and ensures mitotic fidelity in the early C. elegans embryo.
Collapse
Affiliation(s)
- Pablo Lara-Gonzalez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Smriti Variyar
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Shabnam Moghareh
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Anh Cao Ngoc Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Amrutha Kizhedathu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | | | - Aleesa Schlientz
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Neha Varshney
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Andrew Bellaart
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Karen Oegema
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Lee Bardwell
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Arshad Desai
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
37
|
Blow F, Jeffrey K, Chow FWN, Nikonorova IA, Barr MM, Cook AG, Prevo B, Cheerambathur DK, Buck AH. SID-2 is a conserved extracellular vesicle protein that is not associated with environmental RNAi in parasitic nematodes. Open Biol 2024; 14:240190. [PMID: 39501794 PMCID: PMC11538922 DOI: 10.1098/rsob.240190] [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: 07/07/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024] Open
Abstract
In the free-living nematode Caenorhabditis elegans, the transmembrane protein SID-2 imports double-stranded RNA into intestinal cells to trigger systemic RNA interference (RNAi), allowing organisms to sense and respond to environmental cues such as the presence of pathogens. This process, known as environmental RNAi, has not been observed in the most closely related parasites that are also within clade V. Previous sequence-based searches failed to identify sid-2 orthologues in available clade V parasite genomes. In this study, we identified sid-2 orthologues in these parasites using genome synteny and protein structure-based comparison, following identification of a SID-2 orthologue in extracellular vesicles from the murine intestinal parasitic nematode Heligmosomoides bakeri. Expression of GFP-tagged H. bakeri SID-2 in C. elegans showed similar localization to the intestinal apical membrane as seen for GFP-tagged C. elegans SID-2, and further showed mobility in intestinal cells in vesicle-like structures. We tested the capacity of H. bakeri SID-2 to functionally complement environmental RNAi in a C. elegans SID-2 null mutant and show that H. bakeri SID-2 does not rescue the phenotype in this context. Our work identifies SID-2 as a highly abundant EV protein whose ancestral function may be unrelated to environmental RNAi, and rather highlights an association with extracellular vesicles in nematodes.
Collapse
Affiliation(s)
- Frances Blow
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK
| | - Kate Jeffrey
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK
- Wellcome Centre for Cell Biology & Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Franklin Wang-Ngai Chow
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong
| | - Inna A Nikonorova
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, New Jersey NJ 08854, USA
| | - Maureen M Barr
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, New Jersey NJ 08854, USA
| | - Atlanta G Cook
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK
| | - Bram Prevo
- Wellcome Centre for Cell Biology & Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Dhanya K Cheerambathur
- Wellcome Centre for Cell Biology & Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Amy H Buck
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK
| |
Collapse
|
38
|
Al-Refaie N, Padovani F, Hornung J, Pudelko L, Binando F, Del Carmen Fabregat A, Zhao Q, Towbin BD, Cenik ES, Stroustrup N, Padeken J, Schmoller KM, Cabianca DS. Fasting shapes chromatin architecture through an mTOR/RNA Pol I axis. Nat Cell Biol 2024; 26:1903-1917. [PMID: 39300311 PMCID: PMC11567895 DOI: 10.1038/s41556-024-01512-w] [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: 08/16/2023] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Chromatin architecture is a fundamental mediator of genome function. Fasting is a major environmental cue across the animal kingdom, yet how it impacts three-dimensional (3D) genome organization is unknown. Here we show that fasting induces an intestine-specific, reversible and large-scale spatial reorganization of chromatin in Caenorhabditis elegans. This fasting-induced 3D genome reorganization requires inhibition of the nutrient-sensing mTOR pathway, acting through the regulation of RNA Pol I, but not Pol II nor Pol III, and is accompanied by remodelling of the nucleolus. By uncoupling the 3D genome configuration from the animal's nutritional status, we find that the expression of metabolic and stress-related genes increases when the spatial reorganization of chromatin occurs, showing that the 3D genome might support the transcriptional response in fasted animals. Our work documents a large-scale chromatin reorganization triggered by fasting and reveals that mTOR and RNA Pol I shape genome architecture in response to nutrients.
Collapse
Affiliation(s)
- Nada Al-Refaie
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians Universität München, Munich, Germany
| | - Francesco Padovani
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Johanna Hornung
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lorenz Pudelko
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians Universität München, Munich, Germany
| | - Francesca Binando
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Andrea Del Carmen Fabregat
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Qiuxia Zhao
- Department of Molecular Biosciences, University of Texas Austin, Austin, TX, USA
| | | | - Elif Sarinay Cenik
- Department of Molecular Biosciences, University of Texas Austin, Austin, TX, USA
| | - Nicholas Stroustrup
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jan Padeken
- Institute of Molecular Biology, Mainz, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Daphne S Cabianca
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
39
|
Romero-Bueno R, Fragoso-Luna A, Ayuso C, Mellmann N, Kavsek A, Riedel CG, Ward JD, Askjaer P. A human progeria-associated BAF-1 mutation modulates gene expression and accelerates aging in C. elegans. EMBO J 2024; 43:5718-5746. [PMID: 39367234 PMCID: PMC11574047 DOI: 10.1038/s44318-024-00261-8] [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: 07/11/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024] Open
Abstract
Alterations in the nuclear envelope are linked to a variety of rare diseases termed laminopathies. A single amino acid substitution at position 12 (A12T) of the human nuclear envelope protein BAF (Barrier to Autointegration Factor) causes Néstor-Guillermo Progeria Syndrome (NGPS). This premature ageing condition leads to growth retardation and severe skeletal defects, but the underlying mechanisms are unknown. Here, we have generated a novel in vivo model for NGPS by modifying the baf-1 locus in C. elegans to mimic the human NGPS mutation. These baf-1(G12T) mutant worms displayed multiple phenotypes related to fertility, lifespan, and stress resistance. Importantly, nuclear morphology deteriorated faster during aging in baf-1(G12T) compared to wild-type animals, recapitulating an important hallmark of cells from progeria patients. Although localization of BAF-1(G12T) was similar to wild-type BAF-1, lamin accumulation at the nuclear envelope was reduced in mutant worms. Tissue-specific chromatin binding and transcriptome analyses showed reduced BAF-1 association in most genes deregulated by the baf-1(G12T) mutation, suggesting that altered BAF chromatin association induces NGPS phenotypes via altered gene expression.
Collapse
Affiliation(s)
- Raquel Romero-Bueno
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Carretera de Utrera, km 1, 41013, Sevilla, Spain
| | - Adrián Fragoso-Luna
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Carretera de Utrera, km 1, 41013, Sevilla, Spain
| | - Cristina Ayuso
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Carretera de Utrera, km 1, 41013, Sevilla, Spain
| | - Nina Mellmann
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Carretera de Utrera, km 1, 41013, Sevilla, Spain
| | - Alan Kavsek
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Christian G Riedel
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Peter Askjaer
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Carretera de Utrera, km 1, 41013, Sevilla, Spain.
| |
Collapse
|
40
|
Muhammad T, Edwards SL, Morphis AC, Johnson MV, Oliveira VD, Chamera T, Liu S, Nguyen NGT, Li J. Non-cell-autonomous regulation of germline proteostasis by insulin/IGF-1 signaling-induced dietary peptide uptake via PEPT-1. EMBO J 2024; 43:4892-4921. [PMID: 39284915 PMCID: PMC11535032 DOI: 10.1038/s44318-024-00234-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: 04/01/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 11/06/2024] Open
Abstract
Gametogenesis involves active protein synthesis and is proposed to rely on proteostasis. Our previous work in C. elegans indicates that germline development requires coordinated activities of insulin/IGF-1 signaling (IIS) and HSF-1, the central regulator of the heat shock response. However, the downstream mechanisms were not identified. Here, we show that depletion of HSF-1 from germ cells impairs chaperone gene expression, causing protein degradation and aggregation and, consequently, reduced fecundity and gamete quality. Conversely, reduced IIS confers germ cell resilience to HSF-1 depletion-induced protein folding defects and various proteotoxic stresses. Surprisingly, this effect was not mediated by an enhanced stress response, which underlies longevity in low IIS conditions, but by reduced ribosome biogenesis and translation rate. We found that IIS activates the expression of intestinal peptide transporter PEPT-1 by alleviating its repression by FOXO/DAF-16, allowing dietary proteins to be efficiently incorporated into an amino acid pool that fuels germline protein synthesis. Our data suggest this non-cell-autonomous pathway is critical for proteostasis regulation during gametogenesis.
Collapse
Affiliation(s)
- Tahir Muhammad
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Stacey L Edwards
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Allison C Morphis
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Mary V Johnson
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Vitor De Oliveira
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Tomasz Chamera
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Siyan Liu
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | | | - Jian Li
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
41
|
Schlientz AJ, Lee KY, Sebastián Gómez-Cavazos J, Lara-González P, Desai A, Oegema K. The CYK-4 GAP domain regulates cortical targeting of centralspindlin to promote contractile ring assembly and facilitate ring dissolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620943. [PMID: 39554051 PMCID: PMC11565784 DOI: 10.1101/2024.10.29.620943] [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: 11/19/2024]
Abstract
During cytokinesis, an equatorial contractile ring partitions the cell contents. Contractile ring assembly requires an equatorial zone of active GTP-bound RhoA generated by the guanine nucleotide exchange factor ECT21,2. ECT2 is activated by centralspindlin, a complex composed of two molecules each of kinesin-6 and CYK4. During anaphase, Centralspindlin is activated at the central spindle between the separating chromosomes and diffuses to the plasma membrane, where it engages with ECT2 via its N-terminal half3,4. The C-terminal half of CYK4 contains a lipid-binding C1 domain that contributes to plasma membrane targeting5 and a GTPase-activating protein (GAP) domain that has an interaction surface for a Rho family GTPase, whose functions have remained unclear 1,3,4,6,7. Here, using the one-cell stage C. elegans embryo as a model, we show that RhoA and the Rho-binding interface of the CYK4 GAP domain drive the recruitment of centralspindlin to the equatorial cortex. By contrast, a point mutant that selectively disrupts GAP activity does not prevent cortical centralspindlin recruitment but instead substantially delays dissipation of centralspindlin from the cortex. These findings suggest that positive feedback, in which centralspindlin recruitment promotes the generation of active RhoA and active RhoA drives centralspindlin recruitment, is central to the rapid assembly of the contractile ring within a narrow time window. They also indicate that the CYK4 GAP catalytic activity contributes to release of centralspindlin from the cortex, potentially to ensure timely dissolution of the contractile ring.
Collapse
Affiliation(s)
- Aleesa J Schlientz
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, California 92093, USA
| | - Kian-Yong Lee
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - J. Sebastián Gómez-Cavazos
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, California 92093, USA
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Pablo Lara-González
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Arshad Desai
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, California 92093, USA
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Karen Oegema
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, California 92093, USA
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
| |
Collapse
|
42
|
Carrick BH, Crittenden SL, Linsley M, Dos Santos SJC, Wickens M, Kimble J. The PUF RNA-binding protein, FBF-2, maintains stem cells without binding to RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620246. [PMID: 39484565 PMCID: PMC11527184 DOI: 10.1101/2024.10.25.620246] [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: 11/03/2024]
Abstract
Like all canonical PUF proteins, C. elegans FBF-2 binds to specific RNAs via tripartite recognition motifs (TRMs). Here we report that an FBF-2 mutant protein that cannot bind to RNA, is nonetheless biologically active and maintains stem cells. This unexpected result challenges the conventional wisdom that RBPs must bind to RNAs to achieve biological activity. Also unexpectedly, FBF-2 interactions with partner proteins can compensate for loss of RNA-binding. FBF-2 only loses biological activity when its RNA-binding and partner interactions are both defective. These findings highlight the complementary contributions of RNA-binding and protein partner interactions to activity of an RNA-binding protein.
Collapse
Affiliation(s)
- Brian H. Carrick
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
- Present address: MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | | | - MaryGrace Linsley
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
- Present address: Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA
| | - Stephany J. Costa Dos Santos
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
- Present address: WiCell Research Institute, Inc., Madison WI
| | - Marvin Wickens
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
43
|
Wong C, Jurczak EM, Roy R. Neuronal exosomes transport an miRISC cargo to preserve stem cell integrity during energy stress. Cell Rep 2024; 43:114851. [PMID: 39392750 DOI: 10.1016/j.celrep.2024.114851] [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: 01/04/2024] [Revised: 08/19/2024] [Accepted: 09/24/2024] [Indexed: 10/13/2024] Open
Abstract
During periods of nutrient scarcity, many animals undergo germline quiescence to preserve reproductive capacity, and neurons are often necessary for this adaptation. We show here that starvation causes the release of neuronal microRNA (miRNA)/Argonaute-loaded exosomes following AMP kinase-regulated trafficking changes within serotonergic neurons. This neuron-to-germline communication is independent of classical neurotransmission but instead relies on endosome-derived vesicles that carry a pro-quiescent small RNA cargo to modify germline gene expression. Using an miRNA activity sensor, we show that neuronally expressed miRNAs can extinguish the expression of germline mRNA targets in an exosome-dependent manner. Our findings demonstrate how an adaptive neuronal response can change gene expression at a distance by redirecting intracellular trafficking to release neuronal exosomes with specific miRNA cargoes capable of tracking to their appropriate destinations.
Collapse
Affiliation(s)
- Christopher Wong
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Elena M Jurczak
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Richard Roy
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada.
| |
Collapse
|
44
|
Bastien BL, Haury WR, Smisko WR, Hart MP. nlr-1/CNTNAP regulates dopamine circuit structure and foraging behaviors in C. elegans. Commun Biol 2024; 7:1248. [PMID: 39358459 PMCID: PMC11447218 DOI: 10.1038/s42003-024-06936-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The neurexin superfamily, consisting of neurexins and Casprs, play important roles in the development, maintenance, function, and plasticity of neuronal circuits. Caspr/CNTNAP genes are linked to alterations in neuronal circuits and associated with neurodevelopmental and neurodegenerative conditions. Casprs are implicated in multiple neuronal signaling pathways, including dopamine; however, the molecular mechanisms by which Casprs differentially alter specific signaling pathways and downstream behaviors are unclear. We find that the C. elegans Caspr nlr-1 functions in neurons to control foraging behaviors, acting in distinct monoamine neurons to modulate locomotor activity in the presence or absence of food. nlr-1 functions in dopamine neurons to reduce activity in the absence of food, similar to the role of dopamine, and regulates dopamine signaling through D2-like receptors. Furthermore, nlr-1 contributes to proper morphology and presynaptic structure of dopamine neurons, dopamine receptor expression and localization, and the behavioral response to dopamine. We find that nlr-1 similarly regulates another dopamine-dependent behavior, the basal slowing response. Therefore, spatial manipulation of a broadly expressed neuronal gene is sufficient to alter neural circuits and behavior and uncovers important functions masked by global manipulation, highlighting the importance of genetic variation and mechanisms that impact spatial expression of genes to behavior.
Collapse
Affiliation(s)
- Brandon L Bastien
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William R Haury
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William R Smisko
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael P Hart
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
45
|
Ponomarova O, Starbard AN, Belfi A, Anderson AV, Sundaram MV, Walhout AJ. idh-1 neomorphic mutation confers sensitivity to vitamin B12 in Caenorhabditis elegans. Life Sci Alliance 2024; 7:e202402924. [PMID: 39009411 PMCID: PMC11249921 DOI: 10.26508/lsa.202402924] [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: 07/02/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024] Open
Abstract
In humans, a neomorphic isocitrate dehydrogenase mutation (idh-1neo) causes increased levels of cellular D-2-hydroxyglutarate (D-2HG), a proposed oncometabolite. However, the physiological effects of increased D-2HG and whether additional metabolic changes occur in the presence of an idh-1neo mutation are not well understood. We created a Caenorhabditis elegans model to study the effects of the idh-1neo mutation in a whole animal. Comparing the phenotypes exhibited by the idh-1neo to ∆dhgd-1 (D-2HG dehydrogenase) mutant animals, which also accumulate D-2HG, we identified a specific vitamin B12 diet-dependent vulnerability in idh-1neo mutant animals that leads to increased embryonic lethality. Through a genetic screen, we found that impairment of the glycine cleavage system, which generates one-carbon donor units, exacerbates this phenotype. In addition, supplementation with alternate sources of one-carbon donors suppresses the lethal phenotype. Our results indicate that the idh-1neo mutation imposes a heightened dependency on the one-carbon pool and provides a further understanding of how this oncogenic mutation rewires cellular metabolism.
Collapse
Affiliation(s)
- Olga Ponomarova
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Alyxandra N Starbard
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexandra Belfi
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda V Anderson
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Albertha Jm Walhout
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
46
|
Cully D, Cohen NR, Breen PC, Newman MA, Dowen RH. A novel gain-of-function mutation in sgk-1 partially suppresses mTORC2 defects. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001163. [PMID: 39410967 PMCID: PMC11474317 DOI: 10.17912/micropub.biology.001163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
The serine/threonine protein kinase SGK-1 is a downstream target of mTOR complex 2 (mTORC2) and is a conserved regulator of growth and metabolism. In C. elegans , mutations in rict-1 , which encodes an essential component of mTORC2, impairs lipid homeostasis and growth; however, these defects are partially suppressed by an activating mutation in SGK-1 , E116K. Here, we describe a stronger gain-of-function mutation in sgk-1 , L112F, that was identified in a forward genetic screen for rict-1 suppressor mutations . This allele will be useful in further dissecting the mTORC2 pathway and provides new insight into the role of this conserved residue in regulating SGK-1 kinase activity.
Collapse
Affiliation(s)
- David Cully
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Natalie R. Cohen
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Peter C. Breen
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Martin A. Newman
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Robert H. Dowen
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
47
|
Laranjeira AC, Berger S, Kohlbrenner T, Greter NR, Hajnal A. Nutritional vitamin B12 regulates RAS/MAPK-mediated cell fate decisions through one-carbon metabolism. Nat Commun 2024; 15:8178. [PMID: 39289374 PMCID: PMC11408588 DOI: 10.1038/s41467-024-52556-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
Vitamin B12 is an essential nutritional co-factor for the folate and methionine cycles, which together constitute one-carbon metabolism. Here, we show that dietary uptake of vitamin B12 modulates cell fate decisions controlled by the conserved RAS/MAPK signaling pathway in C. elegans. A bacterial diet rich in vitamin B12 increases vulval induction, germ cell apoptosis and oocyte differentiation. These effects are mediated by different one-carbon metabolites in a tissue-specific manner. Vitamin B12 enhances via the choline/phosphatidylcholine metabolism vulval induction by down-regulating fat biosynthesis genes and increasing H3K4 tri-methylation, which results in increased expression of RAS/MAPK target genes. Furthermore, the nucleoside metabolism and H3K4 tri-methylation positively regulate germ cell apoptosis and oocyte production. Using mammalian cells carrying different activated KRAS and BRAF alleles, we show that the effects of methionine on RAS/MAPK-regulated phenotype are conserved in mammals. Our findings suggest that the vitamin B12-dependent one-carbon metabolism is a limiting factor for diverse RAS/MAPK-induced cellular responses.
Collapse
Affiliation(s)
| | - Simon Berger
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Tea Kohlbrenner
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Nadja R Greter
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Alex Hajnal
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
48
|
Tamagawa K, Dayi M, Sun S, Hata R, Kikuchi T, Haruta N, Sugimoto A, Makino T. Evolutionary changes of noncoding elements associated with transition of sexual mode in Caenorhabditis nematodes. SCIENCE ADVANCES 2024; 10:eadn9913. [PMID: 39270031 PMCID: PMC11397494 DOI: 10.1126/sciadv.adn9913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024]
Abstract
The transition of the sexual mode occurs widely in animal evolution. In Caenorhabditis nematodes, androdioecy, a sexual polymorphism composed of males and hermaphrodites having the ability to self-fertilize, has evolved independently multiple times. While the modification of noncoding regulatory elements likely contributed to the evolution of hermaphroditism, little is known about these changes. Here, we conducted a genome-wide analysis of conserved noncoding elements (CNEs) focusing on the evolution of hermaphroditism in Caenorhabditis nematodes. We found that, in androdioecious nematodes, mutations rapidly accumulated in CNEs' neighboring genes associated with sexual traits. Expression analysis indicate that the identified CNEs are involved in spermatogenesis in hermaphrodites and associated with the transition of gene expression from dioecious to androdioecious nematodes. Last, genome editing of a CNE neighboring laf-1 resulted in a change in its expression in the gonadal region undergoing spermatogenesis. Our bioinformatic and experimental analyses highlight the importance of CNEs in gene regulation associated with the development of hermaphrodites.
Collapse
Affiliation(s)
- Katsunori Tamagawa
- Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| | - Mehmet Dayi
- Forestry Vocational School, Duzce University, 81620 Duzce, Türkiye
| | - Simo Sun
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwanoha, Kashiwa City, Japan
| | - Rikako Hata
- Department of Biology, Faculty of Science, Tohoku University, Aoba-ku, Sendai, Japan
| | - Taisei Kikuchi
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwanoha, Kashiwa City, Japan
| | - Nami Haruta
- Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| | - Asako Sugimoto
- Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| | - Takashi Makino
- Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| |
Collapse
|
49
|
Parvand M, Liang JJH, Bozorgmehr T, Born D, Luna Cortes A, Rankin CH. A familial Alzheimer's disease associated mutation in presenilin-1 mediates amyloid-beta independent cell specific neurodegeneration. PLoS One 2024; 19:e0289435. [PMID: 39240956 PMCID: PMC11379242 DOI: 10.1371/journal.pone.0289435] [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: 07/17/2023] [Accepted: 08/23/2024] [Indexed: 09/08/2024] Open
Abstract
Mutations in the presenilin (PS) genes are a predominant cause of familial Alzheimer's disease (fAD). An ortholog of PS in the genetic model organism Caenorhabditis elegans (C. elegans) is sel-12. Mutations in the presenilin genes are commonly thought to lead to fAD by upregulating the expression of amyloid beta (Aβ), however this hypothesis has been challenged by recent evidence. As C. elegans lack amyloid beta (Aβ), the goal of this work was to examine Aβ-independent effects of mutations in sel-12 and PS1/PS2 on behaviour and sensory neuron morphology across the lifespan in a C. elegans model. Olfactory chemotaxis experiments were conducted on sel-12(ok2078) loss-of-function mutant worms. Adult sel-12 mutant worms showed significantly lower levels of chemotaxis to odorants compared to wild-type worms throughout their lifespan, and this deficit increased with age. The chemotaxis phenotype in sel-12 mutant worms is rescued by transgenic over-expression of human wild-type PS1, but not the classic fAD-associated variant PS1C410Y, when expression was driven by either the endogenous sel-12 promoter (Psel-12), a pan-neuronal promoter (Primb-1), or by a promoter whose primary expression was in the sensory neurons responsible for the chemotaxis behavior (Psra-6, Podr-10). The behavioural phenotype was also rescued by over-expressing an atypical fAD-linked mutation in PS1 (PS1ΔS169) that has been reported to leave the Notch pathway intact. An examination of the morphology of polymodal nociceptive (ASH) neurons responsible for the chemotaxis behavior also showed increased neurodegeneration over time in sel-12 mutant worms that could be rescued by the same transgenes that rescued the behaviour, demonstrating a parallel with the observed behavioral deficits. Thus, we report an Aβ-independent neurodegeneration in C. elegans that was rescued by cell specific over-expression of wild-type human presenilin.
Collapse
Affiliation(s)
- Mahraz Parvand
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joseph J H Liang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tahereh Bozorgmehr
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dawson Born
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alvaro Luna Cortes
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catharine H Rankin
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
50
|
Shen A, Hencel K, Parker M, Scott R, Skukan R, Adesina A, Metheringham C, Miska E, Nam Y, Haerty W, Simpson G, Akay A. U6 snRNA m6A modification is required for accurate and efficient splicing of C. elegans and human pre-mRNAs. Nucleic Acids Res 2024; 52:9139-9160. [PMID: 38808663 PMCID: PMC11347140 DOI: 10.1093/nar/gkae447] [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: 09/15/2023] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 05/30/2024] Open
Abstract
pre-mRNA splicing is a critical feature of eukaryotic gene expression. Both cis- and trans-splicing rely on accurately recognising splice site sequences by spliceosomal U snRNAs and associated proteins. Spliceosomal snRNAs carry multiple RNA modifications with the potential to affect different stages of pre-mRNA splicing. Here, we show that the conserved U6 snRNA m6A methyltransferase METT-10 is required for accurate and efficient cis- and trans-splicing of C. elegans pre-mRNAs. The absence of METT-10 in C. elegans and METTL16 in humans primarily leads to alternative splicing at 5' splice sites with an adenosine at +4 position. In addition, METT-10 is required for splicing of weak 3' cis- and trans-splice sites. We identified a significant overlap between METT-10 and the conserved splicing factor SNRNP27K in regulating 5' splice sites with +4A. Finally, we show that editing endogenous 5' splice site +4A positions to +4U restores splicing to wild-type positions in a mett-10 mutant background, supporting a direct role for U6 snRNA m6A modification in 5' splice site recognition. We conclude that the U6 snRNA m6A modification is important for accurate and efficient pre-mRNA splicing.
Collapse
Affiliation(s)
- Aykut Shen
- School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| | - Katarzyna Hencel
- School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| | - Matthew T Parker
- School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Robyn Scott
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Roberta Skukan
- School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| | | | | | - Eric A Miska
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge CB2 1QN, UK
| | - Yunsun Nam
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wilfried Haerty
- School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - Gordon G Simpson
- School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
- Cell & Molecular Sciences, James Hutton Institute, Invergowrie, DD2 5DA, UK
| | - Alper Akay
- School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| |
Collapse
|