1
|
Perfilova KV, Matyuta IO, Minyaev ME, Boyko KM, Cooley RB, Sluchanko NN. High-resolution structure reveals enhanced 14-3-3 binding by a mutant SARS-CoV-2 nucleoprotein variant with improved replicative fitness. Biochem Biophys Res Commun 2025; 767:151915. [PMID: 40318379 DOI: 10.1016/j.bbrc.2025.151915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Replication of many viruses depends on phosphorylation of viral proteins by host protein kinases and subsequent recruitment of host protein partners. The nucleoprotein (N) of SARS-CoV-2 is heavily phosphorylated and recruits human phosphopeptide-binding 14-3-3 proteins early in infection, which is reversed prior to nucleocapsid assembly in new virions. Among the multiple phosphosites of N, which are particularly dense in the serine/arginine-rich interdomain region, phospho-Thr205 is highly relevant for 14-3-3 recruitment by SARS-CoV-2 N. The context of this site is mutated in most SARS-CoV-2 variants of concern. Among mutations that increase infectious virus titers, the S202R mutation (B.1.526 Iota) causes a striking replication boost (∼166-fold), although its molecular consequences have remained unclear. Here, we show that the S202R-mutated N phosphopeptide exhibits a 5-fold higher affinity for human 14-3-3ζ than the Wuhan variant and we rationalize this effect by solving a high-resolution crystal structure of the complex. The structure revealed an enhanced 14-3-3/N interface contributed by the Arg202 side chain that, in contrast to Ser202, formed multiple stabilizing contacts with 14-3-3, including water-mediated H-bonds and guanidinium pi-pi stacking. These findings provide a compelling link between the replicative fitness of SARS-CoV-2 and the N protein's affinity for host 14-3-3 proteins.
Collapse
Affiliation(s)
- Kristina V Perfilova
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, 119071, Russia
| | - Ilya O Matyuta
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, 119071, Russia; Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Mikhail E Minyaev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Konstantin M Boyko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, 119071, Russia
| | - Richard B Cooley
- GCE4All Center, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, 97331, USA
| | - Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, 119071, Russia.
| |
Collapse
|
2
|
Fischer SN, Claussen ER, Kourtis S, Sdelci S, Orchard S, Hermjakob H, Kustatscher G, Drew K. hu.MAP3.0: atlas of human protein complexes by integration of >25,000 proteomic experiments. Mol Syst Biol 2025:10.1038/s44320-025-00121-5. [PMID: 40425816 DOI: 10.1038/s44320-025-00121-5] [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: 11/22/2024] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Macromolecular protein complexes carry out most cellular functions. Unfortunately, we lack the subunit composition for many human protein complexes. To address this gap we integrated >25,000 mass spectrometry experiments using a machine learning approach to identify >15,000 human protein complexes. We show our map of protein complexes is highly accurate and more comprehensive than previous maps, placing nearly 70% of human proteins into their physical contexts. We globally characterize our complexes using mass spectrometry based protein covariation data (ProteomeHD.2) and identify covarying complexes suggesting common functional associations. hu.MAP3.0 generates testable functional hypotheses for 472 uncharacterized proteins which we support using AlphaFold modeling. Additionally, we use AlphaFold modeling to identify 5871 mutually exclusive proteins in hu.MAP3.0 complexes suggesting complexes serve different functional roles depending on their subunit composition. We identify expression as the primary way cells and organisms relieve the conflict of mutually exclusive subunits. Finally, we import our complexes to EMBL-EBI's Complex Portal ( https://www.ebi.ac.uk/complexportal/home ) and provide complexes through our hu.MAP3.0 web interface ( https://humap3.proteincomplexes.org/ ). We expect our resource to be highly impactful to the broader research community.
Collapse
Affiliation(s)
- Samantha N Fischer
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Erin R Claussen
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Savvas Kourtis
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sara Sdelci
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Georg Kustatscher
- Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Kevin Drew
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
3
|
Chapagai D, Strebhardt K, Wyatt MD, McInnes C. Structural regulation of PLK1 activity: implications for cell cycle function and drug discovery. Cancer Gene Ther 2025:10.1038/s41417-025-00907-7. [PMID: 40379873 DOI: 10.1038/s41417-025-00907-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/19/2025]
Abstract
Polo Like Kinase 1 (PLK1), a key regulator of mitosis whose overexpression is often associated with poor survival rates in cancer, continues to be widely investigated as an oncology drug target with clinical trials evaluating second and third generation inhibitors. In addition to the conserved N-terminal kinase domain (KD), a unique characteristic of the Polo-Like kinase family is the C-terminal polo-box domain (PBD). The PBD contains a phosphopeptide binding site that recognizes substrates primed by other kinases and furthermore is responsible for subcellular localization of PLK1 to specific sites in the nucleus including centrosomes and kinetochores. Another role of the PBD is its regulatory ability through domain-domain interactions with the KD to maintain an autoinhibited state of PLK1. Insights into post translational modifications and the PBD - KD domain-domain association have been obtained and show that key events in PLK1 regulation include phosphosubstrate binding, T210 phosphorylation and engagement with the Bora protein. These can induce an open and active conformation where the domain-domain inhibitory interactions no longer dominate. Further regulatory events recently described include the interchange between monomeric and dimeric forms, which can also serve to inhibit or activate PLK1 during the cell cycle. Different oligomeric forms of PLK1, existing as homodimers and heterodimers with PLK2, have been identified and likely play context dependent roles. This review provides an overview of recent information describing structural and mechanistic insights into inhibition of PLK1 and the temporal and spatial requirements of its activation and regulation. It also covers recent insights into the conformational regulation of other members of the Polo-Like kinase family. The implications of the conformational regulation of PLK1 with respect to cell cycle function and drug discovery are significant and are therefore discussed in detail.
Collapse
Affiliation(s)
- Danda Chapagai
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, 02129, USA
| | - Klaus Strebhardt
- Department of Gynecology, University Hospital, Goethe University, Theodor-Stern-Kai 7-9, Frankfurt am Main, 60596, Germany
| | - Michael D Wyatt
- Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, 29208, USA
| | - Campbell McInnes
- Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, 29208, USA.
| |
Collapse
|
4
|
Spooner HC, Dixon RE. 14-3-3 proteins: Regulators of cardiac excitation-contraction coupling and stress responses. J Physiol 2025. [PMID: 40349303 DOI: 10.1113/jp288566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
14-3-3 proteins are highly conserved proteins that regulate numerous cellular processes mostly through phosphorylation-dependent protein-protein interactions. In the heart 14-3-3 proteins play critical roles in cardiac conduction pathways, excitation-contraction (EC) coupling, development and stress responses. This review summarizes the current understanding of cardiac 14-3-3 regulation and function, with particular emphasis on its role in ion channel regulation and β-adrenergic signalling. We discuss how 14-3-3 proteins act through three main mechanisms - masking, clamping, and scaffolding - to regulate target proteins, including Cx43, CaV1.2, NaV1.5, and various potassium channels. The seven mammalian 14-3-3 isoforms display distinct but overlapping functions, with tissue-specific expression patterns and isoform-specific regulation through phosphorylation and dimerization. Recent work has revealed 14-3-3's importance in cardiac development and stress responses, where it generally serves a cardioprotective role. However in some pathological contexts such as ischaemia-reperfusion injury, 14-3-3 can be detrimental. We highlight emerging themes in cardiac 14-3-3 biology, including its role in prolonging β-adrenergic signalling. Understanding the complex regulation of cardiac 14-3-3 and its numerous targets presents both opportunities and challenges for therapeutic development.
Collapse
Affiliation(s)
- Heather C Spooner
- Department of Physiology and Membrane Biology, University of California Davis, School of Medicine, Davis, CA, USA
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, University of California Davis, School of Medicine, Davis, CA, USA
| |
Collapse
|
5
|
Náplavová A, Kozeleková A, Crha R, Gronenborn AM, Hritz J. Harnessing the power of 19F NMR for characterizing dimerization and ligand binding of 14-3-3 proteins. Int J Biol Macromol 2025; 305:141253. [PMID: 39978522 DOI: 10.1016/j.ijbiomac.2025.141253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
The main role of dimeric 14-3-3 proteins is to modulate the activity of several hundred binding partners by interacting with phosphorylated residues of the partner proteins, often located in disordered regions. The inherent flexibility or large size of 14-3-3 complexes hampers their structural characterization by X-ray crystallography, cryo-electron microscopy (EM) and traditional solution nuclear magnetic resonance (NMR) spectroscopy. Here, we employ solution 1D 19F-Trp NMR spectroscopy to characterize substrate binding and dimerization of 14-3-3 proteins, focusing on 14-3-3ζ - an abundant human isoform as an example. Both conserved Trp residues are located in distinct functionally important sites - the dimeric interface and the ligand-binding groove. We substituted them by 5F-Trp, thereby introducing a convenient NMR probe. Fluorination of the two Trp did not impact the stability and interaction properties of 14-3-3ζ in a substantive manner, permitting to carry out 19F NMR experiments to assess 14-3-3's structure and behavior. Importantly, 5F-Trp228 reports on binding of substrates in the amphipathic binding groove of 14-3-3ζ and permitted to distinguish distinct recognition modes. Thus, we established that 19F NMR is a powerful approach to evaluate the binding of partner proteins to 14-3-3 and to characterize the properties of the resulting complexes.
Collapse
Affiliation(s)
- Alexandra Náplavová
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czechia; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czechia
| | - Aneta Kozeleková
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czechia; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czechia
| | - Radek Crha
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czechia; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czechia
| | - Angela M Gronenborn
- Department of Structural Biology, University of Pittsburgh, School of Medicine, 1051 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA 15261, USA
| | - Jozef Hritz
- Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czechia; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czechia; Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czechia.
| |
Collapse
|
6
|
Delalande F, Østergaard SR, Gogl G, Cousido-Siah A, McEwen AG, Men Y, Salimova F, Rohrbacher A, Kostmann C, Nominé Y, Vincentelli R, Eberling P, Carapito C, Travé G, Monsellier E. Holdup Multiplex Assay for High-Throughput Measurement of Protein-Ligand Affinity Constants Using a Mass Spectrometry Readout. J Am Chem Soc 2025; 147:10886-10902. [PMID: 40129024 DOI: 10.1021/jacs.4c11102] [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: 03/26/2025]
Abstract
The accurate description and subsequent modeling of protein interactomes require quantification of their affinities at the proteome-wide scale. Here we develop and validate the Holdup Multiplex, a versatile assay with a mass spectrometry (MS) readout for profiling the affinities of a protein for large pools of peptides. The method can precisely quantify, in one single run, thousands of affinity constants over several orders of magnitude. The throughput, dynamic range, and sensitivity can be pushed to the performance limit of the MS readout. We applied the Holdup Multiplex to quantify in a few sample runs the affinities of the 14-3-3s, phosphoreader proteins highly abundant in humans, for 1000 different phosphopeptides. The seven human 14-3-3 isoforms were found to display similar specificities but staggered affinities, with 14-3-3γ being always the best binder and 14-3-3ε and σ being the weakest. Hundreds of new 14-3-3 binding sites were identified. We also identified dozens of 14-3-3 binding sites, some intervening in key signaling pathways, that were either stabilized or destabilized by the phytotoxin Fusicoccin-A. The results were corroborated by X-ray crystallography. Finally, we demonstrated the transferability of the Holdup Multiplex by quantifying the interactions of a PDZ domain for 5400 PBM peptides at once. The approach is applicable to any category of protein-binding ligands that can be quantifiable by mass spectrometry.
Collapse
Affiliation(s)
- François Delalande
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, Université de Strasbourg, IPHC UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| | - So Ren Østergaard
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Research Park, 2760 Maaloev, Denmark
| | - Gergo Gogl
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Alexandra Cousido-Siah
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Alastair G McEwen
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Yushi Men
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Farida Salimova
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, Université de Strasbourg, IPHC UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| | - Aurélien Rohrbacher
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Camille Kostmann
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Yves Nominé
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257 CNRS-Aix-Marseille Université, 13288 Marseille, France
| | - Pascal Eberling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, Université de Strasbourg, IPHC UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| | - Gilles Travé
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Elodie Monsellier
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| |
Collapse
|
7
|
Tomlinson ACA, Knox JE, Brunsveld L, Ottmann C, Yano JK. The "three body solution": Structural insights into molecular glues. Curr Opin Struct Biol 2025; 91:103007. [PMID: 40014904 DOI: 10.1016/j.sbi.2025.103007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Molecular glues are small molecules that nucleate novel or stabilize natural, protein-protein interactions resulting in a ternary complex. Their success in targeting difficult to drug proteins of interest has led to ever-increasing interest in their use as therapeutics and research tools. While molecular glues and their targets vary in structure, inspection of diverse ternary complexes reveals commonalities. Whether of high or low molecular weight, molecular glues are often rigid and form direct hydrophobic interactions with their target protein. There is growing evidence that these hotspots can accommodate multiple ternary complex binding modes and enable targeting of traditionally undruggable targets. Advances in screening from the molecular glue degrader literature and insights in structure-based drug design, especially from the non-degrading tri-complex work, are likely intersectional.
Collapse
Affiliation(s)
| | | | - Luc Brunsveld
- Eindhoven University of Technology, Eindhoven, Netherlands
| | | | | |
Collapse
|
8
|
Busnadiego I, Lork M, Fernbach S, Schiefer S, Tsolakos N, Hale BG. An atlas of protein phosphorylation dynamics during interferon signaling. Proc Natl Acad Sci U S A 2025; 122:e2412990122. [PMID: 40138345 PMCID: PMC12002234 DOI: 10.1073/pnas.2412990122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
Interferons (IFNs, types I-III) have pleiotropic functions in promoting antiviral and antitumor responses, as well as in modulating inflammation. Dissecting the signaling mechanisms elicited by different IFNs is therefore critical to understand their phenotypes. Here, we use mass spectrometry to investigate the early temporal dynamics of cellular protein phosphorylation in a human lung epithelial cell-line as it responds to stimulation with IFNα2, IFNβ, IFNω, IFNγ, or IFNλ1, representing all IFN types. We report an atlas of over 700 common or unique phosphorylation events reprogrammed by these different IFNs, revealing both previously known and uncharacterized modifications. While the proteins differentially phosphorylated following IFN stimulation have diverse roles, there is an enrichment of factors involved in chromatin remodeling, transcription, and RNA splicing. Functional screening and mechanistic studies identify that several proteins modified in response to IFNs contribute to host antiviral responses, either directly or by supporting IFN-stimulated gene or protein production. Among these, phosphorylation of PLEKHG3 at serine-1081 creates a phospho-regulated binding motif for the docking of 14-3-3 proteins, and together these factors contribute to coordinating efficient IFN-stimulated gene expression independent of early Janus kinase/signal transducer and activator of transcription signaling. Our findings map the global phosphorylation landscapes regulated by IFN types I, II, and III, and provide a key resource to explore their functional consequences.
Collapse
Affiliation(s)
- Idoia Busnadiego
- Institute of Medical Virology, University of Zurich, Zurich8057, Switzerland
| | - Marie Lork
- Institute of Medical Virology, University of Zurich, Zurich8057, Switzerland
| | - Sonja Fernbach
- Institute of Medical Virology, University of Zurich, Zurich8057, Switzerland
| | - Samira Schiefer
- Institute of Medical Virology, University of Zurich, Zurich8057, Switzerland
| | - Nikos Tsolakos
- Institute of Medical Virology, University of Zurich, Zurich8057, Switzerland
| | - Benjamin G. Hale
- Institute of Medical Virology, University of Zurich, Zurich8057, Switzerland
| |
Collapse
|
9
|
Howe J, Barbar EJ. Dynamic interactions of dimeric hub proteins underlie their diverse functions and structures: A comparative analysis of 14-3-3 and LC8. J Biol Chem 2025; 301:108416. [PMID: 40107617 PMCID: PMC12017986 DOI: 10.1016/j.jbc.2025.108416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/22/2025] Open
Abstract
Hub proteins interact with a host of client proteins and regulate multiple cellular functions. Dynamic hubs have a single binding interface for one client at a time resulting in competition among clients with the highest affinity. Dynamic dimeric hubs with two identical sites bind either two different client proteins or two chains of the same client to form homogenous complexes and could also form heterogeneous mixtures of interconverting complexes. Here, we review the interactions of the dimeric hubs 14-3-3 and LC8. 14-3-3 is a phosphoserine/threonine binding protein involved in structuring client proteins and regulating their phosphorylation. LC8 is involved in promoting the dimerization of client peptides and the rigidification of their disordered regions. Both 14-3-3 and LC8 are essential genes, with 14-3-3 playing a crucial role in apoptosis and cell cycle regulation, while LC8 is critical for the assembly of proteins involved in transport, DNA repair, and transcription. Interestingly, both protein dimers can dissociate by phosphorylation, which results in their interactome-wide changes. Their interactions are also regulated by the phosphorylation of their clients. Both form heterogeneous complexes with various functions including phase separation, signaling, and viral hijacking where they restrict the conformational heterogeneity of their dimeric clients that bind nucleic acids. This comparative analysis highlights the importance of dynamic protein-protein interactions in the diversity of functions of 14-3-3 and LC8 and how small differences in structures of interfaces explain why 14-3-3 is primarily involved in the regulation of phosphorylation states while LC8 is primarily involved in the regulation of assembly of large dynamic complexes.
Collapse
Affiliation(s)
- Jesse Howe
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA
| | - Elisar J Barbar
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA.
| |
Collapse
|
10
|
Jansen S, Narasimhan S, Cabre Fernandez P, Iľkovičová L, Kozeleková A, Králová K, Hritz J, Žídek L. Characterization of multiple binding sites on microtubule associated protein 2c recognized by dimeric and monomeric 14-3-3ζ. FEBS J 2025; 292:1991-2016. [PMID: 39877981 PMCID: PMC12001206 DOI: 10.1111/febs.17405] [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: 07/18/2024] [Revised: 10/09/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Microtubule associated protein 2 (MAP2) interacts with the regulatory protein 14-3-3ζ in a cAMP-dependent protein kinase (PKA) phosphorylation dependent manner. Using selective phosphorylation, calorimetry, nuclear magnetic resonance, chemical crosslinking, and X-ray crystallography, we characterized interactions of 14-3-3ζ with various binding regions of MAP2c. Although PKA phosphorylation increases the affinity of MAP2c for 14-3-3ζ in the proline rich region and C-terminal domain, unphosphorylated MAP2c also binds the dimeric 14-3-3ζ via its microtubule binding domain and variable central domain. Monomerization of 14-3-3ζ leads to the loss of affinity for the unphosphorylated residues. In neuroblastoma cell extract, MAP2c is heavily phosphorylated by PKA and the proline kinase ERK2. Although 14-3-3ζ dimer or monomer do not interact with the residues phosphorylated by ERK2, ERK2 phosphorylation of MAP2c in the C-terminal domain reduces the binding of MAP2c to both oligomeric variants of 14-3-3ζ.
Collapse
Affiliation(s)
- Séverine Jansen
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Subhash Narasimhan
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
- National Centre for Biomolecular Research, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Paula Cabre Fernandez
- National Centre for Biomolecular Research, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
- Research Institute Sant PauBarcelonaSpain
| | - Lucia Iľkovičová
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
- National Centre for Biomolecular Research, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Aneta Kozeleková
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
- National Centre for Biomolecular Research, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Kateřina Králová
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Jozef Hritz
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
- National Centre for Biomolecular Research, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
- Department of Chemistry, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Lukáš Žídek
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
- National Centre for Biomolecular Research, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| |
Collapse
|
11
|
Barrera EE, Skrabana R, Bustos DM. Deciphering opening mechanisms of 14-3-3 proteins. Protein Sci 2025; 34:e70108. [PMID: 40130781 PMCID: PMC11934215 DOI: 10.1002/pro.70108] [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: 09/26/2024] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/26/2025]
Abstract
The 14-3-3 proteins are a highly conserved family of regulatory molecules that play crucial roles in various cellular processes. They are known for their ability to bind to phosphorylated serine and threonine residues on target proteins, which allows them to modulate their activity, localization, and stability. In mammals, there are seven known paralogs of 14-3-3 proteins, designated as β, ε, ζ, η, σ, τ, and γ. Each paralog has distinct biological functions and tissue distributions, which allow a diverse range of regulatory roles in cellular processes. The conformational plasticity of 14-3-3s regulates their interaction with protein partners but has not yet been thoroughly characterized. We investigated this topic by classical molecular dynamics simulations and observed how the γ, ε, and ζ paralogs exhibit different opening rates. A PCA analysis identified the main modes of these opening-conformational variations. Using correlation-based tools and simulations with single amino acid substitutions, we have recognized how the amphipathic 14-3-3 groove opening is triggered by a distally located aliphatic-π interaction. The identified residues form a partially conserved small cavity between helices H6, H7, and H8, representing a potential paralog-specific drug site.
Collapse
Affiliation(s)
- Exequiel E. Barrera
- Instituto de Histología y Embriología de Mendoza (IHEM)Universidad Nacional de Cuyo, CONICETMendozaArgentina
| | - Rostislav Skrabana
- Institute of NeuroimmunologySlovak Academy of SciencesBratislavaSlovakia
| | - Diego M. Bustos
- Instituto de Histología y Embriología de Mendoza (IHEM)Universidad Nacional de Cuyo, CONICETMendozaArgentina
- Facultad de Ciencias Exactas y NaturalesUNCUYOMendozaArgentina
| |
Collapse
|
12
|
Zhao J, Liu S, Ren H, Afriyie OE, Zhang M, Xu D, Huang X. Genome-wide identification and comparative evolution of 14-3-3 gene family members in five Brassicaceae species. BMC Genomics 2025; 26:309. [PMID: 40155852 PMCID: PMC11954322 DOI: 10.1186/s12864-025-11513-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The 14-3-3 proteins are highly conserved regulatory eukaryotic proteins, which are crucial in growth, development, and stress responses. However, systematic characterization of the 14-3-3 gene family in Brassicaceae species and their evolutionary relationships have not been comprehensively reported. RESULTS This study conducted genome-wide identification, structural characteristics, and comparative evolutionary analysis of 14-3-3 gene family members in Arabidopsis thaliana, A. lyrata, A. pumila, Camelina sativa, and Brassica oleracea using comparative genomics. Overall, a total of 108 14-3-3 genes, which were phylogenetically classified into ε and non-ε groups were identified in the five species, with the non-ε members exhibiting more similar exon-intron structures and conserved motif patterns. Collinearity analysis revealed that the Brassicaceae 14-3-3 gene family members underwent varying degrees of expansion following whole-genome duplication (WGD) events. Notably, the number of 14-3-3 gene family members between A. lyrata and A. thaliana remained similar despite the former having approximately 1.66-fold larger genome size. In contrast, the number of 14-3-3 gene family members in A. pumila and C. sativa increased in proportionately to their genome size, while gene members in the more distantly related species to A. thaliana, B. oleracea, showed irregular expansion patterns. Selection pressure analysis revealed that 14-3-3 homologs in all the five species underwent purifying selection, with the group ε members experiencing relatively weaker purifying selection. Cloning of ApGRF6-2 gene from A. pumila indicated that the ApGRF6-2 protein was localized in the cell membrane and cytoplasm, while ectopic overexpression of ApGRF6-2 in A. thaliana could promote early flowering by upregulating the expression of floral meristem identity genes. CONCLUSION This study provides a comprehensive and systematic identification of the 14-3-3 gene family members in five Brassicaceae species using updated genome sequences, and the results could form a basis for further validation of functional and molecular mechanisms of 14-3-3 genes in plant growth, development, abiotic stress responses, as well as flowering regulation.
Collapse
Affiliation(s)
- Jingya Zhao
- College of Life Sciences, Shihezi University, Shihezi, 832003, China
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Shengqin Liu
- College of Life Sciences, Shihezi University, Shihezi, 832003, China
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Hui Ren
- College of Life Sciences, Shihezi University, Shihezi, 832003, China
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Owusu Edwin Afriyie
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Mengzhu Zhang
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Dachao Xu
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Xianzhong Huang
- Center for Crop Biotechnology, College of Agriculture, Anhui Science and Technology University, Chuzhou, 233100, China.
| |
Collapse
|
13
|
Romero-Zamora D, Rogers S, Low RRJ, Page SG, Lane BJE, Kosaka S, Robinson AB, French L, Lamm N, Ishikawa F, Hayashi MT, Cesare AJ. A CPC-shelterin-BTR axis regulates mitotic telomere deprotection. Nat Commun 2025; 16:2277. [PMID: 40097392 PMCID: PMC11914695 DOI: 10.1038/s41467-025-57456-8] [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: 02/13/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Telomeres prevent ATM activation by sequestering chromosome termini within telomere loops (t-loops). Mitotic arrest promotes telomere linearity and a localized ATM-dependent telomere DNA damage response (DDR) through an unknown mechanism. Using unbiased interactomics, biochemical screening, molecular biology, and super-resolution imaging, we found that mitotic arrest-dependent (MAD) telomere deprotection requires the combined activities of the Chromosome passenger complex (CPC) on shelterin, and the BLM-TOP3A-RMI1/2 (BTR) complex on t-loops. During mitotic arrest, the CPC component Aurora Kinase B (AURKB) phosphorylated both the TRF1 hinge and TRF2 basic domains. Phosphorylation of the TRF1 hinge domain enhances CPC and TRF1 interaction through the CPC Survivin subunit. Meanwhile, phosphorylation of the TRF2 basic domain promotes telomere linearity, activates a telomere DDR dependent on BTR-mediated double Holliday junction dissolution, and leads to mitotic death. We identify that the TRF2 basic domain functions in mitosis-specific telomere protection and reveal a regulatory role for TRF1 in controlling a physiological ATM-dependent telomere DDR. The data demonstrate that MAD telomere deprotection is a sophisticated active mechanism that exposes telomere ends to signal mitotic stress.
Collapse
Affiliation(s)
- Diana Romero-Zamora
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan
- IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Samuel Rogers
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Ronnie Ren Jie Low
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Scott G Page
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Blake J E Lane
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Shunya Kosaka
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan
- IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Andrew B Robinson
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Lucy French
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Noa Lamm
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Fuyuki Ishikawa
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan
| | - Makoto T Hayashi
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan.
- IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan.
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Anthony J Cesare
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
14
|
Benarroch E. What Is the Function and Relevance of 14-3-3 Proteins in Neurologic Disease? Neurology 2025; 104:e213418. [PMID: 39889260 DOI: 10.1212/wnl.0000000000213418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 02/02/2025] Open
|
15
|
Kamayirese S, Hansen LA, Lovas S. Ligand recognition by 14-3-3 proteins requires negative charges but not necessarily phosphorylation. FEBS Lett 2025; 599:838-847. [PMID: 39757510 PMCID: PMC11931987 DOI: 10.1002/1873-3468.15077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/07/2025]
Abstract
Protein-protein interactions involving 14-3-3 proteins regulate various cellular activities in normal and pathological conditions. These interactions have mostly been reported to be phosphorylation-dependent, but the 14-3-3 proteins also interact with unphosphorylated proteins. In this work, we investigated whether phosphorylation is required, or, alternatively, whether negative charges are sufficient for 14-3-3ε binding. We substituted the pThr residue of pT(502-510) peptide by residues with a varying number of negative charges and investigated the binding of the peptides to 14-3-3ε using MD simulations and biophysical methods. We demonstrated that at least one negative charge is required for the peptides to bind 14-3-3ε, although phosphorylation is not necessary, and that two negative charges are preferable for high affinity binding. This discovery opens up new approaches for designing peptide-based 14-3-3 protein inhibitors.
Collapse
Affiliation(s)
| | - Laura A. Hansen
- Department of Biomedical SciencesCreighton UniversityOmahaNEUSA
| | - Sándor Lovas
- Department of Biomedical SciencesCreighton UniversityOmahaNEUSA
| |
Collapse
|
16
|
Thomas MG, Fernández-Alvarez AJ, Giménez M, Corvetto Aristarain F, Cozza LH, Pimentel J, Pessoa J, Pascual ML, Boscaglia L, Habif M, Corbat A, La Spina PE, Peters T, Bustos DM, Carmo-Fonseca M, Grecco HE, Boccaccio GL. The non-canonical Smoothened-AMPK axis regulates Smaug1 biomolecular condensates. J Cell Sci 2025; 138:JCS263433. [PMID: 39925132 DOI: 10.1242/jcs.263433] [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] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Biomolecular condensates (BMCs) emerge as important players in RNA regulation. The RNA-binding protein Smaug forms cytosolic BMCs in mammals, insects and yeasts and affects mitochondrial function and/or responses to nutrient deprivation. Here, we found that the non-canonical activation of the Smoothened (SMO)-AMPK pathway, which is known to affect energy metabolism, triggers the immediate disassembly of BMCs formed by a number of human and rodent Smaug orthologs, whereas processing bodies remain rather unaltered. A non-phosphorylatable SMO mutant abrogated the effect, involving SMO phosphorylation in human (h)Smaug1 (also known as SAMD4A) BMCs regulation. Three mechanistically different SMO ligands, namely SAG, GSA-10 and cyclopamine, elicited a similar response, which was blocked upon AMPK pharmacological inhibition. Polysome disassembly by puromycin halted Smaug1 BMC dissolution, thus suggesting that unbound transcripts became translationally active. Single-molecule fluorescent in situ hybridization illustrated the release of UQCRC1 mRNA. Finally, Smaug1 is a phosphoprotein bound by 14-3-3 proteins, and the competitive inhibitor difopein blocked the response to non-canonical SMO stimulation. We propose that the regulated condensation and dispersion of Smaug1 BMCs generate translational changes that contribute to metabolic regulation downstream of the non-canonical SMO-AMPK axis.
Collapse
Affiliation(s)
- María Gabriela Thomas
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Ana Julia Fernández-Alvarez
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Macarena Giménez
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Francisco Corvetto Aristarain
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Lucas Helio Cozza
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Jerónimo Pimentel
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - João Pessoa
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Malena Lucía Pascual
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Lara Boscaglia
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Martín Habif
- Instituto de Física de Buenos Aires (IFIBA). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Agustín Corbat
- Instituto de Física de Buenos Aires (IFIBA). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Pablo Ezequiel La Spina
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Tomás Peters
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Diego Martín Bustos
- Instituto de Histología y Embriología (IHEM). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), M5502JMA, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - Maria Carmo-Fonseca
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Hernán Edgardo Grecco
- Instituto de Física de Buenos Aires (IFIBA). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| | - Graciela Lidia Boccaccio
- Fundación Instituto Leloir (FIL), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Department of Molecular and Cellular Biology and Physiology (FBMyC), Facultad de Ciencias Exactas y Naturales (FCEN), University of Buenos Aires (UBA), C1428EHA Buenos Aires, Argentina
| |
Collapse
|
17
|
Sedlov IA, Sluchanko NN. Biochemical signatures strongly demarcate phylogenetic groups of plant 14-3-3 isoforms. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2025; 121:e70017. [PMID: 40051177 DOI: 10.1111/tpj.70017] [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: 05/11/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 05/13/2025]
Abstract
Interaction of dimeric 14-3-3 proteins with phosphotargets regulates various physiological processes in plants, from flowering to transpiration and salt tolerance. Several genes express distinct 14-3-3 "isoforms," particularly numerous in plants, but these are unevenly studied even in model species. Here we systematically investigated twelve 14-3-3 isoforms from Arabidopsis thaliana. While all these proteins can homodimerize, four isoforms representing a supposedly more ancestral, epsilon phylogenetic group (iota, mu, omicron, epsilon), but not their eight non-epsilon counterparts (omega, phi, chi, psi, upsilon, nu, kappa, lambda), exhibit concentration-dependent monomerization, and pronounced surface hydrophobicity at physiologically relevant protein concentrations and under crowding conditions typical for the cell. We show that dramatically lowered thermodynamic stabilities entail aggregation of the epsilon group isoforms at near-physiological temperatures and accelerate their proteolytic degradation in vitro and in plant cell lysates. Mutations in 14-3-3 iota, inspired by structural analysis, helped us rescue non-epsilon behavior and pinpoint key positions responsible for the epsilon/non-epsilon demarcation. Combining two major demarcating positions (namely, 27th and 51st in omega) and differences in biochemical properties, we developed an epsilon/non-epsilon demarcation criterion that classified 89% of available 14-3-3 sequences from Dicots, Monocots, Gymnosperms, Ferns, and Lycophytes with 99.7% accuracy, and reliably predicted biochemical properties of a given 14-3-3 isoform, which we experimentally verified for distant 14-3-3 isoforms from Selaginella moellendorffii. The proven occurrence of isoforms of both groups in primitive plants refines the traditional phylogenetic, solely sequence-based analysis and provides intriguing insights into the evolutionary history of the epsilon phylogenetic group.
Collapse
Affiliation(s)
- Ilya A Sedlov
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071, Moscow, Russia
- School of Biology, Department of Biochemistry, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071, Moscow, Russia
| |
Collapse
|
18
|
Shimamura MI, Satoh K. Challenges and Revisions in Diagnostic Criteria: Advancing Early Detection of Prion Diseases. Int J Mol Sci 2025; 26:2037. [PMID: 40076658 PMCID: PMC11900056 DOI: 10.3390/ijms26052037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Prion diseases are fatal neurological disorders characterized by abnormal protein accumulation in the brain, leading to neurodegeneration, dementia, and ataxia. Sporadic Creutzfeldt-Jakob disease (sCJD), the most common form, accounts for 80-90% of cases and progresses rapidly, with most patients surviving <6 months to a year after symptom onset, indicating the importance of early diagnosis. The disease is classified into six subtypes based on PRNP gene polymorphisms, with differences in protein degradation patterns contributing to the diversity of clinical symptoms. However, diagnosis remains challenging because of the variability in clinical presentation and disease duration. Traditional diagnostic criteria established by the World Health Organization (WHO) rely on clinical findings, electroencephalogram, and cerebrospinal fluid tests, such as the 14-3-3 protein assay. However, these criteria require pathological confirmation, often delaying diagnosis. The recently proposed Hermann's criteria represent a significant advancement by incorporating newer biomarkers, including magnetic resonance imaging, real-time quaking-induced conversion assay, tau protein, and neurofilament light chain. These criteria improve diagnostic sensitivity and specificity but have a slightly higher risk of false positives. This review compares the effectiveness of these biomarkers with the WHO criteria and highlights the importance of early diagnosis for improving patient care.
Collapse
Affiliation(s)
- Mika Inada Shimamura
- Biomedical Research Support Center, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Katsuya Satoh
- Unit of Medical and Dental Sciences, Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
- Leading Medical Research Core Unit, Department of Brain Research Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| |
Collapse
|
19
|
Theisen F, Prestel A, Jacobsen NL, Nyhegn-Eriksen OK, Olsen JG, Kragelund BB. Proline cis/ trans Conformational Selection Controls 14-3-3 Binding. J Am Chem Soc 2025; 147:5714-5724. [PMID: 39909402 PMCID: PMC11848828 DOI: 10.1021/jacs.4c13462] [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: 09/26/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/07/2025]
Abstract
Intrinsically disordered protein regions (IDRs) are structurally dynamic yet functional, often interacting with other proteins through short linear motifs (SLiMs). Proline residues in IDRs introduce conformational heterogeneity on a uniquely slow time scale arising from cis/trans isomerization of the Xaa-Pro peptide bond. Here, we explore the role of proline isomerization in the interaction between the prolactin receptor (PRLR) and 14-3-3. Using NMR spectroscopy, thermodynamic profiling, and molecular dynamics (MD) simulations, we uncover a unique proline isomer-dependent binding, with a cis conformation affinity 3 orders of magnitude higher than the trans. MD simulations identify structural constraints in the narrow 14-3-3 binding groove that provide an explanation for the observed isomer selectivity. The cis preference of WT PRLR introduces a slow kinetic component relevant to signal propagation and a steric component that impacts chain direction. Proline isomerization constitutes a previously unrecognized selective component relevant to the ubiquitous 14-3-3 interactome. Given the prevalence of prolines in IDRs and SLiMs, our study highlights the importance of considering the distinct properties of proline isomers in experimental design and data interpretation to fully comprehend IDR functionality.
Collapse
Affiliation(s)
- Frederik
F. Theisen
- Structural
Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark
- Institut
de Biologie Structurale, 71 avenue des Martyrs, Grenoble 38000, France
| | - Andreas Prestel
- Structural
Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark
| | - Nina L. Jacobsen
- Structural
Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark
| | - Oline K. Nyhegn-Eriksen
- Structural
Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark
| | - Johan G. Olsen
- Structural
Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark
| | - Birthe B. Kragelund
- Structural
Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark
| |
Collapse
|
20
|
Tian Y, Li L, Wu L, Xu Q, Li Y, Pan H, Bing T, Bai X, Finko AV, Li Z, Bian J. Recent Developments in 14-3-3 Stabilizers for Regulating Protein-Protein Interactions: An Update. J Med Chem 2025; 68:2124-2146. [PMID: 39902774 DOI: 10.1021/acs.jmedchem.4c01936] [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: 02/06/2025]
Abstract
14-3-3 proteins play a crucial role in the regulation of protein-protein interactions, impacting various cellular processes and disease mechanisms. Recent advancements have led to the development of stabilizers that enhance the binding of 14-3-3 proteins to clients, presenting promising therapeutic potentials. This perspective provides an updated overview of the latest developments in the field of 14-3-3 stabilizers, with a focus on their design, synthesis, and biological evaluation. We discuss the structural basis for the interaction between 14-3-3 proteins and their ligands, highlighting key modifications that enhance binding affinity and selectivity. Additionally, we explore the therapeutic applications of 14-3-3 stabilizers across major therapeutic areas such as cancer, metabolic disorders, and neurodegenerative diseases. By summarizing recent research findings and technological advancements, this perspective aims to shed light on the current state of 14-3-3 stabilizer developments and outline future directions for optimizing these compounds as effective therapeutic agents.
Collapse
Affiliation(s)
- Yucheng Tian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Longjing Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liuyi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qianqian Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yaojie Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Huawei Pan
- ICE Bioscience, Bldg 15, Yd 18, Kechuang 13th St, Etown, Tongzhou Dist, Beijing 100176, China
| | - Tiejun Bing
- ICE Bioscience, Bldg 15, Yd 18, Kechuang 13th St, Etown, Tongzhou Dist, Beijing 100176, China
| | - Xiumei Bai
- Department of Chemistry, Lomonosov Moscow State University (MSU), Moscow 119991, Russia
| | - Alexander V Finko
- Department of Chemistry, Lomonosov Moscow State University (MSU), Moscow 119991, Russia
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
21
|
Spooner HC, Costa AD, Westhoff M, Hernández-González A, Ibrahimkhail H, Yarov-Yarovoy V, Horne MC, Dickson EJ, Dixon RE. 14-3-3 promotes sarcolemmal expression of cardiac Ca V1.2 and nucleates isoproterenol-triggered channel superclustering. Proc Natl Acad Sci U S A 2025; 122:e2413308122. [PMID: 39869803 PMCID: PMC11804677 DOI: 10.1073/pnas.2413308122] [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: 07/02/2024] [Accepted: 12/16/2024] [Indexed: 01/29/2025] Open
Abstract
The L-type Ca2+ channel (CaV1.2) is essential for cardiac excitation-contraction coupling. To contribute to the inward Ca2+ flux that drives Ca2+-induced-Ca2+-release, CaV1.2 channels must be expressed on the sarcolemma; thus the regulatory mechanisms that tune CaV1.2 expression to meet contractile demand are an emerging area of research. A ubiquitously expressed protein called 14-3-3 has been proposed to affect Ca2+ channel trafficking in nonmyocytes; however, whether 14-3-3 has similar effects on CaV1.2 in cardiomyocytes is unknown. 14-3-3 preferentially binds phospho-serine/threonine residues to affect many cellular processes and is known to regulate cardiac ion channels including NaV1.5 and the human ether-à-go-go-related gene (hERG) potassium channel. Altered 14-3-3 expression and function have been implicated in cardiac pathologies including hypertrophy. Accordingly, we tested the hypothesis that 14-3-3 interacts with CaV1.2 in a phosphorylation-dependent manner and regulates cardiac CaV1.2 trafficking and recycling. Confocal imaging, proximity ligation assays, superresolution imaging, and coimmunoprecipitation revealed a population of 14-3-3 colocalized and closely associated with CaV1.2. The degree of 14-3-3/CaV1.2 colocalization increased upon stimulation of β-adrenergic receptors with isoproterenol. Notably, only the 14-3-3-associated CaV1.2 population displayed increased cluster size with isoproterenol, revealing a role for 14-3-3 as a nucleation factor that directs CaV1.2 superclustering. Isoproterenol-stimulated augmentation of sarcolemmal CaV1.2 expression, Ca2+ currents, and Ca2+ transients in ventricular myocytes were strengthened by 14-3-3 overexpression and attenuated by 14-3-3 inhibition. These data support a model where 14-3-3 interacts with CaV1.2 in a phosphorylation-dependent manner to promote enhanced trafficking/recycling, clustering, and activity during β-adrenergic stimulation.
Collapse
Affiliation(s)
- Heather C. Spooner
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
| | - Alexandre D. Costa
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
| | - Maartje Westhoff
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
| | | | - Husna Ibrahimkhail
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
- Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA95616
| | - Mary C. Horne
- Department of Pharmacology, University of California Davis, Davis, CA95616
| | - Eamonn J. Dickson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
| | - Rose E. Dixon
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA95616
| |
Collapse
|
22
|
Lonare A, Raychaudhuri K, Shah S, Madhu G, Sachdeva A, Basu S, Thorat R, Gupta S, Dalal SN. 14-3-3σ restricts YY1 to the cytoplasm, promoting therapy resistance, and tumor progression in colorectal cancer. Int J Cancer 2025; 156:623-637. [PMID: 39239852 PMCID: PMC11622004 DOI: 10.1002/ijc.35176] [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: 02/29/2024] [Revised: 07/11/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
14-3-3σ functions as an oncogene in colorectal cancer and is associated with therapy resistance. However, the mechanisms underlying these observations are not clear. The results in this report demonstrate that loss of 14-3-3σ in colorectal cancer cells leads to a decrease in tumor formation and increased sensitivity to chemotherapy. The increased sensitivity to chemotherapy is due to a decrease in the expression of UPR pathway genes in the absence of 14-3-3σ. 14-3-3σ promotes expression of the UPR pathway genes by binding to the transcription factor YY1 and preventing the nuclear localization of YY1. YY1, in the absence of 14-3-3σ, shows increased nuclear localization and binds to the promoter of the UPR pathway genes, resulting in decreased gene expression. Similarly, a YY1 mutant that cannot bind to 14-3-3σ also shows increased nuclear localization and is enriched on the promoter of the UPR pathway genes. Finally, inhibition of the UPR pathway with genetic or pharmacological approaches sensitizes colon cancer cells to chemotherapy. Our results identify a novel mechanism by which 14-3-3σ promotes tumor progression and therapy resistance in colorectal cancer by maintaining UPR gene expression.
Collapse
Affiliation(s)
- Amol Lonare
- Cell and Tumour Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
- Homi Bhabha National Institute, Training School ComplexMumbaiIndia
| | - Kumarkrishna Raychaudhuri
- Cell and Tumour Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
- Homi Bhabha National Institute, Training School ComplexMumbaiIndia
| | - Sanket Shah
- Homi Bhabha National Institute, Training School ComplexMumbaiIndia
- Epigenetics and Chromatin Biology Group, Gupta Lab, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi MumbaiIndia
- Present address:
Weill Cornell MedicineNew YorkNew YorkUSA
| | - Gifty Madhu
- Cell and Tumour Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| | - Anoushka Sachdeva
- Cell and Tumour Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| | - Sneha Basu
- Cell and Tumour Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| | - Sanjay Gupta
- Homi Bhabha National Institute, Training School ComplexMumbaiIndia
- Epigenetics and Chromatin Biology Group, Gupta Lab, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi MumbaiIndia
| | - Sorab N. Dalal
- Cell and Tumour Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
- Homi Bhabha National Institute, Training School ComplexMumbaiIndia
| |
Collapse
|
23
|
Obergfell E, Hohmann U, Moretti A, Chen H, Hothorn M. Mechanistic Insights into the Function of 14-3-3 Proteins as Negative Regulators of Brassinosteroid Signaling in Arabidopsis. PLANT & CELL PHYSIOLOGY 2024; 65:1674-1688. [PMID: 38783418 PMCID: PMC11558545 DOI: 10.1093/pcp/pcae056] [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: 10/13/2023] [Revised: 04/24/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
Brassinosteroids (BRs) are vital plant steroid hormones sensed at the cell surface by a membrane signaling complex comprising the receptor kinase BRI1 and a SERK family co-receptor kinase. Activation of this complex lead to dissociation of the inhibitor protein BKI1 from the receptor and to differential phosphorylation of BZR1/BES1 transcription factors by the glycogen synthase kinase 3 protein BIN2. Many phosphoproteins of the BR signaling pathway, including BRI1, SERKs, BKI1 and BZR1/BES1 can associate with 14-3-3 proteins. In this study, we use quantitative ligand binding assays to define the minimal 14-3-3 binding sites in the N-terminal lobe of the BRI1 kinase domain, in BKI1, and in BZR1 from Arabidopsis thaliana. All three motifs require to be phosphorylated to specifically bind 14-3-3s with mid- to low-micromolar affinity. BR signaling components display minimal isoform preference within the 14-3-3 non-ε subgroup. 14-3-3λ and 14-3-3 ω isoform complex crystal structures reveal that BKI1 and BZR1 bind as canonical type II 14-3-3 linear motifs. Disruption of key amino acids in the phosphopeptide binding site through mutation impairs the interaction of 14-3-3λ with all three linear motifs. Notably, quadruple loss-of-function mutants from the non-ε group exhibit gain-of-function BR signaling phenotypes, suggesting a role for 14-3-3 proteins as overall negative regulators of the BR pathway. Collectively, our work provides further mechanistic and genetic evidence for the regulatory role of 14-3-3 proteins at various stages of the BR signaling cascade.
Collapse
Affiliation(s)
- Elsa Obergfell
- Structural Plant Biology Laboratory, Department of Plant Sciences, University of Geneva, 30 Quai E. Ansermet, Geneva 1211, Switzerland
| | - Ulrich Hohmann
- Structural Plant Biology Laboratory, Department of Plant Sciences, University of Geneva, 30 Quai E. Ansermet, Geneva 1211, Switzerland
| | - Andrea Moretti
- Structural Plant Biology Laboratory, Department of Plant Sciences, University of Geneva, 30 Quai E. Ansermet, Geneva 1211, Switzerland
| | - Houming Chen
- Structural Plant Biology Laboratory, Department of Plant Sciences, University of Geneva, 30 Quai E. Ansermet, Geneva 1211, Switzerland
| | - Michael Hothorn
- Structural Plant Biology Laboratory, Department of Plant Sciences, University of Geneva, 30 Quai E. Ansermet, Geneva 1211, Switzerland
| |
Collapse
|
24
|
Kasahara K, Kawade R, Nakakido M, Matsunaga R, Akiba H, Entzminger KC, Maruyama T, Okumura SCJ, Caaveiro JMM, Kuroda D, Tsumoto K. Unveiling the structural mechanisms behind high affinity and selectivity in phosphorylated epitope-specific rabbit antibodies. J Biol Chem 2024; 300:107989. [PMID: 39542251 DOI: 10.1016/j.jbc.2024.107989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/14/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Protein phosphorylation is a crucial process in various cellular functions, and its irregularities have been implicated in several diseases, including cancer. Antibodies are commonly employed to detect protein phosphorylation in research. However, unlike the extensive studies on recognition mechanisms of the phosphate group by proteins such as kinases and phosphatases, only a few studies have explored antibody mechanisms. In this study, we produced and characterized two rabbit monoclonal antibodies that recognize a monophosphorylated Akt peptide. Through crystallography, thermodynamic mutational analyses, and molecular dynamics simulations, we investigated the unique recognition mechanism that enables higher binding affinity and selectivity of the antibodies compared to other generic proteins with lower binding affinity to phosphorylated epitopes. Our results demonstrate that molecular dynamics simulations provide novel insights into the dynamic aspects of molecular recognition of posttranslational modifications by proteins beyond static crystal structures, highlighting how specific atomic level interactions drive the exceptional affinity and selectivity of antibodies.
Collapse
Affiliation(s)
- Keisuke Kasahara
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Raiji Kawade
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makoto Nakakido
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryo Matsunaga
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroki Akiba
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | | | | | | | - Jose M M Caaveiro
- Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka-shi, Japan
| | - Daisuke Kuroda
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan.
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, Japan; Abwiz Bio Inc., San Diego, California, USA; Medical Proteomics Laboratory, Institute of Medical Sciences, The University of Tokyo, Minato-ku, Tokyo, Japan.
| |
Collapse
|
25
|
Kamayirese S, Hansen LA, Lovas S. Negative Charges, Not Necessary Phosphorylation, are Required for Ligand Recognition by 14-3-3 Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613320. [PMID: 39345434 PMCID: PMC11429721 DOI: 10.1101/2024.09.16.613320] [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: 10/01/2024]
Abstract
Protein-protein interactions involving 14-3-3 proteins regulate various cellular activities in normal and pathological conditions. These interactions have mostly been reported to be phosphorylation-dependent, but the 14-3-3 proteins also interact with unphosphorylated proteins. In this work, we investigated whether phosphorylation is required, or, alternatively, whether negative charges are sufficient for 14-3-3ε binding. We substituted the pThr residue of pT(502-510) peptide by residues with varying number of negative charges, and investigated binding of the peptides to 14-3-3ε using MD simulations and biophysical methods. We demonstrated that at least one negative charge is required for the peptides to bind 14-3-3ε while phosphorylation is not necessary, and that two negative charges are preferable for high affinity binding.
Collapse
Affiliation(s)
- Seraphine Kamayirese
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska 68178, United States
| | - Laura A. Hansen
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska 68178, United States
| | - Sándor Lovas
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska 68178, United States
| |
Collapse
|
26
|
Simon JJ, Fowler DM, Maly DJ. Multiplexed profiling of intracellular protein abundance, activity, interactions and druggability with LABEL-seq. Nat Methods 2024; 21:2094-2106. [PMID: 39433876 PMCID: PMC11785348 DOI: 10.1038/s41592-024-02456-7] [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: 05/01/2024] [Accepted: 09/10/2024] [Indexed: 10/23/2024]
Abstract
Here we describe labeling with barcodes and enrichment for biochemical analysis by sequencing (LABEL-seq), an assay for massively parallel profiling of pooled protein variants in human cells. By leveraging the intracellular self-assembly of an RNA-binding domain (RBD) with a stable, variant-encoding RNA barcode, LABEL-seq facilitates the direct measurement of protein properties and functions using simple affinity enrichments of RBD protein fusions, followed by high-throughput sequencing of co-enriched barcodes. Measurement of ~20,000 variant effects for ~1,600 BRaf variants revealed that variation at positions frequently mutated in cancer minimally impacted intracellular abundance but could dramatically alter activity, protein-protein interactions and druggability. Integrative analysis identified networks of positions with similar biochemical roles and enabled modeling of variant effects on cell proliferation and small molecule-promoted degradation. Thus, LABEL-seq enables direct measurement of multiple biochemical properties in a native cellular context, providing insights into protein function, disease mechanisms and druggability.
Collapse
Affiliation(s)
- Jessica J Simon
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|
27
|
Foo B, Amedei H, Kaur S, Jaawan S, Boshnakovska A, Gall T, de Boer RA, Silljé HHW, Urlaub H, Rehling P, Lenz C, Lehnart SE. Unbiased complexome profiling and global proteomics analysis reveals mitochondrial impairment and potential changes at the intercalated disk in presymptomatic R14Δ/+ mice hearts. PLoS One 2024; 19:e0311203. [PMID: 39446877 PMCID: PMC11501035 DOI: 10.1371/journal.pone.0311203] [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: 05/24/2024] [Accepted: 09/15/2024] [Indexed: 10/26/2024] Open
Abstract
Phospholamban (PLN) is a sarco-endoplasmic reticulum (SER) membrane protein that regulates cardiac contraction/relaxation by reversibly inhibiting the SERCA2a Ca2+-reuptake pump. The R14Δ-PLN mutation causes severe cardiomyopathy that is resistant to conventional treatment. Protein complexes and higher-order supercomplexes such as intercalated disk components and Ca+2-cycling domains underlie many critical cardiac functions, a subset of which may be disrupted by R14Δ-PLN. Complexome profiling (CP) is a proteomics workflow for systematic analysis of high molecular weight (MW) protein complexes and supercomplexes. We hypothesize that R14Δ-PLN may alter a subset of these assemblies, and apply CP workflows to explore these changes in presymptomatic R14Δ/+ mice hearts. Ventricular tissues from presymptomatic 28wk-old WT and R14Δ/+ mice were homogenized under non-denaturing conditions, fractionated by size-exclusion chromatography (SEC) with a linear MW-range exceeding 5 MDa, and subjected to quantitative data-independent acquisition mass spectrometry (DIA-MS) analysis. Unfortunately, current workflows for the systematic analysis of CP data proved ill-suited for use in cardiac samples. Most rely upon curated protein complex databases to provide ground-truth for analysis; however, these are derived primarily from cancerous or immortalized cell lines and, consequently, cell-type specific complexes (including cardiac-specific machinery potentially affected in R14Δ-PLN hearts) are poorly covered. We thus developed PERCOM: a novel CP data-analysis strategy that does not rely upon these databases and can, furthermore, be implemented on widely available spreadsheet software. Applying PERCOM to our CP dataset resulted in the identification of 296 proteins with disrupted elution profiles. Hits were significantly enriched for mitochondrial and intercalated disk (ICD) supercomplex components. Changes to mitochondrial supercomplexes were associated with reduced expression of mitochondrial proteins and maximal oxygen consumption rate. The observed alterations to mitochondrial and ICD supercomplexes were replicated in a second cohort of "juvenile" 9wk-old mice. These early-stage changes to key cardiac machinery may contribute to R14Δ-PLN pathogenesis.
Collapse
Affiliation(s)
- Brian Foo
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Hugo Amedei
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Surmeet Kaur
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Samir Jaawan
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Tanja Gall
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, Rotterdam, the Netherlands
| | - Herman H. W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henning Urlaub
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Christof Lenz
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stephan E. Lehnart
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
Fischer SN, Claussen ER, Kourtis S, Sdelci S, Orchard S, Hermjakob H, Kustatscher G, Drew K. hu.MAP3.0: Atlas of human protein complexes by integration of > 25,000 proteomic experiments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617930. [PMID: 39464102 PMCID: PMC11507723 DOI: 10.1101/2024.10.11.617930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Macromolecular protein complexes carry out most functions in the cell including essential functions required for cell survival. Unfortunately, we lack the subunit composition for all human protein complexes. To address this gap we integrated >25,000 mass spectrometry experiments using a machine learning approach to identify > 15,000 human protein complexes. We show our map of protein complexes is highly accurate and more comprehensive than previous maps, placing ~75% of human proteins into their physical contexts. We globally characterize our complexes using protein co-variation data (ProteomeHD.2) and identify co-varying complexes suggesting common functional associations. Our map also generates testable functional hypotheses for 472 uncharacterized proteins which we support using AlphaFold modeling. Additionally, we use AlphaFold modeling to identify 511 mutually exclusive protein pairs in hu.MAP3.0 complexes suggesting complexes serve different functional roles depending on their subunit composition. We identify expression as the primary way cells and organisms relieve the conflict of mutually exclusive subunits. Finally, we import our complexes to EMBL-EBI's Complex Portal (https://www.ebi.ac.uk/complexportal/home) as well as provide complexes through our hu.MAP3.0 web interface (https://humap3.proteincomplexes.org/). We expect our resource to be highly impactful to the broader research community.
Collapse
Affiliation(s)
- Samantha N. Fischer
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607
| | - Erin R. Claussen
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607
| | - Savvas Kourtis
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sara Sdelci
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Georg Kustatscher
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Kevin Drew
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607
| |
Collapse
|
29
|
Zhou R, Hu W, Ma PX, Liu CJ. Versatility of 14-3-3 proteins and their roles in bone and joint-related diseases. Bone Res 2024; 12:58. [PMID: 39406741 PMCID: PMC11480210 DOI: 10.1038/s41413-024-00370-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/30/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Bone and joint-related diseases, including osteoarthritis (OA), rheumatoid arthritis (RA), and bone tumors, pose significant health challenges due to their debilitating effects on the musculoskeletal system. 14-3-3 proteins, a family of conserved regulatory molecules, play a critical role in the pathology of these diseases. This review discusses the intricate structure and multifunctionality of 14-3-3 proteins, their regulation of signaling pathways, and their interactions with other proteins. We underscore the significance of 14-3-3 proteins in the regulation of osteoblasts, osteoclasts, chondrocytes, and bone remodeling, all key factors in the maintenance and dysfunction of bone and joint systems. Specific focus is directed toward elucidating the contribution of 14-3-3 proteins in the pathology of OA, RA, and bone malignancies, where dysregulated 14-3-3-mediated signaling cascades have been implicated in the disease processes. This review illuminates how the perturbation of 14-3-3 protein interactions can lead to the pathological manifestations observed in these disorders, including joint destruction and osteolytic activity. We highlight cutting-edge research that positions 14-3-3 proteins as potential biomarkers for disease progression and as innovative therapeutic targets, offering new avenues for disease intervention and management.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Weirong Hu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Peter X Ma
- Department of Biologic and Materials Sciences and Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
Kiriyama H, Kinoshita SN, Hayashi Y, Honda R, Kasuga S, Kinoshita T, Irieda H, Ohkanda J. Fungal toxin fusicoccin enhances plant growth by upregulating 14-3-3 interaction with plasma membrane H +-ATPase. Sci Rep 2024; 14:23431. [PMID: 39379425 PMCID: PMC11461981 DOI: 10.1038/s41598-024-73979-4] [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: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
Fusicoccin-A (FC-A) is a diterpene glucoside produced by a pathogenic fungus. Since its discovery, FC-A has been widely recognized as a phytotoxin that induces stomatal opening and leaf wilting, eventually leading to plant death. In this study, we present the first evidence that FC-A enhances plant growth by stabilizing the protein-protein interaction between plasma membrane (PM) H+-ATPase and 14-3-3 in guard cells. Long-term treatment of Arabidopsis plants with FC-A resulted in ~ 30% growth enhancement. Structurally similar fusicoccin-J (FC-J) showed a similar degree of growth-promotion activity as FC-A, whereas the more hydrophilic fusicoccin-H (FC-H) exhibited no effect on plant growth, indicating that the enhancement of plant growth observed with FC-A and FC-J involves upregulation of the protein-protein interaction between PM H+-ATPase and 14-3-3 in guard cells, which promotes stomatal opening and photosynthesis.
Collapse
Grants
- 22K19106 Japan Society for the Promotion of Science
- 19K05992 Japan Society for the Promotion of Science
- 20H05687 Japan Society for the Promotion of Science
- 20H04769 Ministry of Education, Culture, Sports, Science and Technology
- 20H05910 Ministry of Education, Culture, Sports, Science and Technology
- LEADER Ministry of Education, Culture, Sports, Science and Technology
- University Research Administration Fund Shinshu University
- 2021 Japan Society for Bioscience, Biotechnology, and Agrochemistry
Collapse
Affiliation(s)
- Hironaru Kiriyama
- Graduate School of Science and Technology, Shinshu University, 8304 Minami-Minowa, Kami-Ina, 399-4598, Nagano, Japan
| | - Satoru N Kinoshita
- Graduate School of Science, Nagoya University, Nagoya, Aichi, 464-8602, Japan
| | - Yuki Hayashi
- Graduate School of Science, Nagoya University, Nagoya, Aichi, 464-8602, Japan
| | - Rikako Honda
- Graduate School of Science and Technology, Shinshu University, 8304 Minami-Minowa, Kami-Ina, 399-4598, Nagano, Japan
| | - Shigemitsu Kasuga
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan
| | - Toshinori Kinoshita
- Graduate School of Science, Nagoya University, Nagoya, Aichi, 464-8602, Japan
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Aichi, 464-8602, Japan
| | - Hiroki Irieda
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan
- Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 8304 Minami- Minowa, Kami-Ina, Nagano, 399-4598, Japan
| | - Junko Ohkanda
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan.
- Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 8304 Minami- Minowa, Kami-Ina, Nagano, 399-4598, Japan.
| |
Collapse
|
31
|
Colino-Lage H, Guerrero-Gómez D, Gómez-Orte E, González X, Martina JA, Dansen TB, Ayuso C, Askjaer P, Puertollano R, Irazoqui JE, Cabello J, Miranda-Vizuete A. Regulation of Caenorhabditis elegans HLH-30 subcellular localization dynamics: Evidence for a redox-dependent mechanism. Free Radic Biol Med 2024; 223:369-383. [PMID: 39059513 PMCID: PMC11977398 DOI: 10.1016/j.freeradbiomed.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Basic Helix-Loop-Helix (bHLH) transcription factors TFEB/TFE3 and HLH-30 are key regulators of autophagy induction and lysosomal biogenesis in mammals and C. elegans, respectively. While much is known about the regulation of TFEB/TFE3, how HLH-30 subcellular dynamics and transactivation are modulated are yet poorly understood. Thus, elucidating the regulation of C. elegans HLH-30 will provide evolutionary insight into the mechanisms governing the function of bHLH transcription factor family. We report here that HLH-30 is retained in the cytoplasm mainly through its conserved Ser201 residue and that HLH-30 physically interacts with the 14-3-3 protein FTT-2 in this location. The FoxO transcription factor DAF-16 is not required for HLH-30 nuclear translocation upon stress, despite that both proteins partner to form a complex that coordinately regulates several organismal responses. Similar as described for DAF-16, the importin IMB-2 assists HLH-30 nuclear translocation, but constitutive HLH-30 nuclear localization is not sufficient to trigger its distinctive transcriptional response. Furthermore, we identify FTT-2 as the target of diethyl maleate (DEM), a GSH depletor that causes a transient nuclear translocation of HLH-30. Together, our work demonstrates that the regulation of TFEB/TFE3 and HLH-30 family members is evolutionarily conserved and that, in addition to a direct redox regulation through its conserved single cysteine residue, HLH-30 can also be indirectly regulated by a redox-dependent mechanism, probably through FTT-2 oxidation.
Collapse
Affiliation(s)
- Hildegard Colino-Lage
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - David Guerrero-Gómez
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eva Gómez-Orte
- Centro de Investigación Biomédica de la Rioja (CIBIR), Logroño, La Rioja, Spain
| | - Xavier González
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tobias B Dansen
- Center for Molecular Medicine, University Medical Center Utrecht, CG Utrecht, the Netherlands
| | - Cristina Ayuso
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Seville, Spain
| | - Peter Askjaer
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Seville, Spain
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Javier E Irazoqui
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Juan Cabello
- Centro de Investigación Biomédica de la Rioja (CIBIR), Logroño, La Rioja, Spain.
| | - Antonio Miranda-Vizuete
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
32
|
de Boer AH. The fusicoccin story revisited. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:5531-5546. [PMID: 38989653 DOI: 10.1093/jxb/erae300] [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: 04/02/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Fusicoccin (FC) is one of the most studied fungal metabolites to date. The finding that the plasma membrane H+-ATPase in combination with 14-3-3 proteins acts as a high-affinity receptor for FC was a breakthrough in the field. Ever since, the binding of FC to the ATPase-14-3-3 receptor complex has taken center stage in explaining all FC-induced physiological effects. However, a more critical review shows that this is not evident for a number of FC-induced effects. This review challenges the notion that all FC-affected processes start with the binding to and activation of the plasma membrane ATPase, and raises the question of whether other proteins with a key role in the respective processes are directly targeted by FC. A second unresolved question is whether FC may be another example of a fungal molecule turning out to be a 'copy' of an as yet unknown plant molecule. In view of the evidence, albeit not conclusive, that plants indeed produce 'FC-like ligands', it is worthwhile making a renewed attempt with modern improved technology to answer this question; the answer might upgrade FC or its structural analogue(s) to the classification of plant hormone.
Collapse
Affiliation(s)
- Albertus H de Boer
- Division of Medicinal Chemistry, Institute of Molecular & Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Obsilova V, Obsil T. Look for the Scaffold: Multifaceted Regulation of Enzyme Activity by 14-3-3 Proteins. Physiol Res 2024; 73:S401-S412. [PMID: 38647170 PMCID: PMC11412345 DOI: 10.33549/physiolres.935306] [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: 09/04/2024] Open
Abstract
Enzyme activity is regulated by several mechanisms, including phosphorylation. Phosphorylation is a key signal transduction process in all eukaryotic cells and is thus crucial for virtually all cellular processes. In addition to its direct effect on protein structure, phosphorylation also affects protein-protein interactions, such as binding to scaffolding 14-3-3 proteins, which selectively recognize phosphorylated motifs. These interactions then modulate the catalytic activity, cellular localisation and interactions of phosphorylated enzymes through different mechanisms. The aim of this mini-review is to highlight several examples of 14-3-3 protein-dependent mechanisms of enzyme regulation previously studied in our laboratory over the past decade. More specifically, we address here the regulation of the human enzymes ubiquitin ligase Nedd4-2, procaspase-2, calcium-calmodulin dependent kinases CaMKK1/2, and death-associated protein kinase 2 (DAPK2) and yeast neutral trehalase Nth1.
Collapse
Affiliation(s)
- V Obsilova
- Institute of Physiology of the Czech Academy of Sciences, Laboratory of Structural Biology of Signaling Proteins, Division BIOCEV, Vestec, Czech Republic. or
| | | |
Collapse
|
34
|
Ru K, Cui L, Wu C, Tan XX, An WT, Wu Q, Ma YT, Hao Y, Xiao X, Bai J, Liu X, Xia XF, Zhao MQ. Exploring the molecular and immune landscape of cellular senescence in lung adenocarcinoma. Front Immunol 2024; 15:1347770. [PMID: 39267750 PMCID: PMC11390420 DOI: 10.3389/fimmu.2024.1347770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction The connection between aging and cancer is complex. Previous research has highlighted the association between the aging process of lung adenocarcinoma (LUAD) cells and the immune response, yet there remains a gap in confirming this through single-cell data validation. Here, we aim to develop a novel aging-related prognostic model for LUAD, and verify the alterations in the genome and immune microenvironment linked to cellular senescence. Methods We integrated a comprehensive collection of senescence genes from the GenAge and CellAge databases and employed the least absolute shrinkage and selection operator (LASSO) Cox analysis to construct and validate a novel prognostic model for LUAD. This model was then utilized to examine the relationship between aging, tumor somatic mutations, and immune cell infiltration. Additionally, we explored the heterogeneity of senescence and intercellular communication within the LUAD tumor microenvironment (TME) through single-cell transcriptomic data analysis. Results By exploring the expression profiles of 586 cellular senescence-related genes in 428 LUAD patients, we constructed an aging-related genes (ARGs) risk model included 10 ARGs and validated it as an independent prognostic predictor for LUAD patients. Notably, patients with low aging scores (LAS group) exhibited better survival, lower tumor mutation burden (TMB), lower somatic mutation frequency, lower tumor proliferation rate, and an immune activated phenotype compared to patients with high aging scores (HAS group). While the HAS group was enriched in tumor cells and showed a lower infiltration of CD8-CCR7, CD8- CXCL13, CD8-GNLY, FCGR3A NK cells, XCL1 NK cells, plasma cell (PC) and other immune subsets. Furthermore, the SPP1 and TENASCIN pathways, associated with tumor immune escape and tumor progression, were also enriched in the HAS group. Additionally, our study also indicated that senescence levels were heterogeneous in the LUAD tumor microenvironment (TME), especially with tumor cells in the LAS group showing higher age scores compared to those in the HAS group. Conclusions Collectively, our findings underscore that ARRS through ARGs serves as a robust biomarker for the prognosis in LUAD.
Collapse
Affiliation(s)
- Kun Ru
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Cui
- Geneplus-Beijing Institute, Beijing, China
| | - Cong Wu
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xin X Tan
- Geneplus-Shenzhen Clinical Laboratory, Shenzhen, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Wen T An
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiang Wu
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, China
| | - Yu T Ma
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yu Hao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao Xiao
- Geneplus-Shenzhen Institute, Shenzhen, China
| | - Jing Bai
- Geneplus-Beijing Institute, Beijing, China
| | - Xiang Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xue F Xia
- Geneplus-Beijing Institute, Beijing, China
| | - Miao Q Zhao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
35
|
Spooner HC, Costa AD, González AH, Ibrahimkhail H, Yarov-Yarovoy V, Horne M, Dickson EJ, Dixon RE. 14-3-3 promotes sarcolemmal expression of cardiac Ca V 1.2 and nucleates isoproterenol-triggered channel super-clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.607987. [PMID: 39229175 PMCID: PMC11370440 DOI: 10.1101/2024.08.16.607987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The L-type Ca 2+ channel (Ca V 1.2) is essential for cardiac excitation-contraction coupling. To contribute to the inward Ca 2+ flux that drives Ca 2+ -induced-Ca 2+ -release, Ca V 1.2 channels must be expressed on the sarcolemma; thus the regulatory mechanisms that tune Ca V 1.2 expression to meet contractile demand are an emerging area of research. A ubiquitously expressed protein called 14-3-3 has been proposed to affect Ca 2+ channel trafficking in non-myocytes, however whether 14-3-3 has similar effects on Ca V 1.2 in cardiomyocytes is unknown. 14-3-3 preferentially binds phospho-serine/threonine residues to affect many cellular processes and is known to regulate cardiac ion channels including Na V 1.5 and hERG. Altered 14-3-3 expression and function have been implicated in cardiac pathologies including hypertrophy. Accordingly, we tested the hypothesis that 14-3-3 interacts with Ca V 1.2 in a phosphorylation-dependent manner and regulates cardiac Ca V 1.2 trafficking and recycling. Confocal imaging, proximity ligation assays, super-resolution imaging, and co-immunoprecipitation revealed a population of 14-3-3 colocalized and closely associated with Ca V 1.2. The degree of 14-3-3/Ca V 1.2 colocalization increased upon stimulation of β -adrenergic receptors with isoproterenol. Notably, only the 14-3-3-associated Ca V 1.2 population displayed increased cluster size with isoproterenol, revealing a role for 14-3-3 as a nucleation factor that directs Ca V 1.2 super-clustering. 14-3-3 overexpression increased basal Ca V 1.2 cluster size and Ca 2+ currents in ventricular myocytes, with maintained channel responsivity to isoproterenol. In contrast, isoproterenol-stimulated augmentation of sarcolemmal Ca V 1.2 expression and currents in ventricular myocytes were abrogated by 14-3-3 inhibition. These data support a model where 14-3-3 interacts with Ca V 1.2 in a phosphorylation-dependent manner to promote enhanced trafficking/recycling, clustering, and activity during β -adrenergic stimulation. Significance Statement The L-type Ca 2+ channel, Ca V 1.2, plays an essential role in excitation-contraction coupling in the heart and in part regulates the overall strength of contraction during basal and fight- or-flight β -adrenergic signaling conditions. Proteins that modulate the trafficking and/or activity of Ca V 1.2 are interesting both from a physiological and pathological perspective, since alterations in Ca V 1.2 can impact action potential duration and cause arrythmias. A small protein called 14-3-3 regulates other ion channels in the heart and other Ca 2+ channels, but how it may interact with Ca V 1.2 in the heart has never been studied. Examining factors that affect Ca V 1.2 at rest and during β -adrenergic stimulation is crucial for our ability to understand and treat disease and aging conditions where these pathways are altered.
Collapse
|
36
|
Abstract
RAF family protein kinases are a key node in the RAS/RAF/MAP kinase pathway, the signaling cascade that controls cellular proliferation, differentiation, and survival in response to engagement of growth factor receptors on the cell surface. Over the past few years, structural and biochemical studies have provided new understanding of RAF autoregulation, RAF activation by RAS and the SHOC2 phosphatase complex, and RAF engagement with HSP90-CDC37 chaperone complexes. These studies have important implications for pharmacologic targeting of the pathway. They reveal RAF in distinct regulatory states and show that the functional RAF switch is an integrated complex of RAF with its substrate (MEK) and a 14-3-3 dimer. Here we review these advances, placing them in the context of decades of investigation of RAF regulation. We explore the insights they provide into aberrant activation of the pathway in cancer and RASopathies (developmental syndromes caused by germline mutations in components of the pathway).
Collapse
Affiliation(s)
- Hyesung Jeon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Emre Tkacik
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
- Systems, Synthetic, and Quantitative Biology PhD Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael J Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Somsen BA, Cossar PJ, Arkin MR, Brunsveld L, Ottmann C. 14-3-3 Protein-Protein Interactions: From Mechanistic Understanding to Their Small-Molecule Stabilization. Chembiochem 2024; 25:e202400214. [PMID: 38738787 DOI: 10.1002/cbic.202400214] [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: 03/08/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/14/2024]
Abstract
Protein-protein interactions (PPIs) are of utmost importance for maintenance of cellular homeostasis. Herein, a central role can be found for 14-3-3 proteins. These hub-proteins are known to bind hundreds of interaction partners, thereby regulating their activity, localization, and/or stabilization. Due to their ability to bind a large variety of client proteins, studies of 14-3-3 protein complexes flourished over the last decades, aiming to gain greater molecular understanding of these complexes and their role in health and disease. Because of their crucial role within the cell, 14-3-3 protein complexes are recognized as highly interesting therapeutic targets, encouraging the discovery of small molecule modulators of these PPIs. We discuss various examples of 14-3-3-mediated regulation of its binding partners on a mechanistic level, highlighting the versatile and multi-functional role of 14-3-3 within the cell. Furthermore, an overview is given on the development of stabilizers of 14-3-3 protein complexes, from initially used natural products to fragment-based approaches. These studies show the potential of 14-3-3 PPI stabilizers as novel agents in drug discovery and as tool compounds to gain greater molecular understanding of the role of 14-3-3-based protein regulation.
Collapse
Affiliation(s)
- Bente A Somsen
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| | - Peter J Cossar
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California, 94143, United States
| | - Luc Brunsveld
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| | - Christian Ottmann
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| |
Collapse
|
38
|
Sinha NK, McKenney C, Yeow ZY, Li JJ, Nam KH, Yaron-Barir TM, Johnson JL, Huntsman EM, Cantley LC, Ordureau A, Regot S, Green R. The ribotoxic stress response drives UV-mediated cell death. Cell 2024; 187:3652-3670.e40. [PMID: 38843833 PMCID: PMC11246228 DOI: 10.1016/j.cell.2024.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/03/2024] [Accepted: 05/09/2024] [Indexed: 06/13/2024]
Abstract
While ultraviolet (UV) radiation damages DNA, eliciting the DNA damage response (DDR), it also damages RNA, triggering transcriptome-wide ribosomal collisions and eliciting a ribotoxic stress response (RSR). However, the relative contributions, timing, and regulation of these pathways in determining cell fate is unclear. Here we use time-resolved phosphoproteomic, chemical-genetic, single-cell imaging, and biochemical approaches to create a chronological atlas of signaling events activated in cells responding to UV damage. We discover that UV-induced apoptosis is mediated by the RSR kinase ZAK and not through the DDR. We identify two negative-feedback modules that regulate ZAK-mediated apoptosis: (1) GCN2 activation limits ribosomal collisions and attenuates ZAK-mediated RSR and (2) ZAK activity leads to phosphodegron autophosphorylation and its subsequent degradation. These events tune ZAK's activity to collision levels to establish regimes of homeostasis, tolerance, and death, revealing its key role as the cellular sentinel for nucleic acid damage.
Collapse
Affiliation(s)
- Niladri K Sinha
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Connor McKenney
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeffrey J Li
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ki Hong Nam
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Emily M Huntsman
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Sergi Regot
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Rachel Green
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
39
|
Nishiyama K, Aihara Y, Suzuki T, Takahashi K, Kinoshita T, Dohmae N, Sato A, Hagihara S. Discovery of a Plant 14-3-3 Inhibitor Possessing Isoform Selectivity and In Planta Activity. Angew Chem Int Ed Engl 2024; 63:e202400218. [PMID: 38658314 DOI: 10.1002/anie.202400218] [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: 01/04/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Synthetic modulators of plant 14-3-3s are promising chemical tools both for understanding the 14-3-3-related signaling pathways and controlling plant physiology. Herein, we describe a novel small-molecule inhibitor for 14-3-3 proteins of Arabidopsis thaliana. The inhibitor was identified from unexpected products in a stock solution in dimethyl sulfoxide (DMSO) of an in-house chemical library. Mass spectroscopy, mutant-based analyses, fluorescence polarization assays, and thermal shift assays revealed that the inhibitor covalently binds to an allosteric site of 14-3-3 with isoform selectivity. Moreover, infiltration of the inhibitor to Arabidopsis leaves suppressed the stomatal aperture. The inhibitor should provide new insight into the design of potent and isoform-selective 14-3-3 modulators.
Collapse
Affiliation(s)
- Kotaro Nishiyama
- Center for Sustainable Resource Science, RIKEN, 2-1 Hirosawa, Wako City, Saitama, 351-0198, Japan
| | - Yusuke Aihara
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University Chikusa, Nagoya, 464-8602, Japan
- PRESTO, Japan Science and Technology Agency (JST), Honcho 4-1-8, Kawaguchi, Saitama, 332-0012, Japan
| | - Takehiro Suzuki
- Center for Sustainable Resource Science, RIKEN, 2-1 Hirosawa, Wako City, Saitama, 351-0198, Japan
| | - Koji Takahashi
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University Chikusa, Nagoya, 464-8602, Japan
- Graduate School of Science, Nagoya University Chikusa, Nagoya, 464-8602, Japan
| | - Toshinori Kinoshita
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University Chikusa, Nagoya, 464-8602, Japan
- Graduate School of Science, Nagoya University Chikusa, Nagoya, 464-8602, Japan
| | - Naoshi Dohmae
- Center for Sustainable Resource Science, RIKEN, 2-1 Hirosawa, Wako City, Saitama, 351-0198, Japan
| | - Ayato Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University Chikusa, Nagoya, 464-8602, Japan
| | - Shinya Hagihara
- Center for Sustainable Resource Science, RIKEN, 2-1 Hirosawa, Wako City, Saitama, 351-0198, Japan
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University Chikusa, Nagoya, 464-8602, Japan
| |
Collapse
|
40
|
Jiang Y, Chen M, Xu N, Li Z, Li X, Yu H, Sun J, Wang A, Huang Y, Wang L. Adaptor protein 14-3-3zeta promotes corneal wound healing via regulating cell homeostasis, a potential novel therapy for corneal injury. Exp Eye Res 2024; 244:109948. [PMID: 38815790 DOI: 10.1016/j.exer.2024.109948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Severe corneal injury can lead to blindness even after prompt treatment. 14-3-3zeta, a member of an adaptor protein family, contributes to tissue repair by enhancing cellular viability and inhibiting fibrosis and inflammation in renal disease or arthritis. However, its role in corneal regeneration is less studied. In this study, filter disc of 2-mm diameter soaked in sodium hydroxide with a concentration of 0.5 N was placed at the center of the cornea for 30 s to establish a mouse model of corneal alkali injury. We found that 14-3-3zeta, which is mainly expressed in the epithelial layer, was upregulated following injury. Overexpression of 14-3-3zeta in ocular tissues via adeno-associated virus-mediated subconjunctival delivery promoted corneal wound healing, showing improved corneal structure and transparency. In vitro studies on human corneal epithelial cells showed that 14-3-3zeta was critical for cell proliferation and migration. mRNA-sequencing in conjunction with KEGG analysis and validation experiments revealed that 14-3-3zeta regulated the mRNA levels of ITGB1, PIK3R1, FGF5, PRKAA1 and the phosphorylation level of Akt, suggesting the involvement of the PI3K-Akt pathway in 14-3-3zeta-mediated tissue repair. 14-3-3zeta is a potential novel therapeutic candidate for treating severe corneal injury.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- 14-3-3 Proteins/metabolism
- 14-3-3 Proteins/genetics
- 14-3-3 Proteins/biosynthesis
- Blotting, Western
- Burns, Chemical/metabolism
- Burns, Chemical/pathology
- Burns, Chemical/drug therapy
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Corneal Injuries/metabolism
- Corneal Injuries/pathology
- Corneal Injuries/genetics
- Disease Models, Animal
- Epithelium, Corneal/metabolism
- Epithelium, Corneal/drug effects
- Epithelium, Corneal/injuries
- Eye Burns/chemically induced
- Gene Expression Regulation
- Homeostasis
- Mice, Inbred C57BL
- Sodium Hydroxide
- Wound Healing/drug effects
- Wound Healing/physiology
Collapse
Affiliation(s)
- Yilin Jiang
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Mingxiong Chen
- School of Medicine, Nankai University, Tianjin, 300071, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Ning Xu
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Zongyuan Li
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Xiaoqi Li
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Hanrui Yu
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Jiaying Sun
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - An Wang
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China
| | - Yifei Huang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Liqiang Wang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China; School of Medicine, Nankai University, Tianjin, 300071, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100089, China.
| |
Collapse
|
41
|
Kapitonova AA, Perfilova KV, Cooley RB, Sluchanko NN. Phosphorylation Code of Human Nucleophosmin Includes Four Cryptic Sites for Hierarchical Binding of 14-3-3 Proteins. J Mol Biol 2024; 436:168592. [PMID: 38702038 DOI: 10.1016/j.jmb.2024.168592] [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: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Nucleophosmin (NPM1) is the 46th most abundant human protein with many functions whose dysregulation leads to various cancers. Pentameric NPM1 resides in the nucleolus but can also shuttle to the cytosol. NPM1 is regulated by multisite phosphorylation, yet molecular consequences of site-specific NPM1 phosphorylation remain elusive. Here we identify four 14-3-3 protein binding sites in NPM1 concealed within its oligomerization and α-helical C-terminal domains that are found phosphorylated in vivo. By combining mutagenesis, in-cell phosphorylation and PermaPhos technology for site-directed incorporation of a non-hydrolyzable phosphoserine mimic, we show how phosphorylation promotes NPM1 monomerization and partial unfolding, to recruit 14-3-3 dimers with low-micromolar affinity. Using fluorescence anisotropy we quantified pairwise interactions of all seven human 14-3-3 isoforms with four recombinant NPM1 phosphopeptides and assessed their druggability by fusicoccin. This revealed a complex hierarchy of 14-3-3 affinities toward the primary (S48, S293) and secondary (S106, S260) sites, differentially modulated by the small molecule. As three of these 14-3-3 binding phosphosites in NPM1 reside within signal sequences, this work suggests a mechanism of NPM1 regulation by which NPM1 phosphorylation can promote 14-3-3 binding to affect NPM1 shuttling between cell compartments. It also provides further evidence that phosphorylation-induced structural rearrangements of globular proteins serve to expose otherwise cryptic 14-3-3-binding sites that are important for cellular function.
Collapse
Affiliation(s)
- Anna A Kapitonova
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Kristina V Perfilova
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Richard B Cooley
- GCE4All Center, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia.
| |
Collapse
|
42
|
Crha R, Kozeleková A, Hofrová A, Iľkovičová L, Gašparik N, Kadeřávek P, Hritz J. Hiding in plain sight: Complex interaction patterns between Tau and 14-3-3ζ protein variants. Int J Biol Macromol 2024; 266:130802. [PMID: 38492709 DOI: 10.1016/j.ijbiomac.2024.130802] [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: 12/22/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Tau protein is an intrinsically disordered protein that plays a key role in Alzheimer's disease (AD). In brains of AD patients, Tau occurs abnormally phosphorylated and aggregated in neurofibrillary tangles (NFTs). Together with Tau, 14-3-3 proteins - abundant cytosolic dimeric proteins - were found colocalized in the NFTs. However, so far, the molecular mechanism of the process leading to pathological changes in Tau structure as well as the direct involvement of 14-3-3 proteins are not well understood. Here, we aimed to reveal the effects of phosphorylation by protein kinase A (PKA) on Tau structural preferences and provide better insight into the interaction between Tau and 14-3-3 proteins. We also addressed the impact of monomerization-inducing phosphorylation of 14-3-3 at S58 on the binding to Tau protein. Using multidimensional nuclear magnetic resonance spectroscopy (NMR), chemical cross-linking analyzed by mass spectrometry (MS) and PAGE, we unveiled differences in their binding affinity, stoichiometry, and interfaces with single-residue resolution. We revealed that the interaction between 14-3-3 and Tau proteins is mediated not only via the 14-3-3 amphipathic binding grooves, but also via less specific interactions with 14-3-3 protein surface and, in the case of monomeric 14-3-3, also partially via the exposed dimeric interface. In addition, the hyperphosphorylation of Tau changes its affinity to 14-3-3 proteins. In conclusion, we propose quite complex interaction mode between the Tau and 14-3-3 proteins.
Collapse
Affiliation(s)
- Radek Crha
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Aneta Kozeleková
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Alena Hofrová
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Lucia Iľkovičová
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Norbert Gašparik
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Pavel Kadeřávek
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jozef Hritz
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| |
Collapse
|
43
|
Kamayirese S, Maity S, Hansen LA, Lovas S. The Development of CDC25A-Derived Phosphoseryl Peptides That Bind 14-3-3ε with High Affinities. Int J Mol Sci 2024; 25:4918. [PMID: 38732131 PMCID: PMC11084659 DOI: 10.3390/ijms25094918] [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: 03/19/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Overexpression of the 14-3-3ε protein is associated with suppression of apoptosis in cutaneous squamous cell carcinoma (cSCC). This antiapoptotic activity of 14-3-3ε is dependent on its binding to CDC25A; thus, inhibiting 14-3-3ε - CDC25A interaction is an attractive therapeutic approach to promote apoptosis in cSCC. In this regard, designing peptide inhibitors of 14-3-3ε - CDC25A interactions is of great interest. This work reports the rational design of peptide analogs of pS, a CDC25A-derived peptide that has been shown to inhibit 14-3-3ε-CDC25A interaction and promote apoptosis in cSCC with micromolar IC50. We designed new peptide analogs in silico by shortening the parent pS peptide from 14 to 9 amino acid residues; then, based on binding motifs of 14-3-3 proteins, we introduced modifications in the pS(174-182) peptide. We studied the binding of the peptides using conventional molecular dynamics (MD) and steered MD simulations, as well as biophysical methods. Our results showed that shortening the pS peptide from 14 to 9 amino acids reduced the affinity of the peptide. However, substituting Gln176 with either Phe or Tyr amino acids rescued the binding of the peptide. The optimized peptides obtained in this work can be candidates for inhibition of 14-3-3ε - CDC25A interactions in cSCC.
Collapse
Affiliation(s)
| | | | | | - Sándor Lovas
- Department of Biomedical Sciences, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
44
|
Simon JJ, Fowler DM, Maly DJ. Multiplexed, multimodal profiling of the intracellular activity, interactions, and druggability of protein variants using LABEL-seq. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590094. [PMID: 38659825 PMCID: PMC11042325 DOI: 10.1101/2024.04.19.590094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Multiplexed assays of variant effect are powerful tools for assessing the impact of protein sequence variation, but are limited to measuring a single protein property and often rely on indirect readouts of intracellular protein function. Here, we developed LAbeling with Barcodes and Enrichment for biochemicaL analysis by sequencing (LABEL-seq), a platform for the multimodal profiling of thousands of protein variants in cultured human cells. Multimodal measurement of ~20,000 variant effects for ~1,600 BRaf variants using LABEL-seq revealed that variation at positions that are frequently mutated in cancer had minimal effects on folding and intracellular abundance but could dramatically alter activity, protein-protein interactions, and druggability. Integrative analysis of our multimodal measurements identified networks of positions with similar roles in regulating BRaf's signaling properties and enabled predictive modeling of variant effects on complex processes such as cell proliferation and small molecule-promoted degradation. LABEL-seq provides a scalable approach for the direct measurement of multiple biochemical effects of protein variants in their native cellular context, yielding insight into protein function, disease mechanisms, and druggability.
Collapse
Affiliation(s)
- Jessica J Simon
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
- Co-corresponding authors: ,
| | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, University of Washington, Seattle, WA, United States
- Co-corresponding authors: ,
| |
Collapse
|
45
|
Ke YD, van Hummel A, Au C, Chan G, Lee WS, van der Hoven J, Przybyla M, Deng Y, Sabale M, Morey N, Bertz J, Feiten A, Ippati S, Stevens CH, Yang S, Gladbach A, Haass NK, Kril JJ, Blair IP, Delerue F, Ittner LM. Targeting 14-3-3θ-mediated TDP-43 pathology in amyotrophic lateral sclerosis and frontotemporal dementia mice. Neuron 2024; 112:1249-1264.e8. [PMID: 38366598 DOI: 10.1016/j.neuron.2024.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/20/2023] [Accepted: 01/22/2024] [Indexed: 02/18/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are characterized by cytoplasmic deposition of the nuclear TAR-binding protein 43 (TDP-43). Although cytoplasmic re-localization of TDP-43 is a key event in the pathogenesis of ALS/FTD, the underlying mechanisms remain unknown. Here, we identified a non-canonical interaction between 14-3-3θ and TDP-43, which regulates nuclear-cytoplasmic shuttling. Neuronal 14-3-3θ levels were increased in sporadic ALS and FTD with TDP-43 pathology. Pathogenic TDP-43 showed increased interaction with 14-3-3θ, resulting in cytoplasmic accumulation, insolubility, phosphorylation, and fragmentation of TDP-43, resembling pathological changes in disease. Harnessing this increased affinity of 14-3-3θ for pathogenic TDP-43, we devised a gene therapy vector targeting TDP-43 pathology, which mitigated functional deficits and neurodegeneration in different ALS/FTD mouse models expressing mutant or non-mutant TDP-43, including when already symptomatic at the time of treatment. Our study identified 14-3-3θ as a mediator of cytoplasmic TDP-43 localization with implications for ALS/FTD pathogenesis and therapy.
Collapse
Affiliation(s)
- Yazi D Ke
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Annika van Hummel
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Carol Au
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Gabriella Chan
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Wei Siang Lee
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Julia van der Hoven
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Magdalena Przybyla
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Yuanyuan Deng
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Miheer Sabale
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Nicolle Morey
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Josefine Bertz
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Astrid Feiten
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Stefania Ippati
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Claire H Stevens
- School of Chemistry and Molecular Bioscience, University of Wollongong and Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Shu Yang
- Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Amadeus Gladbach
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Nikolas K Haass
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Jillian J Kril
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
| | - Ian P Blair
- Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Fabien Delerue
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
46
|
Rrustemi T, Meyer K, Roske Y, Uyar B, Akalin A, Imami K, Ishihama Y, Daumke O, Selbach M. Pathogenic mutations of human phosphorylation sites affect protein-protein interactions. Nat Commun 2024; 15:3146. [PMID: 38605029 PMCID: PMC11009412 DOI: 10.1038/s41467-024-46794-8] [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: 08/09/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Despite their lack of a defined 3D structure, intrinsically disordered regions (IDRs) of proteins play important biological roles. Many IDRs contain short linear motifs (SLiMs) that mediate protein-protein interactions (PPIs), which can be regulated by post-translational modifications like phosphorylation. 20% of pathogenic missense mutations are found in IDRs, and understanding how such mutations affect PPIs is essential for unraveling disease mechanisms. Here, we employ peptide-based interaction proteomics to investigate 36 disease-associated mutations affecting phosphorylation sites. Our results unveil significant differences in interactomes between phosphorylated and non-phosphorylated peptides, often due to disrupted phosphorylation-dependent SLiMs. We focused on a mutation of a serine phosphorylation site in the transcription factor GATAD1, which causes dilated cardiomyopathy. We find that this phosphorylation site mediates interaction with 14-3-3 family proteins. Follow-up experiments reveal the structural basis of this interaction and suggest that 14-3-3 binding affects GATAD1 nucleocytoplasmic transport by masking a nuclear localisation signal. Our results demonstrate that pathogenic mutations of human phosphorylation sites can significantly impact protein-protein interactions, offering insights into potential molecular mechanisms underlying pathogenesis.
Collapse
Affiliation(s)
| | - Katrina Meyer
- Max Delbrück Center (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195, Berlin, Germany
| | - Yvette Roske
- Max Delbrück Center (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Bora Uyar
- Max Delbrück Center (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Altuna Akalin
- Max Delbrück Center (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Koshi Imami
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Kanagawa, Japan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Oliver Daumke
- Max Delbrück Center (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustraße 6, Berlin, Germany
| | - Matthias Selbach
- Max Delbrück Center (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
47
|
Chen Y, Yao Z, Zhao L, Yu M, Chen B, Zou C. Redundant and Distinct Roles of Two 14-3-3 Proteins in Fusarium sacchari, Pathogen of Sugarcane Pokkah Boeng Disease. J Fungi (Basel) 2024; 10:257. [PMID: 38667928 PMCID: PMC11051555 DOI: 10.3390/jof10040257] [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: 02/08/2024] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Fusarium sacchari, a key pathogen of sugarcane, is responsible for the Pokkah boeng disease (PBD) in China. The 14-3-3 proteins have been implicated in critical developmental processes, including dimorphic transition, signal transduction, and carbon metabolism in various phytopathogenic fungi. However, their roles are poorly understood in F. sacchari. This study focused on the characterization of two 14-3-3 protein-encoding genes, FsBmh1 and FsBmh2, within F. sacchari. Both genes were found to be expressed during the vegetative growth stage, yet FsBmh1 was repressed at the sporulation stage in vitro. To elucidate the functions of these genes, the deletion mutants ΔFsBmh1 and ΔFsBmh2 were generated. The ΔFsBmh2 exhibited more pronounced phenotypic defects, such as impaired hyphal branching, septation, conidiation, spore germination, and colony growth, compared to the ΔFsBmh1. Notably, both knockout mutants showed a reduction in virulence, with transcriptome analysis revealing changes associated with the observed phenotypes. To further investigate the functional interplay between FsBmh1 and FsBmh2, we constructed and analyzed mutants with combined deletion and silencing (ΔFsBmh/siFsBmh) as well as overexpression (O-FsBmh). The combinations of ΔFsBmh1/siFsBmh2 or ΔFsBmh2/siFsBmh1 displayed more severe phenotypes than those with single allele deletions, suggesting a functional redundancy between the two 14-3-3 proteins. Yeast two-hybrid (Y2H) assays identified 20 proteins with pivotal roles in primary metabolism or diverse biological functions, 12 of which interacted with both FsBmh1 and FsBmh2. Three proteins were specifically associated with FsBmh1, while five interacted exclusively with FsBmh2. In summary, this research provides novel insights into the roles of FsBmh1 and FsBmh2 in F. sacchari and highlights potential targets for PBD management through the modulation of FsBmh functions.
Collapse
Affiliation(s)
- Yuejia Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry & Province Co-Sponsored Center of Collaborative Innovation for Sugarcane Industry, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.C.); (M.Y.)
| | - Ziting Yao
- Plant Protection Research Institute, Guangxi Academy of Agriculture Science, Nanning 530007, China;
| | - Lixian Zhao
- Guangxi Key Laboratory of Sugarcane Biology, College of Agriculture, Guangxi University, Nanning 530004, China;
| | - Mei Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry & Province Co-Sponsored Center of Collaborative Innovation for Sugarcane Industry, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.C.); (M.Y.)
| | - Baoshan Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry & Province Co-Sponsored Center of Collaborative Innovation for Sugarcane Industry, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.C.); (M.Y.)
- Guangxi Key Laboratory of Sugarcane Biology, College of Agriculture, Guangxi University, Nanning 530004, China;
| | - Chengwu Zou
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry & Province Co-Sponsored Center of Collaborative Innovation for Sugarcane Industry, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.C.); (M.Y.)
- Guangxi Key Laboratory of Sugarcane Biology, College of Agriculture, Guangxi University, Nanning 530004, China;
| |
Collapse
|
48
|
Wang Y, Cao Y, Chen Y, Cheng H, Liu Z, Wang M, Feng Y, Fei B, Cui K, Huang Z. YWHAG promotes colorectal cancer progression by regulating the CTTN-Wnt/β-catenin signaling axis. Med Oncol 2024; 41:100. [PMID: 38538804 DOI: 10.1007/s12032-024-02349-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: 12/07/2023] [Accepted: 02/27/2024] [Indexed: 05/31/2024]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer type globally. Nevertheless, the fundamental mechanisms driving CRC progression remain ambiguous, and the prognosis for the majority of patients diagnosed at an advanced stage is dismal. YWHA/14-3-3 proteins serve as central nodes in several signaling pathways and are closely related to tumorigenesis and progression. However, their exact roles in CRC are still poorly elucidated. In this study, we revealed that YWHAG was the most significantly upregulated member of the YWHA/14-3-3 family in CRC tissues and was associated with a poor prognosis. Subsequent phenotypic experiments showed that YWHAG promoted the proliferation, migration, and invasion of CRC cells. Mechanistically, RNA-seq data showed that multiple signaling pathways, including Wnt and epithelial-mesenchymal transition, were potentially regulated by YWHAG. CTTN was identified as a YWHAG-associated protein, and mediated its tumor-promoting functions by activating the Wnt/β-catenin signaling in CRC cells. In summary, our data indicate that YWHAG facilitates the proliferation, migration, and invasion of CRC cells by modulating the CTTN-Wnt/β-catenin signaling pathway, which offers a novel perspective for the treatment of CRC.
Collapse
Affiliation(s)
- Yuanben Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Ying Chen
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Han Cheng
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhiang Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Mengna Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yuyang Feng
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Bojian Fei
- Department of Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, 1000 He Feng Road, Wuxi, 214122, Jiangsu, China
| | - Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
49
|
Kapitonova AA, Perfilova KV, Cooley RB, Sluchanko NN. Phosphorylation code of human nucleophosmin includes four cryptic sites for hierarchical binding of 14-3-3 proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580064. [PMID: 38405961 PMCID: PMC10888825 DOI: 10.1101/2024.02.13.580064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Nucleophosmin (NPM1) is the 46th most abundant human protein with many functions whose dysregulation leads to various cancers. Pentameric NPM1 resides in the nucleolus but can also shuttle to the cytosol. NPM1 is regulated by multisite phosphorylation, yet molecular consequences of site-specific NPM1 phosphorylation remain elusive. Here we identify four 14-3-3 protein binding sites in NPM1 concealed within its oligomerization and α-helical C-terminal domains that are found phosphorylated in vivo. By combining mutagenesis, in-cell phosphorylation and PermaPhos technology for site-directed incorporation of a non-hydrolyzable phosphoserine mimic, we show how phosphorylation promotes NPM1 monomerization and partial unfolding, to recruit 14-3-3 dimers with low-micromolar affinity. Using fluorescence anisotropy we quantified pairwise interactions of all seven human 14-3-3 isoforms with four recombinant NPM1 phosphopeptides and assessed their druggability by fusicoccin. This revealed a complex hierarchy of 14-3-3 affinities toward the primary (S48, S293) and secondary (S106, S260) sites, differentially modulated by the small molecule. As three of these 14-3-3 binding phospho-sites in NPM1 reside within signal sequences, this work highlights a key mechanism of NPM1 regulation by which NPM1 phosphorylation promotes 14-3-3 binding to control nucleocytoplasmic shuttling. It also provides further evidence that phosphorylation-induced structural rearrangements of globular proteins serve to expose otherwise cryptic 14-3-3-binding sites that are important for cellular function.
Collapse
Affiliation(s)
- Anna A. Kapitonova
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Kristina V. Perfilova
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Richard B. Cooley
- GCE4All Center, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, 97331, USA
| | - Nikolai N. Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| |
Collapse
|
50
|
Liu Z, Lyu X, Chen J, Zhang B, Xie S, Yuan Y, Sun L, Yuan S, Yu H, Ding J, Yang M. Arnicolide C Suppresses Tumor Progression by Targeting 14-3-3θ in Breast Cancer. Pharmaceuticals (Basel) 2024; 17:224. [PMID: 38399439 PMCID: PMC10892132 DOI: 10.3390/ph17020224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Background: Arnicolide C, which is isolated from Centipeda minima, has excellent antitumor effects. However, the potential impacts and related mechanisms of action of arnicolide C in breast cancer remain unknown. Methods: The viability of breast cancer cells was measured using MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and colony formation assays. For analysis of apoptosis and the cell cycle, flow cytometry was used. A molecular docking approach was used to explore the possible targets of arnicolide C. Western blot analysis was used to detect changes in the expression of 14-3-3θ and proteins in related pathways after arnicolide C treatment in breast cancer cells. The anti-breast cancer effect of arnicolide C in vivo was evaluated by establishing cell-derived xenograft (CDX) and patient-derived xenograft (PDX) models. Results: Arnicolide C inhibited proliferation, increased apoptosis, and induced G1 arrest. In particular, molecular docking analysis indicated that arnicolide C binds to 14-3-3θ. Arnicolide C reduced 14-3-3θ expression and inhibited its downstream signaling pathways linked to cell proliferation. Similar results were obtained in the CDX and PDX models. Conclusion: Arnicolide C can have an anti-breast cancer effect both in vitro and in vivo and can induce cell cycle arrest and increase apoptosis in vitro. The molecular mechanism may be related to the effect of arnicolide C on the expression level of 14-3-3θ. However, the specific mechanism through which arnicolide C affects 14-3-3θ protein expression still needs to be determined.
Collapse
Affiliation(s)
- Zhengrui Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaodan Lyu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jiaxu Chen
- College of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Benteng Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Siman Xie
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Yuan
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Jian Ding
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mei Yang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|