1
|
Choi S, Ahn S, Cho KH, Lee SK, Kee JM. Chemoproteomic identification of phosphohistidine acceptors: posttranslational activity regulation of a key glycolytic enzyme. Chem Sci 2025; 16:8014-8022. [PMID: 40201162 PMCID: PMC11974560 DOI: 10.1039/d5sc01024a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
Histidine phosphorylation, an unconventional and understudied posttranslational modification, often involves phosphohistidine (pHis) "acceptor" proteins, which bind to pHis residues and undergo phosphotransfer from pHis. While the roles of pHis acceptors are well-documented in bacterial cell signalling and metabolism, the presence and functions of additional pHis acceptors remain largely unknown. In this study, we introduce a chemoproteomic strategy leveraging a stable analogue of 3-pHis to identify 13 putative pHis acceptors in Escherichia coli. Among these, we identified phosphofructokinase-1 (PfkA), a central enzyme in glycolysis, as a pHis acceptor phosphorylated at His249 by phosphocarrier protein HPr (PtsH). This phosphorylation, modulated by carbon source availability, inhibited PfkA's kinase activity, while the pHis-specific phosphatase signal inhibitory factor X (SixA) reversed the effect, restoring the kinase function. Our findings reveal a novel regulatory mechanism in which histidine phosphorylation dynamically controls a key glycolytic enzyme, implicating a broader role for pHis in bacterial metabolism.
Collapse
Affiliation(s)
- Solbee Choi
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 South Korea
| | - Seungmin Ahn
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 South Korea
| | - Kyung Hyun Cho
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 South Korea
| | - Sung Kuk Lee
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 South Korea
| | - Jung-Min Kee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 South Korea
| |
Collapse
|
2
|
Kuppuswamy S, Watson NJ, Ledford WL, Pavri BA, Zhi W, Gbadebo M, Bonsack F, Xu H, Sukumari-Ramesh S. Brain proteome changes after intracerebral hemorrhage in aged male and female mice. Neurobiol Dis 2025; 212:106936. [PMID: 40320180 DOI: 10.1016/j.nbd.2025.106936] [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: 09/16/2024] [Revised: 03/10/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025] Open
Abstract
Aging is an independent predictor of adverse outcomes after intracerebral hemorrhage (ICH), a stroke subtype with no effective treatment. Despite the expected increase in the incidence of ICH due to population aging and the widespread use of anticoagulants, preclinical studies with aged animal subjects are lacking, and the pathophysiology of ICH in aged subjects has yet to be defined. Herein, we attempt to characterize the brain proteomic changes after ICH using an unbiased label- free quantitative proteomics approach and bioinformatics. To this end, aged male and female mice (18-24 months old) were subjected to sham/ICH. Mice were euthanized on day 3 post-surgery, and ipsilateral brain tissue was collected and subjected to LC-MS/MS analysis. Considering sex as a biological variable, the data derived from males and females were separately analyzed. The proteomics analysis revealed 133 differentially expressed proteins (DEPs) between the sham and ICH groups in male subjects. Among the DEPs, 98 proteins were downregulated, and 35 proteins were upregulated after ICH, compared to sham. In aged female mice, 315 DEPs were identified, of which 221 proteins were downregulated, and 94 proteins were upregulated after ICH compared to sham. The mass spectrometry data was validated using immunohistochemistry or western blot analysis, and the bioinformatics analysis revealed a comprehensive understanding of the signaling pathways associated with ICH. Some DEPs in both aged male and female mice that could play roles in ICH pathology were 14-3-3 proteins and S100-A9. The study also revealed that mitochondrial dysfunction could be a critical regulator of ICH-induced acute brain damage. Overall, the generated proteomics data could help develop hypothesis-driven functional analysis and delineate the complex pathobiology of ICH.
Collapse
Affiliation(s)
- Sivaraman Kuppuswamy
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Noah J Watson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - William Luke Ledford
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Blake A Pavri
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Wenbo Zhi
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Mary Gbadebo
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Frederick Bonsack
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Hongyan Xu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America
| | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1120, 15th Street, CB3515, Augusta, GA 30912, United States of America.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Fu H, Mo X, Ivanov AA. Decoding the functional impact of the cancer genome through protein-protein interactions. Nat Rev Cancer 2025; 25:189-208. [PMID: 39810024 DOI: 10.1038/s41568-024-00784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/16/2025]
Abstract
Acquisition of genomic mutations enables cancer cells to gain fitness advantages under selective pressure and, ultimately, leads to oncogenic transformation. Interestingly, driver mutations, even within the same gene, can yield distinct phenotypes and clinical outcomes, necessitating a mutation-focused approach. Conversely, cellular functions are governed by molecular machines and signalling networks that are mostly controlled by protein-protein interactions (PPIs). The functional impact of individual genomic alterations could be transmitted through regulated nodes and hubs of PPIs. Oncogenic mutations may lead to modified residues of proteins, enabling interactions with other proteins that the wild-type protein does not typically interact with, or preventing interactions with proteins that the wild-type protein usually interacts with. This can result in the rewiring of molecular signalling cascades and the acquisition of an oncogenic phenotype. Here, we review the altered PPIs driven by oncogenic mutations, discuss technologies for monitoring PPIs and provide a functional analysis of mutation-directed PPIs. These driver mutation-enabled PPIs and mutation-perturbed PPIs present a new paradigm for the development of tumour-specific therapeutics. The intersection of cancer variants and altered PPI interfaces represents a new frontier for understanding oncogenic rewiring and developing tumour-selective therapeutic strategies.
Collapse
Affiliation(s)
- Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Xiulei Mo
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Rubino E, Italia M, Giorgio E, Boschi S, Dimartino P, Pippucci T, Roveta F, Cambria CM, Elia G, Marcinnò A, Gallone S, Rogaeva E, Antonucci F, Brusco A, Gardoni F, Rainero I. Exome sequencing reveals a rare damaging variant in GRIN2C in familial late-onset Alzheimer's disease. Alzheimers Res Ther 2025; 17:21. [PMID: 39810256 PMCID: PMC11730494 DOI: 10.1186/s13195-024-01661-y] [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: 07/10/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with both genetic and environmental factors contributing to its pathogenesis. While early-onset AD has well-established genetic determinants, the genetic basis for late-onset AD remains less clear. This study investigates a large Italian family with late-onset autosomal dominant AD, identifying a novel rare missense variant in GRIN2C gene associated with the disease, and evaluates the functional impact of this variant. METHODS Affected and unaffected members from a Northern Italian family were included. Genomic DNA from family members was extracted and initially screened for pathogenic mutations in APP, PSEN1, and PSEN2, and screened for 77 genes associated with neurodegenerative conditions using NeuroX array assay. Exome sequencing was performed on three affected individuals and two healthy relatives. Bioinformatics analyses were conducted. Functional analysis was performed using primary neuronal cultures, and the impact of the variant was assessed through immunocytochemistry and electrophysiology. RESULTS Pathogenic variants were not identified in APP, PSEN1, or PSEN2, nor in the 77 genes in NeuroX array assay. Exome Sequencing revealed the c.3215C > T p.(A1072V) variant in GRIN2C gene (NM 000835.6), encoding for the glutamate ionotropic receptor N-methyl-D-aspartate receptor (NMDA) type subunit 2C (GluN2C). This variant segregated in 6 available AD patients in the family and was absent in 9 healthy relatives. Primary rat hippocampal neurons overexpressing GluN2CA1072V showed an increase in NMDAR-induced currents, suggesting altered glutamatergic transmission. Surface expression assays demonstrated an elevated surface/total ratio of the mutant GluN2C, correlating with the increased NMDAR current. Additionally, immunocytochemistry revealed in neurons expressing the mutant variant a reduced colocalization between the GluN2C subunit and 14-3-3 proteins, which are known to facilitate membrane trafficking of NMDARs. DISCUSSION We identified a rare missense variant in GRIN2C associated with late-onset autosomal dominant Alzheimer's disease. These findings highlight the role of GluN2C-containing NMDARs in glutamatergic signaling and their potential contribution to AD pathogenesis.
Collapse
Affiliation(s)
- Elisa Rubino
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy.
- Center for Alzheimer's Disease and Related Dementias, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, University Hospital, Via Cherasco 15, Turin, 10126, Italy.
| | - Maria Italia
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, 20133, Italy
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, Pavia, 27100, Italy
- Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, 27100, Italy
| | - Silvia Boschi
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Paola Dimartino
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, Pavia, 27100, Italy
| | - Tommaso Pippucci
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria, Via Albertoni 15, Bologna, 40138, Italy
| | - Fausto Roveta
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Clara Maria Cambria
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Via Festa del Perdono 7, Milan, 20122, Italy
| | - Gabriella Elia
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Andrea Marcinnò
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Salvatore Gallone
- Center for Alzheimer's Disease and Related Dementias, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, University Hospital, Via Cherasco 15, Turin, 10126, Italy
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, King's College Circle 1, Toronto, ON, M5S1A8, Canada
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Via Festa del Perdono 7, Milan, 20122, Italy
- Institute of Neuroscience, IN-CNR, Via Raoul Follereau 3, Vedano al Lambro, 20854, Italy
| | - Alfredo Brusco
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
- Medical Genetics Unit, Città della Salute e della Scienza di Torino, University Hospital, Via Santena 19, Turin, 10126, Italy
- Molecular Biotechnology Center Guido Tarone, University of Turin, Piazza Nizza 44B, Turin, 10126, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, 20133, Italy
| | - Innocenzo Rainero
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
- Center for Alzheimer's Disease and Related Dementias, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, University Hospital, Via Cherasco 15, Turin, 10126, Italy
| |
Collapse
|
17
|
Hossain MA. A comprehensive review of targeting RAF kinase in cancer. Eur J Pharmacol 2025; 986:177142. [PMID: 39577552 DOI: 10.1016/j.ejphar.2024.177142] [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: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
RAF kinases, particularly the BRAF isoform, play a crucial role in the MAPK/ERK signaling pathway, regulating key cellular processes such as proliferation, differentiation, and survival. Dysregulation of this pathway often caused by mutations in the BRAF gene or alterations in upstream regulators like Ras and receptor tyrosine kinases contributes significantly to cancer development. Mutations, such as BRAF-V600E, are present in a variety of malignancies, with the highest prevalence in melanoma. Targeted therapies against RAF kinases have achieved substantial success, especially in BRAF-V600E-mutant melanomas, where inhibitors like vemurafenib and dabrafenib have demonstrated remarkable efficacy, leading to improved patient outcomes. These inhibitors have also shown clinical benefits in cancers such as thyroid and colorectal carcinoma, although to a lesser extent. Despite these successes, therapeutic resistance remains a major hurdle. Resistance mechanisms, including RAF dimerization, feedback reactivation of the MAPK pathway, and paradoxical activation of ERK signaling, often lead to diminished efficacy over time, resulting in disease progression or even secondary malignancies. In response, current research is focusing on novel therapeutic strategies, including combination therapies that target multiple components of the pathway simultaneously, such as MEK inhibitors used in tandem with RAF inhibitors. Additionally, next-generation RAF inhibitors are being developed to address resistance and enhance therapeutic specificity. This review discusses the clinical advancements in RAF-targeted therapies, with a focus on ongoing efforts to overcome therapeutic resistance and enhance outcomes for cancer patients. It also underscores the persistent challenges in effectively targeting RAF kinase in oncology.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
18
|
Tian T, He S, Hao H, Guan B, Gong Y, Fan J, Zhu Z, Gao W, Wu Y, Feng N, Wang A, Guo Y, Li X. YWHAG promotes bladder cancer metastasis by regulating TMOD3 to activate ERK1/2 and JNK phosphorylation in the MAPK pathway. J Transl Med 2024; 22:1159. [PMID: 39741303 DOI: 10.1186/s12967-024-06003-y] [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: 08/28/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE This study aims to investigate the molecular mechanisms by which YWHAG (Tyrosine 3-Monooxygenase/Tryptophan 5-Monooxygenase Activation Protein Gamma) promotes metastasis in bladder cancer. Specifically, it seeks to elucidate the role of YWHAG in driving cancer cell invasion and its potential as a prognostic marker for bladder cancer progression. METHODS The expression pattern of YWHAG in both primary and metastatic bladder cancer tissues was analyzed using immunohistochemistry (IHC) to determine its correlation with clinical stage and prognosis in bladder cancer patients. The functional role of YWHAG in bladder cancer progression was examined through a series of in vitro and in vivo experiments. Transcriptome sequencing was employed to identify the key signaling pathways regulated by YWHAG. The interaction between YWHAG and TMOD3 (Tropomodulin 3) was confirmed through pull-down assays coupled with mass spectrometry, co-immunoprecipitation (Co-IP), and cell immunofluorescence studies. Finally, TMOD3 knockdown experiments were conducted to verify whether the pro-metastatic effects of YWHAG in bladder cancer are mediated through TMOD3. RESULTS YWHAG expression was significantly elevated in metastatic bladder cancer tissues compared to primary tumor tissues, and its expression positively correlated with advanced clinical stages and poor prognosis in patients. In vitro and in vivo experiments demonstrated that YWHAG knockdown significantly reduced the invasive, metastatic, and colonization capabilities of bladder cancer cells. Transcriptome analysis revealed that YWHAG knockdown markedly inhibited the phosphorylation of ERK1/2 (extracellular signal-related kinases 1 and 2) and JNK (JUN N-terminal kinases), key components of the MAPK (mitogen-activated protein kinase) signaling pathway. Mechanistically, YWHAG was found to promote bladder cancer cell invasion and metastasis by regulating TMOD3, which subsequently activates the MAPK pathway. CONCLUSION YWHAG upregulates TMOD3 expression, leading to the activation of ERK1/2 phosphorylation in the MAPK pathway, thereby promoting the invasion and metastasis of bladder cancer cells.
Collapse
Affiliation(s)
- Tai Tian
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- Department of Urology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Han Hao
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Bao Guan
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Jian Fan
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Zhenpeng Zhu
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Wenzhi Gao
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Yucai Wu
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
| | - Ninghan Feng
- Department of Urology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, 214002, China.
| | - Aixiang Wang
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China.
- Institute of Urology, Peking University, Beijing, 100034, China.
| | - Yuexian Guo
- Department of Urology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China.
- Institute of Urology, Peking University, Beijing, 100034, China.
| |
Collapse
|
19
|
Martinez Fiesco JA, Li N, Alvarez de la Cruz A, Metcalfe RD, Beilina A, Cookson MR, Zhang P. 14-3-3 binding maintains the Parkinson's associated kinase LRRK2 in an inactive state. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624879. [PMID: 39605327 PMCID: PMC11601620 DOI: 10.1101/2024.11.22.624879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a central player in cellular signaling and a significant contributor to Parkinson's disease (PD) pathogenesis. 14-3-3 proteins are essential regulators of LRRK2, modulating its activity. Here, we present the cryo- electron microscopy structure of the LRRK2:14-3-3 2 autoinhibitory complex, showing that a 14-3-3 dimer stabilizes an autoinhibited LRRK2 monomer by binding to key phosphorylation sites and the COR-A and COR-B subdomains within the Roc-COR GTPase domain of LRRK2. This interaction locks LRRK2 in an inactive conformation, restricting LRR domain mobility and preventing dimerization and oligomer formation. Our mutagenesis studies reveal that PD-associated mutations at the COR:14-3-3 interface and within the GTPase domain reduce 14-3-3 binding, diminishing its inhibitory effect on LRRK2. These findings provide a structural basis for understanding how LRRK2 likely remains dormant within cells, illuminate aspects of critical PD biomarkers, and suggest therapeutic strategies to enhance LRRK2-14-3-3 interactions to treat PD and related disorders.
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Anbalagan GK, Agarwal P, Ghosh SK. Evidence of 14-3-3 proteins contributing to kinetochore integrity and chromosome congression during mitosis. J Cell Sci 2024; 137:jcs261928. [PMID: 38988319 PMCID: PMC11698032 DOI: 10.1242/jcs.261928] [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: 12/24/2023] [Accepted: 07/05/2024] [Indexed: 07/12/2024] Open
Abstract
The 14-3-3 family of proteins are conserved across eukaryotes and serve myriad important regulatory functions in the cell. Homo- and hetero-dimers of these proteins mainly recognize their ligands via conserved motifs to modulate the localization and functions of those effector ligands. In most of the genetic backgrounds of Saccharomyces cerevisiae, disruption of both 14-3-3 homologs (Bmh1 and Bmh2) are either lethal or cells survive with severe growth defects, including gross chromosomal missegregation and prolonged cell cycle arrest. To elucidate their contributions to chromosome segregation, in this work, we investigated their centromere- and kinetochore-related functions of Bmh1 and Bmh2. Analysis of appropriate deletion mutants shows that Bmh isoforms have cumulative and non-shared isoform-specific contributions in maintaining the proper integrity of the kinetochore ensemble. Consequently, Bmh mutant cells exhibited perturbations in kinetochore-microtubule (KT-MT) dynamics, characterized by kinetochore declustering, mis-localization of kinetochore proteins and Mad2-mediated transient G2/M arrest. These defects also caused an asynchronous chromosome congression in bmh mutants during metaphase. In summary, this report advances the knowledge on contributions of budding yeast 14-3-3 proteins in chromosome segregation by demonstrating their roles in kinetochore integrity and chromosome congression.
Collapse
Affiliation(s)
| | - Prakhar Agarwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, 400 076, India
| | - Santanu Kumar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, 400 076, India
| |
Collapse
|
26
|
Wan S, Wang S, Yang X, Cui Y, Guan H, Xiao W, Liu F. Regulation of H9C2 cell hypertrophy by 14-3-3η via inhibiting glycolysis. PLoS One 2024; 19:e0307696. [PMID: 39038022 PMCID: PMC11262655 DOI: 10.1371/journal.pone.0307696] [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: 02/18/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
It has been reported that Ywhah (14-3-3η) reduces glycolysis. However, it remains unclear about the downstream mechanism by which glycolysis is regulated by 14-3-3η in cardiac hypertrophy. As an important regulator, Yes-associated protein (YAP) interacts with 14-3-3η to participate in the initiation and progression of various diseases in vivo. In this study, the model of H9C2 cardiomyocyte hypertrophy was established by triiodothyronine (T3) or rotenone stimulation to probe into the action mechanism of 14-3-3η. Interestingly, the overexpression of 14-3-3η attenuated T3 or rotenone induced cardiomyocyte hypertrophy and decreased glycolysis in H9C2 cardiomyocytes, whereas the knockdown of 14-3-3η had an opposite effect. Mechanistically, 14-3-3η can reduce the expression level of YAP and bind to it to reduce its nuclear translocation. In addition, changing YAP may affect the expression of lactate dehydrogenase A (LDHA), a glycolysis-related protein. Meanwhile, LDHA is also a possible target for 14-3-3η to mediate glycolysis based on changes in pyruvate, a substrate of LDHA. Collectively, 14-3-3η can suppress cardiomyocyte hypertrophy via decreasing the nucleus translocation of YAP and glycolysis, which indicates that 14-3-3η could be a promising target for inhibiting cardiac hypertrophy.
Collapse
Affiliation(s)
- Sha Wan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Songhao Wang
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Xianfei Yang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
| | - Yalan Cui
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
- Clinical Pathology Department, The Second People’s Hospital of Yichang, Yichang, China
| | - Heng Guan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Wenping Xiao
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Fang Liu
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| |
Collapse
|
27
|
Chen J, Liu L, Wang G, Chen G, Liu X, Li M, Han L, Song W, Wang S, Li C, Wang Z, Huang Y, Gu C, Yang Z, Zhou Z, Zhao J, Zhang X. The AGAMOUS-LIKE 16-GENERAL REGULATORY FACTOR 1 module regulates axillary bud outgrowth via catabolism of abscisic acid in cucumber. THE PLANT CELL 2024; 36:2689-2708. [PMID: 38581430 PMCID: PMC11218829 DOI: 10.1093/plcell/koae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/02/2024] [Accepted: 03/01/2024] [Indexed: 04/08/2024]
Abstract
Lateral branches are important components of shoot architecture and directly affect crop yield and production cost. Although sporadic studies have implicated abscisic acid (ABA) biosynthesis in axillary bud outgrowth, the function of ABA catabolism and its upstream regulators in shoot branching remain elusive. Here, we showed that the MADS-box transcription factor AGAMOUS-LIKE 16 (CsAGL16) is a positive regulator of axillary bud outgrowth in cucumber (Cucumis sativus). Functional disruption of CsAGL16 led to reduced bud outgrowth, whereas overexpression of CsAGL16 resulted in enhanced branching. CsAGL16 directly binds to the promoter of the ABA 8'-hydroxylase gene CsCYP707A4 and promotes its expression. Loss of CsCYP707A4 function inhibited axillary bud outgrowth and increased ABA levels. Elevated expression of CsCYP707A4 or treatment with an ABA biosynthesis inhibitor largely rescued the Csagl16 mutant phenotype. Moreover, cucumber General Regulatory Factor 1 (CsGRF1) interacts with CsAGL16 and antagonizes CsAGL16-mediated CsCYP707A4 activation. Disruption of CsGRF1 resulted in elongated branches and decreased ABA levels in the axillary buds. The Csagl16 Csgrf1 double mutant exhibited a branching phenotype resembling that of the Csagl16 single mutant. Therefore, our data suggest that the CsAGL16-CsGRF1 module regulates axillary bud outgrowth via CsCYP707A4-mediated ABA catabolism in cucumber. Our findings provide a strategy to manipulate ABA levels in axillary buds during crop breeding to produce desirable branching phenotypes.
Collapse
Affiliation(s)
- Jiacai Chen
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Liu Liu
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Guanghui Wang
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Guangxin Chen
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaofeng Liu
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Min Li
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Lijie Han
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Weiyuan Song
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Shaoyun Wang
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Chuang Li
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Zhongyi Wang
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Yuxiang Huang
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Chaoheng Gu
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Zhengan Yang
- College of Landscape and Horticulture, Yunnan Agricultural University, Kunming, Yunnan 650201, China
| | - Zhaoyang Zhou
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Jianyu Zhao
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaolan Zhang
- Beijing Key Laboratory of Growth and Developmental Regulation for Protected Vegetable Crops, Department of Vegetable Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
28
|
Winter DL, Wairara AR, Bennett JL, Donald WA, Glover DJ. Protein Interaction Kinetics Delimit the Performance of Phosphorylation-Driven Protein Switches. ACS Synth Biol 2024; 13:1781-1797. [PMID: 38830815 DOI: 10.1021/acssynbio.4c00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Post-translational modifications (PTMs) such as phosphorylation and dephosphorylation can rapidly alter protein surface chemistry and structural conformation, which can switch protein-protein interactions (PPIs) within signaling networks. Recently, de novo-designed phosphorylation-responsive protein switches have been created that harness kinase- and phosphatase-mediated phosphorylation to modulate PPIs. PTM-driven protein switches are promising tools for investigating PTM dynamics in living cells, developing biocompatible nanodevices, and engineering signaling pathways to program cell behavior. However, little is known about the physical and kinetic constraints of PTM-driven protein switches, which limits their practical application. In this study, we present a framework to evaluate two-component PTM-driven protein switches based on four performance metrics: effective concentration, dynamic range, response time, and reversibility. Our computational models reveal an intricate relationship between the binding kinetics, phosphorylation kinetics, and switch concentration that governs the sensitivity and reversibility of PTM-driven protein switches. Building upon the insights of the interaction modeling, we built and evaluated novel phosphorylation-driven protein switches consisting of phosphorylation-sensitive coiled coils as sensor domains fused to fluorescent proteins as actuator domains. By modulating the phosphorylation state of the switches with a specific protein kinase and phosphatase, we demonstrate fast, reversible transitions between "on" and "off" states. The response of the switches linearly correlated to the kinase concentration, demonstrating its potential as a biosensor for kinase measurements in real time. As intended, the switches responded to specific kinase activity with an increase in the fluorescence signal and our model could be used to distinguish between two mechanisms of switch activation: dimerization or a structural rearrangement. The protein switch kinetics model developed here should enable PTM-driven switches to be designed with ideal performance for specific applications.
Collapse
Affiliation(s)
- Daniel L Winter
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Synthetic Biology Future Science Platform, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Canberra, ACT 2601, Australia
| | - Adelgisa R Wairara
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jack L Bennett
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - William A Donald
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Dominic J Glover
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
29
|
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
|
30
|
Spencer-Smith R. The RAF cysteine-rich domain: Structure, function, and role in disease. Adv Cancer Res 2024; 164:69-91. [PMID: 39306370 DOI: 10.1016/bs.acr.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
RAF kinases, consisting of ARAF, BRAF and CRAF, are direct effectors of RAS GTPases and critical for signal transduction through the RAS-MAPK pathway. Driver mutations in BRAF are commonplace in human cancer, while germline mutations in BRAF and CRAF cause RASopathy development syndromes. However, there remains a lack of effective drugs that target RAF function, which is partially due to the complexity of the RAF activation cycle. Therefore, greater understanding of RAF regulation is required to identify new approaches that target its function in disease. A key piece of this puzzle is the RAF zinc finger, often referred to as the cysteine-rich domain (CRD). The CRD is a lipid and protein binding domain which plays complex and opposing roles in the RAF activation cycle. Firstly, it supports the RAS-RAF interaction during RAF activation by binding to phosphatidylserine (PS) in the plasma membrane and by making direct RAS contacts. Conversely, under quiescent conditions the CRD also plays a critical role in maintaining RAF in a closed, autoinhibited state. However, the interplay between these activities and their relative importance for RAF activation were not well understood. Recent structural and biochemical studies have contributed greatly to our understanding of these roles and identified functional differences between BRAF CRD and that of CRAF. This chapter provides an in-depth review of the CRDs roles in RAF regulation and how they may inform novel approaches to target RAF function.
Collapse
Affiliation(s)
- Russell Spencer-Smith
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina.
| |
Collapse
|
31
|
Sakai K, Aoki K, Goto Y. Live-cell fluorescence imaging and optogenetic control of PKA kinase activity in fission yeast Schizosaccharomyces pombe. Yeast 2024; 41:349-363. [PMID: 38583078 DOI: 10.1002/yea.3937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/21/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024] Open
Abstract
The cAMP-PKA signaling pathway plays a crucial role in sensing and responding to nutrient availability in the fission yeast Schizosaccharomyces pombe. This pathway monitors external glucose levels to control cell growth and sexual differentiation. However, the temporal dynamics of the cAMP-PKA pathway in response to external stimuli remains unclear mainly due to the lack of tools to quantitatively visualize the activity of the pathway. Here, we report the development of the kinase translocation reporter (KTR)-based biosensor spPKA-KTR1.0, which allows us to measure the dynamics of PKA activity in fission yeast cells. The spPKA-KTR1.0 is derived from the transcription factor Rst2, which translocates from the nucleus to the cytoplasm upon PKA activation. We found that spPKA-KTR1.0 translocates between the nucleus and cytoplasm in a cAMP-PKA pathway-dependent manner, indicating that the spPKA-KTR1.0 is a reliable indicator of the PKA activity in fission yeast cells. In addition, we implemented a system that simultaneously visualizes and manipulates the cAMP-PKA signaling dynamics by introducing bPAC, a photoactivatable adenylate cyclase, in combination with spPKA-KTR1.0. This system offers an opportunity for investigating the role of the signaling dynamics of the cAMP-PKA pathway in fission yeast cells with higher temporal resolution.
Collapse
Affiliation(s)
- Keiichiro Sakai
- Quantitative Biology Research Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Division of Integrated Life Science, Department of Gene Mechanisms, Laboratory of Cell Cycle Regulation, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yuhei Goto
- Quantitative Biology Research Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
Moors TE, Morella ML, Bertran-Cobo C, Geut H, Udayar V, Timmermans-Huisman E, Ingrassia AMT, Brevé JJP, Bol JGJM, Bonifati V, Jagasia R, van de Berg WDJ. Altered TFEB subcellular localization in nigral neurons of subjects with incidental, sporadic and GBA-related Lewy body diseases. Acta Neuropathol 2024; 147:67. [PMID: 38581586 PMCID: PMC10998821 DOI: 10.1007/s00401-024-02707-z] [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: 08/03/2023] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 04/08/2024]
Abstract
Transcription factor EB (TFEB) is a master regulator of genes involved in the maintenance of autophagic and lysosomal homeostasis, processes which have been implicated in the pathogenesis of GBA-related and sporadic Parkinson's disease (PD), and dementia with Lewy bodies (DLB). TFEB activation results in its translocation from the cytosol to the nucleus. Here, we investigated TFEB subcellular localization and its relation to intracellular alpha-synuclein (aSyn) accumulation in post-mortem human brain of individuals with either incidental Lewy body disease (iLBD), GBA-related PD/DLB (GBA-PD/DLB) or sporadic PD/DLB (sPD/DLB), compared to control subjects. We analyzed nigral dopaminergic neurons using high-resolution confocal and stimulated emission depletion (STED) microscopy and semi-quantitatively scored the TFEB subcellular localization patterns. We observed reduced nuclear TFEB immunoreactivity in PD/DLB patients compared to controls, both in sporadic and GBA-related cases, as well as in iLBD cases. Nuclear depletion of TFEB was more pronounced in neurons with Ser129-phosphorylated (pSer129) aSyn accumulation in all groups. Importantly, we observed previously-unidentified TFEB-immunopositive perinuclear clusters in human dopaminergic neurons, which localized at the Golgi apparatus. These TFEB clusters were more frequently observed and more severe in iLBD, sPD/DLB and GBA-PD/DLB compared to controls, particularly in pSer129 aSyn-positive neurons, but also in neurons lacking detectable aSyn accumulation. In aSyn-negative cells, cytoplasmic TFEB clusters were more frequently observed in GBA-PD/DLB and iLBD patients, and correlated with reduced GBA enzymatic activity as well as increased Braak LB stage. Altered TFEB distribution was accompanied by a reduction in overall mRNA expression levels of selected TFEB-regulated genes, indicating a possible early dysfunction of lysosomal regulation. Overall, we observed cytoplasmic TFEB retention and accumulation at the Golgi in cells without apparent pSer129 aSyn accumulation in iLBD and PD/DLB patients. This suggests potential TFEB impairment at the early stages of cellular disease and underscores TFEB as a promising therapeutic target for synucleinopathies.
Collapse
Affiliation(s)
- Tim E Moors
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Martino L Morella
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Cesc Bertran-Cobo
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Hanneke Geut
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Vinod Udayar
- Roche Pharma Research and Early Development; Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Evelien Timmermans-Huisman
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Angela M T Ingrassia
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - John J P Brevé
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - John G J M Bol
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Vincenzo Bonifati
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ravi Jagasia
- Roche Pharma Research and Early Development; Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands.
| |
Collapse
|
36
|
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
|
37
|
Dedden D, Nitsche J, Schneider EV, Thomsen M, Schwarz D, Leuthner B, Grädler U. Cryo-EM Structures of CRAF 2/14-3-3 2 and CRAF 2/14-3-3 2/MEK1 2 Complexes. J Mol Biol 2024; 436:168483. [PMID: 38331211 DOI: 10.1016/j.jmb.2024.168483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/22/2023] [Accepted: 02/02/2024] [Indexed: 02/10/2024]
Abstract
RAF protein kinases are essential effectors in the MAPK pathway and are important cancer drug targets. Structural understanding of RAF activation is so far based on cryo-electron microscopy (cryo-EM) and X-ray structures of BRAF in different conformational states as inactive or active complexes with KRAS, 14-3-3 and MEK1. In this study, we have solved the first cryo-EM structures of CRAF2/14-3-32 at 3.4 Å resolution and CRAF2/14-3-32/MEK12 at 4.2 Å resolution using CRAF kinase domain expressed as constitutively active Y340D/Y341D mutant in insect cells. The overall architecture of our CRAF2/14-3-32 and CRAF2/14-3-32/MEK12 cryo-EM structures is highly similar to corresponding BRAF structures in complex with 14-3-3 or 14-3-3/MEK1 and represent the activated dimeric RAF conformation. Our CRAF cryo-EM structures provide additional insights into structural understanding of the activated CRAF2/14-3-32/MEK12 complex.
Collapse
Affiliation(s)
- Dirk Dedden
- Proteros biostructures GmbH, Bunsenstraße 7a, D-82152 Planegg-Martinsried, Germany
| | - Julius Nitsche
- Proteros biostructures GmbH, Bunsenstraße 7a, D-82152 Planegg-Martinsried, Germany
| | | | - Maren Thomsen
- Proteros biostructures GmbH, Bunsenstraße 7a, D-82152 Planegg-Martinsried, Germany
| | - Daniel Schwarz
- The Healthcare Business of Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Birgitta Leuthner
- The Healthcare Business of Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Ulrich Grädler
- The Healthcare Business of Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany.
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Scheible N, Henning PM, McCubbin AG. Calmodulin-Domain Protein Kinase PiCDPK1 Interacts with the 14-3-3-like Protein NtGF14 to Modulate Pollen Tube Growth. PLANTS (BASEL, SWITZERLAND) 2024; 13:451. [PMID: 38337984 PMCID: PMC10857193 DOI: 10.3390/plants13030451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Calcium-mediated signaling pathways are known to play important roles in the polar growth of pollen tubes. The calcium-dependent protein kinase, PiCDPK1, has been shown to be involved in regulating this process through interaction with a guanine dissociation inhibitor, PiRhoGDI1. To more fully understand the role of PiCDPK1 in pollen tube extension, we designed a pull-down study to identify additional substrates of this kinase. These experiments identified 123 putative interactors. Two of the identified proteins were predicted to directly interact with PiCDPK1, and this possibility was investigated in planta. The first, NtGF14, a 14-3-3-like protein, did not produce a noticeable phenotype when overexpressed in pollen alone but partially rescued the spherical tube phenotype caused by PiCDPK1 over-expression when co-over-expressed with the kinase. The second, NtREN1, a GTPase activating protein (GAP), severely inhibited pollen tube germination when over-expressed, and its co-over-expression with PiCDPK1 did not substantially affect this phenotype. These results suggest a novel in vivo interaction between NtGF14 and PiCDPK1 but do not support the direct interaction between PiCDPK1 and NtREN1. We demonstrate the utility of the methodology used to identify potential protein interactions while confirming the necessity of additional studies to confirm their validity. Finally, additional support was found for intersection between PiCDPK1 and RopGTPase pathways to control polar growth at the pollen tube tip.
Collapse
Affiliation(s)
| | | | - Andrew G. McCubbin
- School of Biological Sciences, Washington State University, Pullman, WA 99164, USA; (N.S.); (P.M.H.)
| |
Collapse
|
41
|
Zeylan M, Senyuz S, Picón-Pagès P, García-Elías A, Tajes M, Muñoz FJ, Oliva B, Garcia-Ojalvo J, Barbu E, Vicente R, Nattel S, Ois A, Puig-Pijoan A, Keskin O, Gursoy A. Shared Proteins and Pathways of Cardiovascular and Cognitive Diseases: Relation to Vascular Cognitive Impairment. J Proteome Res 2024; 23:560-573. [PMID: 38252700 PMCID: PMC10846560 DOI: 10.1021/acs.jproteome.3c00289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/29/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
One of the primary goals of systems medicine is the detection of putative proteins and pathways involved in disease progression and pathological phenotypes. Vascular cognitive impairment (VCI) is a heterogeneous condition manifesting as cognitive impairment resulting from vascular factors. The precise mechanisms underlying this relationship remain unclear, which poses challenges for experimental research. Here, we applied computational approaches like systems biology to unveil and select relevant proteins and pathways related to VCI by studying the crosstalk between cardiovascular and cognitive diseases. In addition, we specifically included signals related to oxidative stress, a common etiologic factor tightly linked to aging, a major determinant of VCI. Our results show that pathways associated with oxidative stress are quite relevant, as most of the prioritized vascular cognitive genes and proteins were enriched in these pathways. Our analysis provided a short list of proteins that could be contributing to VCI: DOLK, TSC1, ATP1A1, MAPK14, YWHAZ, CREB3, HSPB1, PRDX6, and LMNA. Moreover, our experimental results suggest a high implication of glycative stress, generating oxidative processes and post-translational protein modifications through advanced glycation end-products (AGEs). We propose that these products interact with their specific receptors (RAGE) and Notch signaling to contribute to the etiology of VCI.
Collapse
Affiliation(s)
- Melisa
E. Zeylan
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Simge Senyuz
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Pol Picón-Pagès
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Anna García-Elías
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Marta Tajes
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Francisco J. Muñoz
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Baldomero Oliva
- Laboratory
of Structural Bioinformatics (GRIB), Department of Medicine and Life
Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Jordi Garcia-Ojalvo
- Laboratory
of Dynamical Systems Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Eduard Barbu
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Raul Vicente
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Stanley Nattel
- Department
of Medicine and Research Center, Montreal Heart Institute and Université
de Montréal; Institute of Pharmacology, West German Heart and
Vascular Center, University Duisburg-Essen,
Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux 33000, France
| | - Angel Ois
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Albert Puig-Pijoan
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Ozlem Keskin
- Department
of Chemical and Biological Engineering, Koç University, Istanbul 34450, Türkiye
| | - Attila Gursoy
- Department
of Computer Engineering, Koç University, Istanbul 34450, Türkiye
| |
Collapse
|
42
|
Khan A, Bealy MA, Alharbi B, Khan S, Alharethi SH, Al-Soud WA, Mohammad T, Hassan MI, Alshammari N, Ahmed Al-Keridis L. Discovering potential inhibitors of Raf proto-oncogene serine/threonine kinase 1: a virtual screening approach towards anticancer drug development. J Biomol Struct Dyn 2024; 42:1846-1857. [PMID: 37104027 DOI: 10.1080/07391102.2023.2204380] [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: 02/17/2023] [Accepted: 04/08/2023] [Indexed: 04/28/2023]
Abstract
Raf proto-oncogene serine/threonine kinase 1 (RAF1 or c-Raf) is a serine/threonine protein kinase crucial in regulating cell growth, differentiation, and survival. Any disruption or overexpression of RAF1 can result in neoplastic transformation and other disorders such as cardiomyopathy, Noonan syndrome, leopard syndrome, etc. RAF1 has been identified as a potential therapeutic target in drug development against various complex diseases, including cancer, due to its remarkable role in disease progression. Here, we carried out a multitier virtual screening study involving different in-silico approaches to discover potential inhibitors of RAF1. After applying the Lipinski rule of five, we retrieved all phytocompounds from the IMPPAT database based on their physicochemical properties. We performed a molecular docking-based virtual screening and got top hits with the best binding affinity and ligand efficiency. Then we screened out the selected hits using the PAINS filter, ADMET properties, and other druglike features. Eventually, PASS evaluation identifies two phytocompounds, Moracin C and Tectochrysin, with appreciable anti-cancerous properties. Finally, all-atom molecular dynamics simulation (MDS) followed by interaction analysis was performed on the elucidated compounds in complex with RAF1 for 200 ns to investigate their time-evolution dynamics and interaction mechanism. Molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) and Dynamical Cross-Correlation Matrix (DCCM) analyses then followed these results from the simulated trajectories. According to the results, the elucidated compounds stabilize the RAF1 structure and lead to fewer conformational alterations. The results of the current study indicated that Moracin C and Tectochrysin could serve as potential inhibitors of RAF1 after required validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Afsha Khan
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Mohamed Ahmed Bealy
- Department of Pathology, College of Medicine, University of Ha'il, Hail, Saudi Arabia
| | - Bandar Alharbi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha'il, Hail, Saudi Arabia
| | - Shama Khan
- Faculty of Health Science, South Africa Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudi Arabia
| | - Waleed Abu Al-Soud
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Lamya Ahmed Al-Keridis
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
43
|
Davarinejad H, Arvanitis-Vigneault A, Nygard D, Lavallée-Adam M, Couture JF. Modus operandi: Chromatin recognition by α-helical histone readers. Structure 2024; 32:8-17. [PMID: 37922903 DOI: 10.1016/j.str.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/25/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023]
Abstract
Histone reader domains provide a mechanism for sensing states of coordinated nuclear processes marked by histone proteins' post-translational modifications (PTMs). Among a growing number of discovered histone readers, the 14-3-3s, ankyrin repeat domains (ARDs), tetratricopeptide repeats (TPRs), bromodomains (BRDs), and HEAT domains are a group of domains using various mechanisms to recognize unmodified or modified histones, yet they all are composed of an α-helical fold. In this review, we compare how these readers fold to create protein domains that are very diverse in their tertiary structures, giving rise to intriguing peptide binding mechanisms resulting in vastly different footprints of their targets.
Collapse
Affiliation(s)
- Hossein Davarinejad
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Alexis Arvanitis-Vigneault
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Dallas Nygard
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Mathieu Lavallée-Adam
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Jean-François Couture
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
44
|
Nussinov R, Liu Y, Zhang W, Jang H. Protein conformational ensembles in function: roles and mechanisms. RSC Chem Biol 2023; 4:850-864. [PMID: 37920394 PMCID: PMC10619138 DOI: 10.1039/d3cb00114h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/02/2023] [Indexed: 11/04/2023] Open
Abstract
The sequence-structure-function paradigm has dominated twentieth century molecular biology. The paradigm tacitly stipulated that for each sequence there exists a single, well-organized protein structure. Yet, to sustain cell life, function requires (i) that there be more than a single structure, (ii) that there be switching between the structures, and (iii) that the structures be incompletely organized. These fundamental tenets called for an updated sequence-conformational ensemble-function paradigm. The powerful energy landscape idea, which is the foundation of modernized molecular biology, imported the conformational ensemble framework from physics and chemistry. This framework embraces the recognition that proteins are dynamic and are always interconverting between conformational states with varying energies. The more stable the conformation the more populated it is. The changes in the populations of the states are required for cell life. As an example, in vivo, under physiological conditions, wild type kinases commonly populate their more stable "closed", inactive, conformations. However, there are minor populations of the "open", ligand-free states. Upon their stabilization, e.g., by high affinity interactions or mutations, their ensembles shift to occupy the active states. Here we discuss the role of conformational propensities in function. We provide multiple examples of diverse systems, including protein kinases, lipid kinases, and Ras GTPases, discuss diverse conformational mechanisms, and provide a broad outlook on protein ensembles in the cell. We propose that the number of molecules in the active state (inactive for repressors), determine protein function, and that the dynamic, relative conformational propensities, rather than the rigid structures, are the hallmark of cell life.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University Tel Aviv 69978 Israel
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Wengang Zhang
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| |
Collapse
|
45
|
Kefalas G, Rotin D. Primate-specific isoform of Nedd4-1 regulates substrate binding via Ser/Thr phosphorylation and 14-3-3 binding. Sci Rep 2023; 13:17903. [PMID: 37863970 PMCID: PMC10589272 DOI: 10.1038/s41598-023-44761-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023] Open
Abstract
Nedd4 (Nedd4-1) is an E3 ubiquitin ligase involved in crucial biological processes such as growth factor receptor signaling. While canonical Nedd4-1 comprises a C2-WW(4)-HECT domain architecture, alternative splicing produces non-canonical isoforms that are poorly characterized. Here we characterized Nedd4-1(NE), a primate-specific isoform of Nedd4-1 that contains a large N-terminal Extension (NE) that replaces most of the C2 domain. We show that Nedd4-1(NE) mRNA is ubiquitously expressed in human tissues and cell lines. Moreover, we found that Nedd4-1(NE) is more active than the canonical Nedd4-1 isoform, likely due to the absence of a C2 domain-mediated autoinhibitory mechanism. Additionally, we identified two Thr/Ser phosphoresidues in the NE region that act as binding sites for 14-3-3 proteins, and show that phosphorylation on these sites reduces substrate binding. Finally, we show that the NE region can act as a binding site for the RPB2 subunit of RNA polymerase II, a unique substrate of Nedd4-1(NE) but not the canonical Nedd4-1. Taken together, our results demonstrate that alternative splicing of the ubiquitin ligase Nedd4-1 can produce isoforms that differ in their catalytic activity, binding partners and substrates, and mechanisms of regulation.
Collapse
Affiliation(s)
- George Kefalas
- Cell Biology Program, the Hospital for Sick Children, PGCRL 19-9715, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Biochemistry Department, University of Toronto, Toronto, ON, M5G 0A4, Canada
| | - Daniela Rotin
- Cell Biology Program, the Hospital for Sick Children, PGCRL 19-9715, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
- Biochemistry Department, University of Toronto, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
46
|
Vetoshkina D, Borisova-Mubarakshina M. Reversible protein phosphorylation in higher plants: focus on state transitions. Biophys Rev 2023; 15:1079-1093. [PMID: 37974979 PMCID: PMC10643769 DOI: 10.1007/s12551-023-01116-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/10/2023] [Indexed: 11/19/2023] Open
Abstract
Reversible protein phosphorylation is one of the comprehensive mechanisms of cell metabolism regulation in eukaryotic organisms. The review describes the impact of the reversible protein phosphorylation on the regulation of growth and development as well as in adaptation pathways and signaling network in higher plant cells. The main part of the review is devoted to the role of the reversible phosphorylation of light-harvesting proteins of photosystem II and the state transition process in fine-tuning the photosynthetic activity of chloroplasts. A separate section of the review is dedicated to comparing the mechanisms and functional significance of state transitions in higher plants, algae, and cyanobacteria that allows the evolution aspects of state transitions meaning in various organisms to be discussed. Environmental factors affecting the state transitions are also considered. Additionally, we gain insight into the possible influence of STN7-dependent phosphorylation of the target proteins on the global network of reversible protein phosphorylation in plant cells as well as into the probable effect of the STN7 kinase inhibition on long-term acclimation pathways in higher plants.
Collapse
Affiliation(s)
- D.V. Vetoshkina
- Institute of Basic Biological Problems of the Russian Academy of Sciences, Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya st., 2, Pushchino, Russia
| | - M.M. Borisova-Mubarakshina
- Institute of Basic Biological Problems of the Russian Academy of Sciences, Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya st., 2, Pushchino, Russia
| |
Collapse
|
47
|
Zhu R, Cao B, Sun M, Wu J, Li J. Genome-Wide Identification and Evolution of the GRF Gene Family and Functional Characterization of PbGRF18 in Pear. Int J Mol Sci 2023; 24:14690. [PMID: 37834136 PMCID: PMC10572701 DOI: 10.3390/ijms241914690] [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: 08/30/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Proteins encoded by the G-box regulating factor (GRF, also called 14-3-3) gene family are involved in protein-protein interactions and mediate signaling transduction, which play important roles in plant growth, development, and stress responses. However, there were no detailed investigations of the GRF gene family in pear at present. In this study, we identified 25 GRF family members in the pear genome. Based on a phylogenetic analysis, the 25 GRF genes were clustered into two groups; the ε group and the non-ε group. Analyses of the exon-intron structures and motifs showed that the gene structures were conserved within each of the ε and non-ε groups. Gene duplication analysis indicated that most of the PbGRF gene expansion that occurred in both groups was due to WGD/segmental duplication. Phosphorylation sites analysis showed that the main phosphorylation sites of PbGRF proteins were serine residues. For gene expression, five PbGRF genes (PbGRF7, PbGRF11, PbGRF16, PbGRF21, and PbGRF23) were highly expressed in fruits, and PbGRF18 was highly expressed in all tissues. Further analysis revealed that eight PbGRF genes were significantly differentially expressed after treatment with different sugars; the expression of PbGRF7, PbGRF8, and PbGRF11 significantly increased, implying the involvement of these genes in sugar signaling. In addition, subcellular localization studies showed that the tested GRF proteins localize to the plasma membrane, and transgenic analysis showed that PbGRF18 can increase the sugar content in tomato leaves and fruit. The results of our research establish a foundation for functional determination of PbGRF proteins, and will help to promote a further understanding of the regulatory network in pear fruit development.
Collapse
Affiliation(s)
- Rongxiang Zhu
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China (M.S.)
- Guangxi Key Laboratory of Plant Functional Phytochemicals and Sustainable Utilization, Guangxi Institute of Botany, Guangxi Zhuang Autonomous Region and Chinese Academy of Sciences, Guilin 541006, China
| | - Beibei Cao
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China (M.S.)
| | - Manyi Sun
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China (M.S.)
| | - Jun Wu
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China (M.S.)
- Zhongshan Biological Breeding Laboratory, Nanjing 210014, China
| | - Jiaming Li
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China (M.S.)
- Academy for Advanced Interdisciplinary Studies, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
48
|
Barone M, Ramayo-Caldas Y, Estellé J, Tambosco K, Chadi S, Maillard F, Gallopin M, Planchais J, Chain F, Kropp C, Rios-Covian D, Sokol H, Brigidi P, Langella P, Martín R. Gut barrier-microbiota imbalances in early life lead to higher sensitivity to inflammation in a murine model of C-section delivery. MICROBIOME 2023; 11:140. [PMID: 37394428 PMCID: PMC10316582 DOI: 10.1186/s40168-023-01584-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 05/25/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Most interactions between the host and its microbiota occur at the gut barrier, and primary colonizers are essential in the gut barrier maturation in the early life. The mother-offspring transmission of microorganisms is the most important factor influencing microbial colonization in mammals, and C-section delivery (CSD) is an important disruptive factor of this transfer. Recently, the deregulation of symbiotic host-microbe interactions in early life has been shown to alter the maturation of the immune system, predisposing the host to gut barrier dysfunction and inflammation. The main goal of this study is to decipher the role of the early-life gut microbiota-barrier alterations and its links with later-life risks of intestinal inflammation in a murine model of CSD. RESULTS The higher sensitivity to chemically induced inflammation in CSD mice is related to excessive exposure to a too diverse microbiota too early in life. This early microbial stimulus has short-term consequences on the host homeostasis. It switches the pup's immune response to an inflammatory context and alters the epithelium structure and the mucus-producing cells, disrupting gut homeostasis. This presence of a too diverse microbiota in the very early life involves a disproportionate short-chain fatty acids ratio and an excessive antigen exposure across the vulnerable gut barrier in the first days of life, before the gut closure. Besides, as shown by microbiota transfer experiments, the microbiota is causal in the high sensitivity of CSD mice to chemical-induced colitis and in most of the phenotypical parameters found altered in early life. Finally, supplementation with lactobacilli, the main bacterial group impacted by CSD in mice, reverts the higher sensitivity to inflammation in ex-germ-free mice colonized by CSD pups' microbiota. CONCLUSIONS Early-life gut microbiota-host crosstalk alterations related to CSD could be the linchpin behind the phenotypic effects that lead to increased susceptibility to an induced inflammation later in life in mice. Video Abstract.
Collapse
Affiliation(s)
- M. Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Y. Ramayo-Caldas
- INRAE, AgroParisTech, GABI, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - J. Estellé
- INRAE, AgroParisTech, GABI, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - K. Tambosco
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - S. Chadi
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - F. Maillard
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - M. Gallopin
- CNRS, CEA, l’Institut de Biologie Intégrative de La Cellule (I2BC), Paris-Saclay University, 91405 Orsay, France
| | - J. Planchais
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - F. Chain
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - C. Kropp
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - D. Rios-Covian
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - H. Sokol
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Gastroenterology Department, Centre de Recherche Saint-Antoine, Centre de Recherche Saint-Antoine, CRSA, AP-HP, INSERM, Saint Antoine Hospital, Sorbonne Université, 75012 Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - P. Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - P. Langella
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - R. Martín
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| |
Collapse
|
49
|
Dong X, Feng F, Li Y, Li L, Chen S, Zhou JM. 14-3-3 proteins facilitate the activation of MAP kinase cascades by upstream immunity-related kinases. THE PLANT CELL 2023; 35:2413-2428. [PMID: 36943771 PMCID: PMC10226567 DOI: 10.1093/plcell/koad088] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/24/2023] [Accepted: 02/14/2023] [Indexed: 05/30/2023]
Abstract
Activation of mitogen-activated protein kinase (MAP kinase) cascades is essential for plant immunity. Upon activation by surface-localized immune receptors, receptor-like cytoplasmic kinases (RLCKs) in the cytoplasm phosphorylate MAP kinase kinase kinases (MAPKKKs) to initiate MAP kinase activation. Surprisingly, we found that both the phosphorylation of Arabidopsis (Arabidopsis thaliana) MAPKKKs and the subsequent activation of MAP kinase cascades require the λ and κ isoforms of 14-3-3 proteins, which directly interact with multiple RLCKs and MAPKKKs. The N- and C-termini of MAPKKK5 interact intramolecularly to inhibit the access to the C terminus by RLCKs, whereas the 14-3-3 proteins relieve this inhibition and facilitate the interaction of RLCKs with the C-terminus of MAPKKK5. This enables the phosphorylation of MAPKK5 at Ser599 and Ser682, thus promoting MAP kinase activation and enhancing plant disease resistance. Our study reveals a role of 14-3-3 proteins as scaffolds and activators in the regulation of the RLCK-MAPKKK5 module and provides insight into the mechanism of plant immune signaling.
Collapse
Affiliation(s)
- Xiaojing Dong
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Feng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Yangjun Li
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Li
- National Institute of Biological Sciences, Beijing 102206, China
| | - She Chen
- National Institute of Biological Sciences, Beijing 102206, China
| | - Jian-Min Zhou
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing 100101, China
- Chinese Academy of Sciences Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
- Hainan Yazhou Bay Seed Laboratory, Sanya, Hainan 572025, China
| |
Collapse
|
50
|
Sheikh AH, Zacharia I, Pardal AJ, Dominguez-Ferreras A, Sueldo DJ, Kim JG, Balmuth A, Gutierrez JR, Conlan BF, Ullah N, Nippe OM, Girija AM, Wu CH, Sessa G, Jones AME, Grant MR, Gifford ML, Mudgett MB, Rathjen JP, Ntoukakis V. Dynamic changes of the Prf/Pto tomato resistance complex following effector recognition. Nat Commun 2023; 14:2568. [PMID: 37142566 PMCID: PMC10160066 DOI: 10.1038/s41467-023-38103-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 04/16/2023] [Indexed: 05/06/2023] Open
Abstract
In both plants and animals, nucleotide-binding leucine-rich repeat (NLR) immune receptors play critical roles in pathogen recognition and activation of innate immunity. In plants, NLRs recognise pathogen-derived effector proteins and initiate effector-triggered immunity (ETI). However, the molecular mechanisms that link NLR-mediated effector recognition and downstream signalling are not fully understood. By exploiting the well-characterised tomato Prf/Pto NLR resistance complex, we identified the 14-3-3 proteins TFT1 and TFT3 as interacting partners of both the NLR complex and the protein kinase MAPKKKα. Moreover, we identified the helper NRC proteins (NLR-required for cell death) as integral components of the Prf /Pto NLR recognition complex. Notably our studies revealed that TFTs and NRCs interact with distinct modules of the NLR complex and, following effector recognition, dissociate facilitating downstream signalling. Thus, our data provide a mechanistic link between activation of immune receptors and initiation of downstream signalling cascades.
Collapse
Affiliation(s)
- Arsheed H Sheikh
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Center for Desert Agriculture, BESE Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Iosif Zacharia
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Alonso J Pardal
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | | | - Daniela J Sueldo
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Hogskoleringen 1, 7491, Trondheim, Norway
| | - Jung-Gun Kim
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Alexi Balmuth
- J.R. Simplot Company, Boise, ID, USA
- The Sainsbury Laboratory, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Jose R Gutierrez
- The Sainsbury Laboratory, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Brendon F Conlan
- Research School of Biology, The Australian National University, Acton, 2601, ACT, Australia
| | - Najeeb Ullah
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Olivia M Nippe
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Anil M Girija
- School of Plant Sciences and Food Security, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Chih-Hang Wu
- The Sainsbury Laboratory, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Guido Sessa
- School of Plant Sciences and Food Security, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | | | - Murray R Grant
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Miriam L Gifford
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Warwick Integrative Synthetic Biology Centre, University of Warwick, Coventry, CV4 7AL, UK
| | - Mary Beth Mudgett
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - John P Rathjen
- Research School of Biology, The Australian National University, Acton, 2601, ACT, Australia
| | - Vardis Ntoukakis
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
- Warwick Integrative Synthetic Biology Centre, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|