1
|
Andlovic B, Valenti D, Centorrino F, Picarazzi F, Hristeva S, Hiltmann M, Wolf A, Cantrelle FX, Mori M, Landrieu I, Levy LM, Klebl B, Tzalis D, Genski T, Eickhoff J, Ottmann C. Fragment-Based Interrogation of the 14-3-3/TAZ Protein-Protein Interaction. Biochemistry 2024; 63:2196-2206. [PMID: 39172504 PMCID: PMC11375770 DOI: 10.1021/acs.biochem.4c00248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
The identification of chemical starting points for the development of molecular glues is challenging. Here, we employed fragment screening and identified an allosteric stabilizer of the complex between 14-3-3 and a TAZ-derived peptide. The fragment binds preferentially to the 14-3-3/TAZ peptide complex and shows moderate stabilization in differential scanning fluorimetry and microscale thermophoresis. The binding site of the fragment was predicted by molecular dynamics calculations to be distant from the 14-3-3/TAZ peptide interface, located between helices 8 and 9 of the 14-3-3 protein. This site was confirmed by nuclear magnetic resonance and X-ray protein crystallography, revealing the first example of an allosteric stabilizer for 14-3-3 protein-protein interactions.
Collapse
Affiliation(s)
- Blaž Andlovic
- Lead
Discovery Center GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Dario Valenti
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
- Taros
Chemicals GmbH & Co. KG, Emil-Figge-Straße 76a, 44227 Dortmund, Germany
| | - Federica Centorrino
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Francesca Picarazzi
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Stanimira Hristeva
- Taros
Chemicals GmbH & Co. KG, Emil-Figge-Straße 76a, 44227 Dortmund, Germany
| | | | - Alexander Wolf
- Lead
Discovery Center GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - François-Xavier Cantrelle
- CNRS
EMR9002 Integrative Structural Biology, University of Lille, F-59000 Lille, France
- University
of Lille, Inserm, Institut Pasteur de Lille, U1167—RID-AGE—Risk
Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
| | - Mattia Mori
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Isabelle Landrieu
- CNRS
EMR9002 Integrative Structural Biology, University of Lille, F-59000 Lille, France
- University
of Lille, Inserm, Institut Pasteur de Lille, U1167—RID-AGE—Risk
Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
| | - Laura M. Levy
- Taros
Chemicals GmbH & Co. KG, Emil-Figge-Straße 76a, 44227 Dortmund, Germany
| | - Bert Klebl
- Lead
Discovery Center GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Dimitrios Tzalis
- Taros
Chemicals GmbH & Co. KG, Emil-Figge-Straße 76a, 44227 Dortmund, Germany
| | - Thorsten Genski
- Taros
Chemicals GmbH & Co. KG, Emil-Figge-Straße 76a, 44227 Dortmund, Germany
| | - Jan Eickhoff
- Lead
Discovery Center GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Christian Ottmann
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
2
|
Somsen BA, Cossar PJ, Arkin MR, Brunsveld L, Ottmann C. 14-3-3 Protein-Protein Interactions: From Mechanistic Understanding to Their Small-Molecule Stabilization. Chembiochem 2024; 25:e202400214. [PMID: 38738787 DOI: 10.1002/cbic.202400214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/14/2024]
Abstract
Protein-protein interactions (PPIs) are of utmost importance for maintenance of cellular homeostasis. Herein, a central role can be found for 14-3-3 proteins. These hub-proteins are known to bind hundreds of interaction partners, thereby regulating their activity, localization, and/or stabilization. Due to their ability to bind a large variety of client proteins, studies of 14-3-3 protein complexes flourished over the last decades, aiming to gain greater molecular understanding of these complexes and their role in health and disease. Because of their crucial role within the cell, 14-3-3 protein complexes are recognized as highly interesting therapeutic targets, encouraging the discovery of small molecule modulators of these PPIs. We discuss various examples of 14-3-3-mediated regulation of its binding partners on a mechanistic level, highlighting the versatile and multi-functional role of 14-3-3 within the cell. Furthermore, an overview is given on the development of stabilizers of 14-3-3 protein complexes, from initially used natural products to fragment-based approaches. These studies show the potential of 14-3-3 PPI stabilizers as novel agents in drug discovery and as tool compounds to gain greater molecular understanding of the role of 14-3-3-based protein regulation.
Collapse
Affiliation(s)
- Bente A Somsen
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| | - Peter J Cossar
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California, 94143, United States
| | - Luc Brunsveld
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| | - Christian Ottmann
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. Box 513, MB Eindhoven, 5600, Eindhoven, The Netherlands
| |
Collapse
|
3
|
Konstantinidou M, Visser EJ, Vandenboorn E, Chen S, Jaishankar P, Overmans M, Dutta S, Neitz RJ, Renslo AR, Ottmann C, Brunsveld L, Arkin MR. Structure-Based Optimization of Covalent, Small-Molecule Stabilizers of the 14-3-3σ/ERα Protein-Protein Interaction from Nonselective Fragments. J Am Chem Soc 2023; 145:20328-20343. [PMID: 37676236 PMCID: PMC10515640 DOI: 10.1021/jacs.3c05161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Indexed: 09/08/2023]
Abstract
The stabilization of protein-protein interactions (PPIs) has emerged as a promising strategy in chemical biology and drug discovery. The identification of suitable starting points for stabilizing native PPIs and their subsequent elaboration into selective and potent molecular glues lacks structure-guided optimization strategies. We have previously identified a disulfide fragment that stabilized the hub protein 14-3-3σ bound to several of its clients, including ERα and C-RAF. Here, we show the structure-based optimization of the nonselective fragment toward selective and highly potent small-molecule stabilizers of the 14-3-3σ/ERα complex. The more elaborated molecular glues, for example, show no stabilization of 14-3-3σ/C-RAF up to 150 μM compound. Orthogonal biophysical assays, including mass spectrometry and fluorescence anisotropy, were used to establish structure-activity relationships. The binding modes of 37 compounds were elucidated with X-ray crystallography, which further assisted the concomitant structure-guided optimization. By targeting specific amino acids in the 14-3-3σ/ERα interface and locking the conformation with a spirocycle, the optimized covalent stabilizer 181 achieved potency, cooperativity, and selectivity similar to the natural product Fusicoccin-A. This case study showcases the value of addressing the structure, kinetics, and cooperativity for molecular glue development.
Collapse
Affiliation(s)
- Markella Konstantinidou
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| | - Emira J. Visser
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Edmee Vandenboorn
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Sheng Chen
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| | - Priyadarshini Jaishankar
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| | - Maurits Overmans
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Shubhankar Dutta
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| | - R. Jeffrey Neitz
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| | - Adam R. Renslo
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| | - Christian Ottmann
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Michelle R. Arkin
- Department
of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, California 94143, United States
| |
Collapse
|
4
|
Visser EJ, Jaishankar P, Sijbesma E, Pennings MAM, Vandenboorn EMF, Guillory X, Neitz RJ, Morrow J, Dutta S, Renslo AR, Brunsveld L, Arkin MR, Ottmann C. From Tethered to Freestanding Stabilizers of 14-3-3 Protein-Protein Interactions through Fragment Linking. Angew Chem Int Ed Engl 2023; 62:e202308004. [PMID: 37455289 DOI: 10.1002/anie.202308004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Small-molecule stabilization of protein-protein interactions (PPIs) is a promising strategy in chemical biology and drug discovery. However, the systematic discovery of PPI stabilizers remains a largely unmet challenge. Herein we report a fragment-linking approach targeting the interface of 14-3-3 and a peptide derived from the estrogen receptor alpha (ERα) protein. Two classes of fragments-a covalent and a noncovalent fragment-were co-crystallized and subsequently linked, resulting in a noncovalent hybrid molecule in which the original fragment interactions were largely conserved. Supported by 20 crystal structures, this initial hybrid molecule was further optimized, resulting in selective, 25-fold stabilization of the 14-3-3/ERα interaction. The high-resolution structures of both the single fragments, their co-crystal structures and those of the linked fragments document a feasible strategy to develop orthosteric PPI stabilizers by linking to an initial tethered fragment.
Collapse
Affiliation(s)
- Emira J Visser
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| | - Priyadarshini Jaishankar
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| | - Marloes A M Pennings
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| | - Edmee M F Vandenboorn
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| | - Xavier Guillory
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| | - R Jeffrey Neitz
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - John Morrow
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Shubhankar Dutta
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ Eindhoven, The Netherlands
| |
Collapse
|
5
|
Andlovic B, Heilmann G, Ninck S, Andrei SA, Centorrino F, Higuchi Y, Kato N, Brunsveld L, Arkin M, Menninger S, Choidas A, Wolf A, Klebl B, Kaschani F, Kaiser M, Eickhoff J, Ottmann C. IFNα primes cancer cells for Fusicoccin-induced cell death via 14-3-3 PPI stabilization. Cell Chem Biol 2023; 30:573-590.e6. [PMID: 37130519 DOI: 10.1016/j.chembiol.2023.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 02/02/2023] [Accepted: 04/06/2023] [Indexed: 05/04/2023]
Abstract
The natural product family of the fusicoccanes (FCs) has been shown to display anti-cancer activity, especially when combined with established therapeutic agents. FCs stabilize 14-3-3 protein-protein interactions (PPIs). Here, we tested combinations of a small library of FCs with interferon α (IFNα) on different cancer cell lines and report a proteomics approach to identify the specific 14-3-3 PPIs that are induced by IFNα and stabilized by FCs in OVCAR-3 cells. Among the identified 14-3-3 target proteins are THEMIS2, receptor interacting protein kinase 2 (RIPK2), EIF2AK2, and several members of the LDB1 complex. Biophysical and structural biology studies confirm these 14-3-3 PPIs as physical targets of FC stabilization, and transcriptome as well as pathway analyses suggest possible explanations for the observed synergistic effect of IFNα/FC treatment on cancer cells. This study elucidates the polypharmacological effects of FCs in cancer cells and identifies potential targets from the vast interactome of 14-3-3s for therapeutic intervention in oncology.
Collapse
Affiliation(s)
- Blaž Andlovic
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, the Netherlands; Lead Discovery Center GmbH, 44227 Dortmund, Germany
| | - Geronimo Heilmann
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, 45117 Essen, Germany
| | - Sabrina Ninck
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, 45117 Essen, Germany
| | - Sebastian A Andrei
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, the Netherlands
| | - Federica Centorrino
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, the Netherlands
| | - Yusuke Higuchi
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Ibaraki, Japan
| | - Nobuo Kato
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Ibaraki, Japan
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, the Netherlands
| | - Michelle Arkin
- Small Molecule Discovery Center and Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Axel Choidas
- Lead Discovery Center GmbH, 44227 Dortmund, Germany
| | | | - Bert Klebl
- Lead Discovery Center GmbH, 44227 Dortmund, Germany
| | - Farnusch Kaschani
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, 45117 Essen, Germany
| | - Markus Kaiser
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, 45117 Essen, Germany
| | - Jan Eickhoff
- Lead Discovery Center GmbH, 44227 Dortmund, Germany
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, the Netherlands.
| |
Collapse
|
6
|
Egbert CM, Warr LR, Pennington KL, Thornton MM, Vaughan AJ, Ashworth SW, Heaton MJ, English N, Torres MP, Andersen JL. The Integration of Proteome-Wide PTM Data with Protein Structural and Sequence Features Identifies Phosphorylations that Mediate 14-3-3 Interactions. J Mol Biol 2023; 435:167890. [PMID: 36402225 PMCID: PMC10099770 DOI: 10.1016/j.jmb.2022.167890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/18/2022] [Accepted: 11/07/2022] [Indexed: 11/18/2022]
Abstract
14-3-3s are abundant proteins that regulate essentially all aspects of cell biology, including cell cycle, motility, metabolism, and cell death. 14-3-3s work by docking to phosphorylated Ser/Thr residues on a large network of client proteins and modulating client protein function in a variety of ways. In recent years, aided by improvements in proteomics, the discovery of 14-3-3 client proteins has far outpaced our ability to understand the biological impact of individual 14-3-3 interactions. The rate-limiting step in this process is often the identification of the individual phospho-serines/threonines that mediate 14-3-3 binding, which are difficult to distinguish from other phospho-sites by sequence alone. Furthermore, trial-and-error molecular approaches to identify these phosphorylations are costly and can take months or years to identify even a single 14-3-3 docking site phosphorylation. To help overcome this challenge, we used machine learning to analyze predictive features of 14-3-3 binding sites. We found that accounting for intrinsic protein disorder and the unbiased mass spectrometry identification rate of a given phosphorylation significantly improves the identification of 14-3-3 docking site phosphorylations across the proteome. We incorporated these features, coupled with consensus sequence prediction, into a publicly available web app, called "14-3-3 site-finder". We demonstrate the strength of this approach through its ability to identify 14-3-3 binding sites that do not conform to the loose consensus sequence of 14-3-3 docking phosphorylations, which we validate with 14-3-3 client proteins, including TNK1, CHEK1, MAPK7, and others. In addition, by using this approach, we identify a phosphorylation on A-kinase anchor protein-13 (AKAP13) at Ser2467 that dominantly controls its interaction with 14-3-3.
Collapse
Affiliation(s)
- C M Egbert
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - L R Warr
- Department of Statistics, Brigham Young University, Provo, UT, USA
| | - K L Pennington
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA; Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - M M Thornton
- Department of Computer Science, Brigham Young University, Provo, UT, USA
| | - A J Vaughan
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - S W Ashworth
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - M J Heaton
- Department of Statistics, Brigham Young University, Provo, UT, USA
| | - N English
- Quantitative Bioscience Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - M P Torres
- Quantitative Bioscience Program, Georgia Institute of Technology, Atlanta, GA, USA; School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - J L Andersen
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
7
|
Chatterjee C, Singh SK. Peptide and protein chemistry approaches to study the tumor suppressor protein p53. Org Biomol Chem 2022; 20:5500-5509. [PMID: 35786742 PMCID: PMC10112546 DOI: 10.1039/d2ob00902a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The tumor suppressor and master gene regulator protein p53 has been the subject of intense investigation for several decades due to its mutation in about half of all human cancers. However, mechanistic studies of p53 in cells are complicated by its many dynamic binding partners and heterogeneous post-translational modifications. The design of therapeutics that rescue p53 functions in cells requires a mechanistic understanding of its protein-protein interactions in specific protein complexes and identifying changes in p53 activity by diverse post-translational modifications. This review highlights the important roles that peptide and protein chemistry have played in biophysical and biochemical studies aimed at elucidating p53 regulation by several key binding partners. The design of various peptide inhibitors that rescue p53 function in cells and new opportunities in targeting p53-protein interactions are discussed. In addition, the review highlights the importance of a protein semisynthesis approach to comprehend the role of site-specific PTMs in p53 regulation.
Collapse
Affiliation(s)
- Champak Chatterjee
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA.
| | - Sumeet K Singh
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
8
|
Abstract
AbstractThe druggable genome is limited by structural features that can be targeted by small molecules in disease-relevant proteins. While orthosteric and allosteric protein modulators have been well studied, they are limited to antagonistic/agonistic functions. This approach to protein modulation leaves many disease-relevant proteins as undruggable targets. Recently, protein-protein interaction modulation has emerged as a promising therapeutic field for previously undruggable protein targets. Molecular glues and heterobifunctional degraders such as PROTACs can facilitate protein interactions and bring the proteasome into proximity to induce targeted protein degradation. In this review, we discuss the function and rational design of molecular glues, heterobifunctional degraders, and hydrophobic tag degraders. We also review historic and novel molecular glues and targets and discuss the challenges and opportunities in this new therapeutic field.
Collapse
|
9
|
Most Probable Druggable Pockets in Mutant p53-Arg175His Clusters Extracted from Gaussian Accelerated Molecular Dynamics Simulations. Protein J 2022; 41:27-43. [PMID: 35099676 DOI: 10.1007/s10930-022-10041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 12/24/2022]
Abstract
p53, a tumor suppressor protein, is essential for preventing cancer development. Enhancing our understanding of the human p53 function and its modifications in carcinogenesis will aid in developing more highly effective strategies for cancer prevention and treatment. In this study, we have modeled five human p53 forms, namely, inactive, distal-active, proximal-active, distal-Arg175His mutant, and proximal-Arg175His mutant forms. These forms have been investigated using Gaussian accelerated molecular dynamics (GaMD) simulations in OPC water model at physiological temperature and pH. Our observations, obtained throughout [Formula: see text] of production run, are in good agreement with the relevant results in the classical molecular dynamics (MD) studies. Therefore, GaMD method is more economic and efficient method than the classical MD method for studying biomolecular systems. The featured dynamics of the five human p53-DBD forms include noticeable conformational changes of L1 and [Formula: see text]-[Formula: see text] loops as well as [Formula: see text]-[Formula: see text] and [Formula: see text]-[Formula: see text] turns. We have identified two clusters that represent two distinct conformational states in each p53-DBD form. The free-energy profiles of these clusters demonstrate the flexibility of the protein to undergo a conformational transition between the two clusters. We have predicted two out of seven possible druggability pockets on the clusters of the Arg175His forms. These two druggability pockets are near the mutation site and are expected to be actual pockets, which will be helpful for the compound clinical progression studies.
Collapse
|
10
|
Sijbesma E, Hallenbeck KK, Andrei SA, Rust RR, Adriaans JMC, Brunsveld L, Arkin MR, Ottmann C. Exploration of a 14-3-3 PPI Pocket by Covalent Fragments as Stabilizers. ACS Med Chem Lett 2021; 12:976-982. [PMID: 34136078 PMCID: PMC8201753 DOI: 10.1021/acsmedchemlett.1c00088] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
![]()
The systematic discovery
of functional fragments binding to the
composite interface of protein complexes is a first critical step
for the development of orthosteric stabilizers of protein–protein
interactions (PPIs). We have previously shown that disulfide trapping
successfully yielded covalent stabilizers for the PPI of 14-3-3 with
the estrogen receptor ERα. Here we provide an assessment of
the composite PPI target pocket and the molecular characteristics
of various fragments binding to a specific subpocket. Evaluating structure–activity
relationships highlights the basic principles for PPI stabilization
by these covalent fragments that engage a relatively large and exposed
binding pocket at the protein/peptide interface with a “molecular
glue” mode of action.
Collapse
Affiliation(s)
- Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Kenneth K. Hallenbeck
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco 94134, United States
| | - Sebastian A. Andrei
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Reanne R. Rust
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Joris M. C. Adriaans
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco 94134, United States
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
11
|
Kuusk A, Boyd H, Chen H, Ottmann C. Small-molecule modulation of p53 protein-protein interactions. Biol Chem 2021; 401:921-931. [PMID: 32049643 DOI: 10.1515/hsz-2019-0405] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022]
Abstract
Small-molecule modulation of protein-protein interactions (PPIs) is a very promising but also challenging area in drug discovery. The tumor suppressor protein p53 is one of the most frequently altered proteins in human cancers, making it an attractive target in oncology. 14-3-3 proteins have been shown to bind to and positively regulate p53 activity by protecting it from MDM2-dependent degradation or activating its DNA binding affinity. PPIs can be modulated by inhibiting or stabilizing specific interactions by small molecules. Whereas inhibition has been widely explored by the pharmaceutical industry and academia, the opposite strategy of stabilizing PPIs still remains relatively underexploited. This is rather interesting considering the number of natural compounds like rapamycin, forskolin and fusicoccin that exert their activity by stabilizing specific PPIs. In this review, we give an overview of 14-3-3 interactions with p53, explain isoform specific stabilization of the tumor suppressor protein, explore the approach of stabilizing the 14-3-3σ-p53 complex and summarize some promising small molecules inhibiting the p53-MDM2 protein-protein interaction.
Collapse
Affiliation(s)
- Ave Kuusk
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, NL-5600MB Eindhoven, The Netherlands
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, S-43183 Mölndal, Sweden
| | - Helen Boyd
- Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| | - Hongming Chen
- Guangzhou Regenerative Medicine and Health-Guangdong Laboratory, Science Park, Guangzhou 510530, China
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, NL-5600MB Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, D-45141 Essen, Germany
| |
Collapse
|
12
|
Sugita K, Kuwabara M, Matsuo A, Kamo S, Matsuzawa A. Stereoselective Convergent Synthesis of Carbon Skeleton of Cotylenin A Aglycone. SYNTHESIS-STUTTGART 2021. [DOI: 10.1055/s-0040-1706684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
AbstractIn this paper, the synthesis of the carbon skeleton of cotylenin A aglycone is described. The key reactions, including an intramolecular aldol reaction, an aldol coupling reaction, and a ring-closing metathesis, allow for the effective and stereoselective access to the carbon skeleton of cotylenin A aglycone. The stereochemistry was confirmed by single-crystal X-ray crystallographic analyses of related compounds.
Collapse
|
13
|
Gencel-Augusto J, Lozano G. p53 tetramerization: at the center of the dominant-negative effect of mutant p53. Genes Dev 2021; 34:1128-1146. [PMID: 32873579 PMCID: PMC7462067 DOI: 10.1101/gad.340976.120] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, Gencel-Augusto and Lozano summarize the data on p53 mutants with a functional tetramerization domain that form mixed tetramers and in some cases have dominant-negative effects (DNE) that inactivate wild-type p53. They conclude that the DNE is mostly observed after DNA damage but fails in other contexts. The p53 tumor suppressor functions as a tetrameric transcription factor to regulate hundreds of genes—many in a tissue-specific manner. Missense mutations in cancers in the p53 DNA-binding and tetramerization domains cement the importance of these domains in tumor suppression. p53 mutants with a functional tetramerization domain form mixed tetramers, which in some cases have dominant-negative effects (DNE) that inactivate wild-type p53. DNA damage appears necessary but not sufficient for DNE, indicating that upstream signals impact DNE. Posttranslational modifications and protein–protein interactions alter p53 tetramerization affecting transcription, stability, and localization. These regulatory components limit the dominant-negative effects of mutant p53 on wild-type p53 activity. A deeper understanding of the molecular basis for DNE may drive development of drugs that release WT p53 and allow tumor suppression.
Collapse
Affiliation(s)
- Jovanka Gencel-Augusto
- Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas 77030, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Guillermina Lozano
- Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas 77030, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
14
|
Falcicchio M, Ward JA, Macip S, Doveston RG. Regulation of p53 by the 14-3-3 protein interaction network: new opportunities for drug discovery in cancer. Cell Death Discov 2020; 6:126. [PMID: 33298896 PMCID: PMC7669891 DOI: 10.1038/s41420-020-00362-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/02/2020] [Accepted: 10/23/2020] [Indexed: 01/17/2023] Open
Abstract
Most cancers evolve to disable the p53 pathway, a key tumour suppressor mechanism that prevents transformation and malignant cell growth. However, only ~50% exhibit inactivating mutations of p53, while in the rest its activity is suppressed by changes in the proteins that modulate the pathway. Therefore, restoring p53 activity in cells in which it is still wild type is a highly attractive therapeutic strategy that could be effective in many different cancer types. To this end, drugs can be used to stabilise p53 levels by modulating its regulatory pathways. However, despite the emergence of promising strategies, drug development has stalled in clinical trials. The need for alternative approaches has shifted the spotlight to the 14-3-3 family of proteins, which strongly influence p53 stability and transcriptional activity through direct and indirect interactions. Here, we present the first detailed review of how 14-3-3 proteins regulate p53, with special emphasis on the mechanisms involved in their binding to different members of the pathway. This information will be important to design new compounds that can reactivate p53 in cancer cells by influencing protein-protein interactions. The intricate relationship between the 14-3-3 isoforms and the p53 pathway suggests that many potential drug targets for p53 reactivation could be identified and exploited to design novel antineoplastic therapies with a wide range of applications.
Collapse
Affiliation(s)
- Marta Falcicchio
- Leicester Institute for Structural and Chemical Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK
- School of Chemistry, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - Jake A Ward
- Leicester Institute for Structural and Chemical Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - Salvador Macip
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK.
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain.
| | - Richard G Doveston
- Leicester Institute for Structural and Chemical Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK.
- School of Chemistry, University of Leicester, University Road, Leicester, LE1 7RH, UK.
| |
Collapse
|
15
|
Ballone A, Lau RA, Zweipfenning FPA, Ottmann C. A new soaking procedure for X-ray crystallographic structural determination of protein-peptide complexes. Acta Crystallogr F Struct Biol Commun 2020; 76:501-507. [PMID: 33006579 PMCID: PMC7531243 DOI: 10.1107/s2053230x2001122x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/16/2020] [Indexed: 11/29/2022] Open
Abstract
Interactions between a protein and a peptide motif of its protein partner are prevalent in nature. Often, a protein also has multiple interaction partners. X-ray protein crystallography is commonly used to examine these interactions in terms of bond distances and angles as well as to describe hotspots within protein complexes. However, the crystallization process presents a significant bottleneck in structure determination since it often requires notably time-consuming screening procedures, which involve testing a broad range of crystallization conditions via a trial-and-error approach. This difficulty is also increased as each protein-peptide complex does not necessarily crystallize under the same conditions. Here, a new co-crystallization/peptide-soaking method is presented which circumvents the need to return to the initial lengthy crystal screening and optimization processes for each consequent new complex. The 14-3-3σ protein, which has multiple interacting partners with specific peptidic motifs, was used as a case study. It was found that co-crystals of 14-3-3σ and a low-affinity peptide from one of its partners, c-Jun, could easily be soaked with another interacting peptide to quickly and easily generate new structures at high resolution. Not only does this significantly reduce the production time, but new 14-3-3-peptide structures that were previously not accessible with the 14-3-3σ isoform, despite screening hundreds of other different conditions, were now also able to be resolved. The findings achieved in this study may be considered as a supporting and practical guide to potentially enable the acceleration of the crystallization process of any protein-peptide system.
Collapse
Affiliation(s)
- Alice Ballone
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Roxanne A. Lau
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Fabian P. A. Zweipfenning
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| |
Collapse
|
16
|
Nadel CM, Ran X, Gestwicki JE. Luminescence complementation assay for measurement of binding to protein C-termini in live cells. Anal Biochem 2020; 611:113947. [PMID: 32918866 DOI: 10.1016/j.ab.2020.113947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 11/28/2022]
Abstract
Protein-protein interactions (PPIs) involving the extreme C-terminus serve important scaffolding and regulatory functions. Here, we leveraged NanoBiT technology to build a luminescent complementation assay for use in studying this subcategory of PPI. As a model system, we fused one component of NanoBiT to the disordered C-terminus of heat shock protein (Hsp70) and the other to its binding partner, the tetratricopeptide repeat (TPR) domain of CHIP/STUB1. We found that HEK293 cells that stably express these chimeras under a doxycycline promoter produced a robust luminescence signal. This signal was sensitive to mutations and it was further tuned by the expression of competitive C-termini. Using this system, we identified a promising, membrane permeable inhibitor of the Hsp70-CHIP interaction. More broadly, we anticipate that NanoBiT is well-suited for studying PPIs that involve C-termini.
Collapse
Affiliation(s)
- Cory M Nadel
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Xu Ran
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
17
|
Yuan S, Gopal JV, Ren S, Chen L, Liu L, Gao Z. Anticancer fungal natural products: Mechanisms of action and biosynthesis. Eur J Med Chem 2020; 202:112502. [PMID: 32652407 DOI: 10.1016/j.ejmech.2020.112502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
Abstract
Many fungal metabolites show promising anticancer properties both in vitro and in animal models, and some synthetic analogs of those metabolites have progressed into clinical trials. However, currently, there are still no fungi-derived agents approved as anticancer drugs. Two potential reasons could be envisioned: 1) lacking a clear understanding of their anticancer mechanism of action, 2) unable to supply enough materials to support the preclinical and clinic developments. In this review, we will summarize recent efforts on elucidating the anticancer mechanisms and biosynthetic pathways of several promising anticancer fungal natural products.
Collapse
Affiliation(s)
- Siwen Yuan
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jannu Vinay Gopal
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shuya Ren
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Litong Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China
| | - Zhizeng Gao
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China.
| |
Collapse
|
18
|
Wolter M, de Vink P, Neves JF, Srdanović S, Higuchi Y, Kato N, Wilson A, Landrieu I, Brunsveld L, Ottmann C. Selectivity via Cooperativity: Preferential Stabilization of the p65/14-3-3 Interaction with Semisynthetic Natural Products. J Am Chem Soc 2020; 142:11772-11783. [PMID: 32501683 PMCID: PMC8022324 DOI: 10.1021/jacs.0c02151] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Natural
compounds are an important class of potent drug molecules
including some retrospectively found to act as stabilizers of protein–protein
interactions (PPIs). However, the design of synthetic PPI stabilizers
remains an understudied approach. To date, there are limited examples
where cooperativity has been utilized to guide the optimization of
a PPI stabilizer. The 14-3-3 scaffold proteins provide an excellent
platform to explore PPI stabilization because these proteins mediate
several hundred PPIs, and a class of natural compounds, the fusicoccanes,
are known to stabilize a subset of 14-3-3 protein interactions. 14-3-3
has been reported to negatively regulate the p65 subunit of the NF-κB
transcription factor, which qualifies this protein complex as a potential
target for drug discovery to control cell proliferation. Here, we
report the high-resolution crystal structures of two 14-3-3 binding
motifs of p65 in complex with 14-3-3. A semisynthetic natural product
derivative, DP-005, binds to an interface pocket of the p65/14-3-3
complex and concomitantly stabilizes it. Cooperativity analyses of
this interaction, and other disease relevant 14-3-3-PPIs, demonstrated
selectivity of DP-005 for the p65/14-3-3 complex. The adaptation of
a cooperative binding model provided a general approach to characterize
stabilization and to assay for selectivity of PPI stabilizers.
Collapse
Affiliation(s)
- Madita Wolter
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - Pim de Vink
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - João Filipe Neves
- U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille F-59000, France.,CNRS ERL9002 Integrative Structural Biology, Lille F-59000, France
| | - Sonja Srdanović
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, United Kingdom
| | - Yusuke Higuchi
- The Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Nobuo Kato
- The Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Andrew Wilson
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, United Kingdom.,Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, United Kingdom
| | - Isabelle Landrieu
- U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille F-59000, France.,CNRS ERL9002 Integrative Structural Biology, Lille F-59000, France
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, P.O. Box 513, Eindhoven 5600 MB, The Netherlands.,Department of Organic Chemistry, University of Duisburg-Essen, 45117, Essen, Germany
| |
Collapse
|
19
|
Folding and structural polymorphism of p53 C-terminal domain: One peptide with many conformations. Arch Biochem Biophys 2020; 684:108342. [DOI: 10.1016/j.abb.2020.108342] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/20/2020] [Accepted: 03/11/2020] [Indexed: 11/19/2022]
|
20
|
ProtCID: a data resource for structural information on protein interactions. Nat Commun 2020; 11:711. [PMID: 32024829 PMCID: PMC7002494 DOI: 10.1038/s41467-020-14301-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022] Open
Abstract
Structural information on the interactions of proteins with other molecules is plentiful, and for some proteins and protein families, there may be 100s of available structures. It can be very difficult for a scientist who is not trained in structural bioinformatics to access this information comprehensively. Previously, we developed the Protein Common Interface Database (ProtCID), which provided clusters of the interfaces of full-length protein chains as a means of identifying biological assemblies. Because proteins consist of domains that act as modular functional units, we have extended the analysis in ProtCID to the individual domain level. This has greatly increased the number of large protein-protein clusters in ProtCID, enabling the generation of hypotheses on the structures of biological assemblies of many systems. The analysis of domain families allows us to extend ProtCID to the interactions of domains with peptides, nucleic acids, and ligands. ProtCID provides complete annotations and coordinate sets for every cluster. The authors previously developed the Protein Common Interface Database (ProtCID), which compares and clusters the interfaces of pairs of full-length protein chains with defined Pfam domain architectures in different PDB entries to identify biological assemblies. Here the authors extend ProtCID to the clustering of domain-domain interactions that also allows analyzing domain interactions with peptides, nucleic acids, and ligands.
Collapse
|
21
|
Kuusk A, Neves JF, Bravo-Rodriguez K, Gunnarsson A, Ruiz-Blanco YB, Ehrmann M, Chen H, Landrieu I, Sanchez-Garcia E, Boyd H, Ottmann C, Doveston RG. Adoption of a Turn Conformation Drives the Binding Affinity of p53 C-Terminal Domain Peptides to 14-3-3σ. ACS Chem Biol 2020; 15:262-271. [PMID: 31742997 DOI: 10.1021/acschembio.9b00893] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The interaction between the adapter protein 14-3-3σ and transcription factor p53 is important for preserving the tumor-suppressor functions of p53 in the cell. A phosphorylated motif within the C-terminal domain (CTD) of p53 is key for binding to the amphipathic groove of 14-3-3. This motif is unique among 14-3-3 binding partners, and the precise dynamics of the interaction is not yet fully understood. Here, we investigate this interaction at the molecular level by analyzing the binding of different length p53 CTD peptides to 14-3-3σ using ITC, SPR, NMR, and MD simulations. We observed that the propensity of the p53 peptide to adopt turn-like conformation plays an important role in the binding to the 14-3-3σ protein. Our study contributes to elucidate the molecular mechanism of the 14-3-3-p53 binding and provides useful insight into how conformation properties of a ligand influence protein binding.
Collapse
Affiliation(s)
- Ave Kuusk
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | | | | | | | | | - Hongming Chen
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
- Chemistry and Chemical Biology Centre, Guangzhou Regenerative Medicine and Health-Guangdong Laboratory, Guangzhou, China
| | | | | | - Helen Boyd
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, U.K
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Richard G. Doveston
- Leicester Institute of Structural and Chemical Biology and School of Chemistry, University of Leicester, University Road, Leicester LE1 7RH, U.K
| |
Collapse
|
22
|
Quantum mechanics/molecular mechanics multiscale modeling of biomolecules. ADVANCES IN PHYSICAL ORGANIC CHEMISTRY 2020. [DOI: 10.1016/bs.apoc.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
23
|
Valenti D, Neves JF, Cantrelle FX, Hristeva S, Lentini Santo D, Obšil T, Hanoulle X, Levy LM, Tzalis D, Landrieu I, Ottmann C. Set-up and screening of a fragment library targeting the 14-3-3 protein interface. MEDCHEMCOMM 2019; 10:1796-1802. [PMID: 31814953 PMCID: PMC6839876 DOI: 10.1039/c9md00215d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/20/2019] [Indexed: 12/16/2022]
Abstract
Protein-protein interactions (PPIs) are at the core of regulation mechanisms in biological systems and consequently became an attractive target for therapeutic intervention. PPIs involving the adapter protein 14-3-3 are representative examples given the broad range of partner proteins forming a complex with one of its seven human isoforms. Given the challenges represented by the nature of these interactions, fragment-based approaches offer a valid alternative for the development of PPI modulators. After having assembled a fragment set tailored on PPIs' modulation, we started a screening campaign on the sigma isoform of 14-3-3 adapter proteins. Through the use of both mono- and bi-dimensional nuclear magnetic resonance spectroscopy measurements, coupled with differential scanning fluorimetry, three fragment hits were identified. These molecules bind the protein at two different regions distant from the usual binding groove highlighting new possibilities for selective modulation of 14-3-3 complexes.
Collapse
Affiliation(s)
- Dario Valenti
- Medicinal Chemistry , Taros Chemicals GmbH & Co. KG , Emil-Figge-Straße 76a , 44227 , Dortmund , Germany .
- Department of Biomedical Engineering and Institute for Complex Molecular Systems , Technische Universiteit Eindhoven , Den Dolech 2 , 5612 AZ Eindhoven , The Netherlands .
| | | | | | - Stanimira Hristeva
- Medicinal Chemistry , Taros Chemicals GmbH & Co. KG , Emil-Figge-Straße 76a , 44227 , Dortmund , Germany .
| | - Domenico Lentini Santo
- Department of Physical and Macromolecular Chemistry , Faculty of Science , Charles University , 12843 Prague , Czech Republic
| | - Tomáš Obšil
- Department of Physical and Macromolecular Chemistry , Faculty of Science , Charles University , 12843 Prague , Czech Republic
- Department of Structural Biology of Signaling Proteins , Division BIOCEV , Institute of Physiology of the Czech Academy of Sciences , Prumyslova 595, 252 50 Vestec , Czech Republic
| | - Xavier Hanoulle
- Univ. Lille , CNRS , UMR 8576 - UGSF , F-59000 Lille , France .
| | - Laura M Levy
- Medicinal Chemistry , Taros Chemicals GmbH & Co. KG , Emil-Figge-Straße 76a , 44227 , Dortmund , Germany .
| | - Dimitrios Tzalis
- Medicinal Chemistry , Taros Chemicals GmbH & Co. KG , Emil-Figge-Straße 76a , 44227 , Dortmund , Germany .
| | | | - Christian Ottmann
- Department of Biomedical Engineering and Institute for Complex Molecular Systems , Technische Universiteit Eindhoven , Den Dolech 2 , 5612 AZ Eindhoven , The Netherlands .
- Department of Chemistry , University of Duisburg-Essen , Universitätsstraße 7 , 45117 , Essen , Germany
| |
Collapse
|
24
|
Shi M, Xu D. Molecular Dynamics Investigations Suggest a Non-specific Recognition Strategy of 14-3-3σ Protein by Tweezer: Implication for the Inhibition Mechanism. Front Chem 2019; 7:237. [PMID: 31058132 PMCID: PMC6478809 DOI: 10.3389/fchem.2019.00237] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 03/26/2019] [Indexed: 02/04/2023] Open
Abstract
The supramolecular complex formed between protein and designed molecule has become one of the most efficient ways to modify protein functions. As one of the more well-studied model systems, 14-3-3 family proteins play an important role in regulating intracellular signaling pathways via protein-protein interactions. In this work, we selected 14-3-3σ as the target protein. Molecular dynamics simulations and binding free energy calculations were applied to identify the possible binding sites and understand its recognition ability of the supramolecular inhibitor, the tweezer molecule (CLR01). On the basis of our simulation, major interactions between lysine residues and CLR01 come from the van der Waals interactions between the long alkyl chain of lysine and the cavity formed by the norbornadiene and benzene rings of the inhibitor. Apart from K214, which was found to be crystallized with this inhibitor, other lysine sites have also shown their abilities to form inclusion complexes with the inhibitor. Such non-specific recognition features of CLR01 against 14-3-3σ can be used in the modification of protein functions via supramolecular chemistry.
Collapse
Affiliation(s)
- Mingsong Shi
- College of Chemistry, Sichuan University, Chengdu, China
| | - Dingguo Xu
- College of Chemistry, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Clinical candidates modulating protein-protein interactions: The fragment-based experience. Eur J Med Chem 2019; 167:76-95. [DOI: 10.1016/j.ejmech.2019.01.084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/23/2022]
|
26
|
Sijbesma E, Hallenbeck KK, Leysen S, de Vink PJ, Skóra L, Jahnke W, Brunsveld L, Arkin MR, Ottmann C. Site-Directed Fragment-Based Screening for the Discovery of Protein–Protein Interaction Stabilizers. J Am Chem Soc 2019; 141:3524-3531. [DOI: 10.1021/jacs.8b11658] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Kenneth K. Hallenbeck
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California, San Francisco 94143, United States
| | - Seppe Leysen
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Pim J. de Vink
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lukasz Skóra
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Wolfgang Jahnke
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Centre (SMDC), University of California, San Francisco 94143, United States
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, 47057 Essen, Germany
| |
Collapse
|
27
|
Woods AG, Wormwood KL, Iosifescu DV, Murrough J, Darie CC. Protein Biomarkers in Major Depressive Disorder: An Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:585-600. [DOI: 10.1007/978-3-030-15950-4_35] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
28
|
Li N, Xie C, Lu NH. p53, a potential predictor of Helicobacter pylori infection-associated gastric carcinogenesis? Oncotarget 2018; 7:66276-66286. [PMID: 27556187 PMCID: PMC5323233 DOI: 10.18632/oncotarget.11414] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori (H. pylori) is an ancient and persistent inhabitant of the human stomach that is closely linked to the development of gastric cancer (GC). . Emerging evidence suggests that H. pylori strain interactions with gastric epithelial cells subvert the best- characterized p53 tumour suppressor pathway. A high prevalence of p53 mutations is related to H. pylori infection. H. pylori also accelerates p53 protein degradation by disturbing the MDM2-P53 feedback loop. Additionally, H. pylori triggers the alteration of other p53 isoforms. Dysregulation of p53 by H. pylori infection contributes to gastric carcinogenesis by mediating cell proliferation and apoptosis. This review focuses on the regulation of p53 in H. pylori infection-associated GC.
Collapse
Affiliation(s)
- Nianshuang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Nong-Hua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
29
|
Shi X, Reimers JR. Understanding non-linear effects from Hill-type dynamics with application to decoding of p53 signaling. Sci Rep 2018; 8:2147. [PMID: 29391550 PMCID: PMC5795017 DOI: 10.1038/s41598-018-20466-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
Analytical equations are derived depicting four possible scenarios resulting from pulsed signaling of a system subject to Hill-type dynamics. Pulsed Hill-type dynamics involves the binding of multiple signal molecules to a receptor and occurs e.g., when transcription factor p53 orchestrates cancer prevention, during calcium signaling, and during circadian rhythms. The scenarios involve: (i) enhancement of high-affinity binders compared to low-affinity ones, (ii) slowing reactions involving high-affinity binders, (iii) transfer of the clocking of low-affinity binders from the signal molecule to the products, and (iv) a unique clocking process that produces incremental increases in the activity of high-affinity binders with each signal pulse. In principle, these mostly non-linear effects could control cellular outcomes. An applications to p53 signaling is developed, with binding to most gene promoters identified as category (iii) responses. However, currently unexplained enhancement of high-affinity promoters such as CDKN1a (p21) by pulsed signaling could be an example of (i). In general, provision for all possible scenarios is required in the design of mathematical models incorporating pulsed Hill-type signaling as some aspect.
Collapse
Affiliation(s)
- Xiaomin Shi
- International Centre for Quantum and Molecular Structures and Mathematics Department, Shanghai University, Shanghai, 200444, China.
| | - Jeffrey R Reimers
- International Centre for Quantum and Molecular Structures and Physics Department, Shanghai University, Shanghai, 200444, China.
- School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
30
|
Sluchanko NN. Association of Multiple Phosphorylated Proteins with the 14-3-3 Regulatory Hubs: Problems and Perspectives. J Mol Biol 2017; 430:20-26. [PMID: 29180038 DOI: 10.1016/j.jmb.2017.11.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 01/11/2023]
Abstract
14-3-3 proteins are well-known universal regulators binding a vast number of partners by recognizing their phosphorylated motifs, typically located within the intrinsically disordered regions. The abundance of such phosphomotifs ensures the involvement of 14-3-3 proteins in sophisticated protein-protein interaction networks that govern vital cellular processes. Thousands of 14-3-3 partners have been either experimentally identified or predicted, but the spatiotemporal hierarchy of the processes based on 14-3-3 interactions is not clearly understood. This is exacerbated by the lack of available structural information on full regulatory complexes involving 14-3-3, which resist high-resolution structural studies due to the presence of intrinsically disordered regions. Although deducing three-dimensional structures is of particular urgency, structural advances are lagging behind the rate at which novel 14-3-3 partners are discovered. Here I attempted to critically review the current state of the field and in particular to dissect the unknowns, focusing on questions that could help in moving the frontiers forward.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology" of the Russian Academy of Sciences, 119071 Moscow, Russian Federation; Department of Biophysics, School of Biology, Moscow State University, 119991 Moscow, Russian Federation.
| |
Collapse
|
31
|
Bier D, Mittal S, Bravo-Rodriguez K, Sowislok A, Guillory X, Briels J, Heid C, Bartel M, Wettig B, Brunsveld L, Sanchez-Garcia E, Schrader T, Ottmann C. The Molecular Tweezer CLR01 Stabilizes a Disordered Protein-Protein Interface. J Am Chem Soc 2017; 139:16256-16263. [PMID: 29039919 PMCID: PMC5691318 DOI: 10.1021/jacs.7b07939] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Indexed: 12/13/2022]
Abstract
Protein regions that are involved in protein-protein interactions (PPIs) very often display a high degree of intrinsic disorder, which is reduced during the recognition process. A prime example is binding of the rigid 14-3-3 adapter proteins to their numerous partner proteins, whose recognition motifs undergo an extensive disorder-to-order transition. In this context, it is highly desirable to control this entropy-costly process using tailored stabilizing agents. This study reveals how the molecular tweezer CLR01 tunes the 14-3-3/Cdc25CpS216 protein-protein interaction. Protein crystallography, biophysical affinity determination and biomolecular simulations unanimously deliver a remarkable finding: a supramolecular "Janus" ligand can bind simultaneously to a flexible peptidic PPI recognition motif and to a well-structured adapter protein. This binding fills a gap in the protein-protein interface, "freezes" one of the conformational states of the intrinsically disordered Cdc25C protein partner and enhances the apparent affinity of the interaction. This is the first structural and functional proof of a supramolecular ligand targeting a PPI interface and stabilizing the binding of an intrinsically disordered recognition motif to a rigid partner protein.
Collapse
Affiliation(s)
- David Bier
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Sumit Mittal
- Max-Planck-Institut
für Kohlenforschung, Kaiser-Wilhelm-Platz 1, 45470 Mülheim an der Ruhr, Germany
| | - Kenny Bravo-Rodriguez
- Max-Planck-Institut
für Kohlenforschung, Kaiser-Wilhelm-Platz 1, 45470 Mülheim an der Ruhr, Germany
| | - Andrea Sowislok
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Xavier Guillory
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Jeroen Briels
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Christian Heid
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Maria Bartel
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Burkhard Wettig
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Luc Brunsveld
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Elsa Sanchez-Garcia
- Max-Planck-Institut
für Kohlenforschung, Kaiser-Wilhelm-Platz 1, 45470 Mülheim an der Ruhr, Germany
| | - Thomas Schrader
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| | - Christian Ottmann
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
- Department
of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117 Essen, Germany
| |
Collapse
|
32
|
Stevers LM, Sijbesma E, Botta M, MacKintosh C, Obsil T, Landrieu I, Cau Y, Wilson AJ, Karawajczyk A, Eickhoff J, Davis J, Hann M, O'Mahony G, Doveston RG, Brunsveld L, Ottmann C. Modulators of 14-3-3 Protein-Protein Interactions. J Med Chem 2017; 61:3755-3778. [PMID: 28968506 PMCID: PMC5949722 DOI: 10.1021/acs.jmedchem.7b00574] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Direct
interactions between proteins are essential for the regulation
of their functions in biological pathways. Targeting the complex network
of protein–protein interactions (PPIs) has now been widely
recognized as an attractive means to therapeutically intervene in
disease states. Even though this is a challenging endeavor and PPIs
have long been regarded as “undruggable” targets, the
last two decades have seen an increasing number of successful examples
of PPI modulators, resulting in growing interest in this field. PPI
modulation requires novel approaches and the integrated efforts of
multiple disciplines to be a fruitful strategy. This perspective focuses
on the hub-protein 14-3-3, which has several hundred identified protein
interaction partners, and is therefore involved in a wide range of
cellular processes and diseases. Here, we aim to provide an integrated
overview of the approaches explored for the modulation of 14-3-3 PPIs
and review the examples resulting from these efforts in both inhibiting
and stabilizing specific 14-3-3 protein complexes by small molecules,
peptide mimetics, and natural products.
Collapse
Affiliation(s)
- Loes M Stevers
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy , University of Siena , Via Aldo Moro 2 , 53100 Siena , Italy
| | - Carol MacKintosh
- Division of Cell and Developmental Biology, School of Life Sciences , University of Dundee , Dundee DD1 4HN , United Kingdom
| | - Tomas Obsil
- Department of Physical and Macromolecular Chemistry, Faculty of Science , Charles University , Prague 116 36 , Czech Republic
| | | | - Ylenia Cau
- Department of Biotechnology, Chemistry and Pharmacy , University of Siena , Via Aldo Moro 2 , 53100 Siena , Italy
| | - Andrew J Wilson
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , United Kingdom.,Astbury Center For Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , United Kingdom
| | | | - Jan Eickhoff
- Lead Discovery Center GmbH , Dortmund 44227 , Germany
| | - Jeremy Davis
- UCB Celltech , 216 Bath Road , Slough SL1 3WE , United Kingdom
| | - Michael Hann
- GlaxoSmithKline , Gunnels Wood Road , Stevenage, Hertfordshire SG1 2NY , United Kingdom
| | - Gavin O'Mahony
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit , AstraZeneca Gothenburg , Pepparedsleden 1 , SE-431 83 Mölndal , Sweden
| | - Richard G Doveston
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands.,Department of Chemistry , University of Duisburg-Essen , Universitätstraße 7 , 45141 Essen , Germany
| |
Collapse
|
33
|
Sluchanko NN, Tugaeva KV, Greive SJ, Antson AA. Chimeric 14-3-3 proteins for unraveling interactions with intrinsically disordered partners. Sci Rep 2017; 7:12014. [PMID: 28931924 PMCID: PMC5607241 DOI: 10.1038/s41598-017-12214-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023] Open
Abstract
In eukaryotes, several “hub” proteins integrate signals from different interacting partners that bind through intrinsically disordered regions. The 14-3-3 protein hub, which plays wide-ranging roles in cellular processes, has been linked to numerous human disorders and is a promising target for therapeutic intervention. Partner proteins usually bind via insertion of a phosphopeptide into an amphipathic groove of 14-3-3. Structural plasticity in the groove generates promiscuity allowing accommodation of hundreds of different partners. So far, accurate structural information has been derived for only a few 14-3-3 complexes with phosphopeptide-containing proteins and a variety of complexes with short synthetic peptides. To further advance structural studies, here we propose a novel approach based on fusing 14-3-3 proteins with the target partner peptide sequences. Such chimeric proteins are easy to design, express, purify and crystallize. Peptide attachment to the C terminus of 14-3-3 via an optimal linker allows its phosphorylation by protein kinase A during bacterial co-expression and subsequent binding at the amphipathic groove. Crystal structures of 14-3-3 chimeras with three different peptides provide detailed structural information on peptide-14-3-3 interactions. This simple but powerful approach, employing chimeric proteins, can reinvigorate studies of 14-3-3/phosphoprotein assemblies, including those with challenging low-affinity partners, and may facilitate the design of novel biosensors.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology" of the Russian Academy of Sciences, 119071, Moscow, Russian Federation. .,Department of biophysics, School of Biology, Moscow State University, 119991, Moscow, Russian Federation.
| | - Kristina V Tugaeva
- A.N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology" of the Russian Academy of Sciences, 119071, Moscow, Russian Federation.,Department of biochemistry, School of Biology, Moscow State University, 119991, Moscow, Russian Federation
| | - Sandra J Greive
- York Structural Biology Laboratory, Department of Chemistry, University of York, York, YO10 5DD, United Kingdom
| | - Alfred A Antson
- York Structural Biology Laboratory, Department of Chemistry, University of York, York, YO10 5DD, United Kingdom
| |
Collapse
|
34
|
Doveston RG, Kuusk A, Andrei SA, Leysen S, Cao Q, Castaldi MP, Hendricks A, Brunsveld L, Chen H, Boyd H, Ottmann C. Small-molecule stabilization of the p53 - 14-3-3 protein-protein interaction. FEBS Lett 2017. [DOI: 10.1002/1873-3468.12723] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Richard G. Doveston
- Laboratory of Chemical Biology; Department of Biomedical Engineering and Institute for Complex Molecular Systems; Eindhoven University of Technology; The Netherlands
| | - Ave Kuusk
- Laboratory of Chemical Biology; Department of Biomedical Engineering and Institute for Complex Molecular Systems; Eindhoven University of Technology; The Netherlands
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca R&D Gothenburg; Mölndal Sweden
| | - Sebastian A. Andrei
- Laboratory of Chemical Biology; Department of Biomedical Engineering and Institute for Complex Molecular Systems; Eindhoven University of Technology; The Netherlands
| | - Seppe Leysen
- Laboratory of Chemical Biology; Department of Biomedical Engineering and Institute for Complex Molecular Systems; Eindhoven University of Technology; The Netherlands
| | - Qing Cao
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca; Waltham MA USA
| | - Maria P. Castaldi
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca; Waltham MA USA
| | - Adam Hendricks
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca; Waltham MA USA
| | - Luc Brunsveld
- Laboratory of Chemical Biology; Department of Biomedical Engineering and Institute for Complex Molecular Systems; Eindhoven University of Technology; The Netherlands
| | - Hongming Chen
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca R&D Gothenburg; Mölndal Sweden
| | - Helen Boyd
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca R&D Gothenburg; Mölndal Sweden
| | - Christian Ottmann
- Laboratory of Chemical Biology; Department of Biomedical Engineering and Institute for Complex Molecular Systems; Eindhoven University of Technology; The Netherlands
- Department of Chemistry; University of Duisburg-Essen; Germany
| |
Collapse
|
35
|
Structural interface between LRRK2 and 14-3-3 protein. Biochem J 2017; 474:1273-1287. [PMID: 28202711 DOI: 10.1042/bcj20161078] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/09/2017] [Accepted: 02/14/2017] [Indexed: 02/03/2023]
Abstract
Binding of 14-3-3 proteins to leucine-rich repeat protein kinase 2 (LRRK2) is known to be impaired by many Parkinson's disease (PD)-relevant mutations. Abrogation of this interaction is connected to enhanced LRRK2 kinase activity, which in turn is implicated in increased ubiquitination of LRRK2, accumulation of LRRK2 into inclusion bodies and reduction in neurite length. Hence, the interaction between 14-3-3 and LRRK2 is of significant interest as a possible drug target for the treatment of PD. However, LRRK2 possesses multiple sites that, upon phosphorylation, can bind to 14-3-3, thus rendering the interaction relatively complex. Using biochemical assays and crystal structures, we characterize the multivalent interaction between these two proteins.
Collapse
|
36
|
Sluchanko NN, Beelen S, Kulikova AA, Weeks SD, Antson AA, Gusev NB, Strelkov SV. Structural Basis for the Interaction of a Human Small Heat Shock Protein with the 14-3-3 Universal Signaling Regulator. Structure 2017; 25:305-316. [PMID: 28089448 PMCID: PMC5321513 DOI: 10.1016/j.str.2016.12.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/14/2016] [Accepted: 12/12/2016] [Indexed: 12/31/2022]
Abstract
By interacting with hundreds of protein partners, 14-3-3 proteins coordinate vital cellular processes. Phosphorylation of the small heat shock protein, HSPB6, within its intrinsically disordered N-terminal domain activates its interaction with 14-3-3, ultimately triggering smooth muscle relaxation. After analyzing the binding of an HSPB6-derived phosphopeptide to 14-3-3 using isothermal calorimetry and X-ray crystallography, we have determined the crystal structure of the complete assembly consisting of the 14-3-3 dimer and full-length HSPB6 dimer and further characterized this complex in solution using fluorescence spectroscopy, small-angle X-ray scattering, and limited proteolysis. We show that selected intrinsically disordered regions of HSPB6 are transformed into well-defined conformations upon the interaction, whereby an unexpectedly asymmetric structure is formed. This structure provides the first atomic resolution snapshot of a human small HSP in functional state, explains how 14-3-3 proteins sequester their regulatory partners, and can inform the design of small-molecule interaction modifiers to be used as myorelaxants.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- Laboratory of Structural Biochemistry of Proteins, A.N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, 119071 Moscow, Russia.
| | - Steven Beelen
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Alexandra A Kulikova
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia
| | - Stephen D Weeks
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Alfred A Antson
- York Structural Biology Laboratory, Department of Chemistry, University of York, York YO10 5YW, UK
| | - Nikolai B Gusev
- Department of Biochemistry, School of Biology, Moscow State University, 119991 Moscow, Russia
| | - Sergei V Strelkov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
37
|
p53 Proteoforms and Intrinsic Disorder: An Illustration of the Protein Structure-Function Continuum Concept. Int J Mol Sci 2016; 17:ijms17111874. [PMID: 27834926 PMCID: PMC5133874 DOI: 10.3390/ijms17111874] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/27/2016] [Accepted: 11/03/2016] [Indexed: 01/10/2023] Open
Abstract
Although it is one of the most studied proteins, p53 continues to be an enigma. This protein has numerous biological functions, possesses intrinsically disordered regions crucial for its functionality, can form both homo-tetramers and isoform-based hetero-tetramers, and is able to interact with many binding partners. It contains numerous posttranslational modifications, has several isoforms generated by alternative splicing, alternative promoter usage or alternative initiation of translation, and is commonly mutated in different cancers. Therefore, p53 serves as an important illustration of the protein structure–function continuum concept, where the generation of multiple proteoforms by various mechanisms defines the ability of this protein to have a multitude of structurally and functionally different states. Considering p53 in the light of a proteoform-based structure–function continuum represents a non-canonical and conceptually new contemplation of structure, regulation, and functionality of this important protein.
Collapse
|
38
|
Characterization and small-molecule stabilization of the multisite tandem binding between 14-3-3 and the R domain of CFTR. Proc Natl Acad Sci U S A 2016; 113:E1152-61. [PMID: 26888287 DOI: 10.1073/pnas.1516631113] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cystic fibrosis is a fatal genetic disease, most frequently caused by the retention of the CFTR (cystic fibrosis transmembrane conductance regulator) mutant protein in the endoplasmic reticulum (ER). The binding of the 14-3-3 protein to the CFTR regulatory (R) domain has been found to enhance CFTR trafficking to the plasma membrane. To define the mechanism of action of this protein-protein interaction, we have examined the interaction in vitro. The disordered multiphosphorylated R domain contains nine different 14-3-3 binding motifs. Furthermore, the 14-3-3 protein forms a dimer containing two amphipathic grooves that can potentially bind these phosphorylated motifs. This results in a number of possible binding mechanisms between these two proteins. Using multiple biochemical assays and crystal structures, we show that the interaction between them is governed by two binding sites: The key binding site of CFTR (pS768) occupies one groove of the 14-3-3 dimer, and a weaker, secondary binding site occupies the other binding groove. We show that fusicoccin-A, a natural-product tool compound used in studies of 14-3-3 biology, can stabilize the interaction between 14-3-3 and CFTR by selectively interacting with a secondary binding motif of CFTR (pS753). The stabilization of this interaction stimulates the trafficking of mutant CFTR to the plasma membrane. This definition of the druggability of the 14-3-3-CFTR interface might offer an approach for cystic fibrosis therapeutics.
Collapse
|
39
|
Bier D, Bartel M, Sies K, Halbach S, Higuchi Y, Haranosono Y, Brummer T, Kato N, Ottmann C. Small-Molecule Stabilization of the 14-3-3/Gab2 Protein-Protein Interaction (PPI) Interface. ChemMedChem 2015; 11:911-8. [DOI: 10.1002/cmdc.201500484] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Indexed: 02/06/2023]
Affiliation(s)
- David Bier
- Department of Chemistry; University of Duisburg-Essen; Universitätstr. 7 45141 Essen Germany
| | - Maria Bartel
- Department of Biomedical Engineering; Institute for Complex Molecular, Systems; Eindhoven University of Technology; P.O. Box 513 5600 MB Eindhoven The Netherlands
| | - Katharina Sies
- Institute of Molecular Medicine & Cell Research (IMMZ); Faculty of Medicine; University of Freiburg; Stefan-Meier-Str. 17 79104 Freiburg Germany
| | - Sebastian Halbach
- Institute of Molecular Medicine & Cell Research (IMMZ); Faculty of Medicine; University of Freiburg; Stefan-Meier-Str. 17 79104 Freiburg Germany
- Faculty of Biology; University of Freiburg; Schänzlestr. 1 79104 Freiburg Germany
- Spemann Graduate School of Biology & Medicine; University of Freiburg; Albertstr. 19A 79104 Freiburg Germany
| | - Yusuke Higuchi
- The Institute of Scientific & Industrial Research; Osaka University; Osaka 567-0047 Japan
| | - Yu Haranosono
- The Institute of Scientific & Industrial Research; Osaka University; Osaka 567-0047 Japan
| | - Tilman Brummer
- Institute of Molecular Medicine & Cell Research (IMMZ); Faculty of Medicine; University of Freiburg; Stefan-Meier-Str. 17 79104 Freiburg Germany
- BIOSS: Centre for Biological Signaling Studies; University of Freiburg; Schänzlestr. 18 79104 Freiburg Germany
| | - Nobuo Kato
- The Institute of Scientific & Industrial Research; Osaka University; Osaka 567-0047 Japan
| | - Christian Ottmann
- Department of Chemistry; University of Duisburg-Essen; Universitätstr. 7 45141 Essen Germany
- Department of Biomedical Engineering; Institute for Complex Molecular, Systems; Eindhoven University of Technology; P.O. Box 513 5600 MB Eindhoven The Netherlands
| |
Collapse
|
40
|
Abstract
Modulation of protein-protein interactions (PPIs) is becoming increasingly important in drug discovery and chemical biology. While a few years ago this 'target class' was deemed to be largely undruggable an impressing number of publications and success stories now show that targeting PPIs with small, drug-like molecules indeed is a feasible approach. Here, we summarize the current state of small-molecule inhibition and stabilization of PPIs and review the active molecules from a structural and medicinal chemistry angle, especially focusing on the key examples of iNOS, LFA-1 and 14-3-3.
Collapse
|
41
|
Babula JJ, Liu JY. Integrate Omics Data and Molecular Dynamics Simulations toward Better Understanding of Human 14-3-3 Interactomes and Better Drugs for Cancer Therapy. J Genet Genomics 2015; 42:531-547. [PMID: 26554908 DOI: 10.1016/j.jgg.2015.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/03/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022]
Abstract
The 14-3-3 protein family is among the most extensively studied, yet still largely mysterious protein families in mammals to date. As they are well recognized for their roles in apoptosis, cell cycle regulation, and proliferation in healthy cells, aberrant 14-3-3 expression has unsurprisingly emerged as instrumental in the development of many cancers and in prognosis. Interestingly, while the seven known 14-3-3 isoforms in humans have many similar functions across cell types, evidence of isoform-specific functions and localization has been observed in both healthy and diseased cells. The strikingly high similarity among 14-3-3 isoforms has made it difficult to delineate isoform-specific functions and for isoform-specific targeting. Here, we review our knowledge of 14-3-3 interactome(s) generated by high-throughput techniques, bioinformatics, structural genomics and chemical genomics and point out that integrating the information with molecular dynamics (MD) simulations may bring us new opportunity to the design of isoform-specific inhibitors, which can not only be used as powerful research tools for delineating distinct interactomes of individual 14-3-3 isoforms, but also can serve as potential new anti-cancer drugs that selectively target aberrant 14-3-3 isoform.
Collapse
Affiliation(s)
- JoAnne J Babula
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Jing-Yuan Liu
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA; Department of Computer and Information Science, Indiana University Purdue University Indianapolis, 723 W. Michigan St., Indianapolis, IN 46202, USA.
| |
Collapse
|
42
|
Joo Y, Schumacher B, Landrieu I, Bartel M, Smet-Nocca C, Jang A, Choi HS, Jeon NL, Chang KA, Kim HS, Ottmann C, Suh YH. Involvement of 14-3-3 in tubulin instability and impaired axon development is mediated by Tau. FASEB J 2015; 29:4133-44. [PMID: 26103986 DOI: 10.1096/fj.14-265009] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 06/15/2015] [Indexed: 01/06/2023]
Abstract
14-3-3 proteins act as adapters that exert their function by interacting with their various protein partners. 14-3-3 proteins have been implicated in a variety of human diseases including neurodegenerative diseases. 14-3-3 proteins have recently been reported to be abundant in the neurofibrillary tangles (NFTs) observed inside the neurons of brains affected by Alzheimer's disease (AD). These NFTs are mainly constituted of phosphorylated Tau protein, a microtubule-associated protein known to bind 14-3-3. Despite this indication of 14-3-3 protein involvement in the AD pathogenesis, the role of 14-3-3 in the Tauopathy remains to be clarified. In the present study, we shed light on the role of 14-3-3 proteins in the molecular pathways leading to Tauopathies. Overexpression of the 14-3-3σ isoform resulted in a disruption of the tubulin cytoskeleton and prevented neuritic outgrowth in neurons. NMR studies validated the phosphorylated residues pSer214 and pSer324 in Tau as the 2 primary sites for 14-3-3 binding, with the crystal structure of 14-3-3σ in complex with Tau-pSer214 and Tau-pSer324 revealing the molecular details of the interaction. These data suggest a rationale for a possible pharmacologic intervention of the Tau/14-3-3 interaction.
Collapse
Affiliation(s)
- Yuyoung Joo
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Benjamin Schumacher
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Isabelle Landrieu
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Maria Bartel
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Caroline Smet-Nocca
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ahram Jang
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hee Soon Choi
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Noo Li Jeon
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Keun-A Chang
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hye-Sun Kim
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Christian Ottmann
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Yoo-Hun Suh
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
43
|
Small molecules, peptides and natural products: getting a grip on 14-3-3 protein-protein modulation. Future Med Chem 2015; 6:903-21. [PMID: 24962282 DOI: 10.4155/fmc.14.47] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
One of the proteins that is found in a diverse range of eukaryotic protein-protein interactions is the adaptor protein 14-3-3. As 14-3-3 is a hub protein with very diverse interactions, it is a good model to study various protein-protein interactions. A wide range of classes of molecules, peptides, small molecules or natural products, has been used to modify the protein interactions, providing both stabilization or inhibition of the interactions of 14-3-3 with its binding partners. The first protein crystal structures were solved in 1995 and gave molecular insights for further research. The plant analog of 14-3-3 binds to a plant plasma membrane H(+)-ATPase and this protein complex is stabilized by the fungal phytotoxin fusicoccin A. The knowledge gained from the process in plants was transferred to and applied in human models to find stabilizers or inhibitors of 14-3-3 interaction in human cellular pathways.
Collapse
|
44
|
Gaglia G, Lahav G. Constant rate of p53 tetramerization in response to DNA damage controls the p53 response. Mol Syst Biol 2014; 10:753. [PMID: 25344068 PMCID: PMC4299375 DOI: 10.15252/msb.20145168] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The dynamics of the tumor suppressor protein p53 have been previously investigated in single
cells using fluorescently tagged p53. Such approach reports on the total abundance of p53 but does
not provide a measure for functional p53. We used fluorescent protein-fragment complementation assay
(PCA) to quantify in single cells the dynamics of p53 tetramers, the functional units of p53. We
found that while total p53 increases proportionally to the input strength, p53 tetramers are formed
in cells at a constant rate. This breaks the linear input–output relation and dampens the p53
response. Disruption of the p53-binding protein ARC led to a dose-dependent rate of tetramers
formation, resulting in enhanced tetramerization and induction of p53 target genes. Our work
suggests that constraining the p53 response in face of variable inputs may protect cells from
committing to terminal outcomes and highlights the importance of quantifying the active form of
signaling molecules in single cells. Quantification of the dynamics of p53 tetramers in single cells using a fluorescent
protein-fragment complementation assay reveals that, while total p53 increases proportionally to the
DNA damage strength, p53 tetramers are formed at a constant rate.
Collapse
Affiliation(s)
- Giorgio Gaglia
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Galit Lahav
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Mori M, Vignaroli G, Botta M. Small molecules modulation of 14-3-3 protein-protein interactions. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 10:e541-7. [PMID: 24451646 DOI: 10.1016/j.ddtec.2012.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
14-3-3 is a family of highly conserved regulatory proteins which is attracting a significant interest due to its potential role as target for pharmacological intervention against cancer and neurodegenerative disorders. Although modulating protein-protein interactions (PPI) is still conceived as a challenging task in drug discovery, in past few years peptide inhibitors and small molecular modulators of 14-3-3 PPI have been described. Here we examine structural and biological features of 14-3-3 and propose an overview on techniques used for discovering small molecular inhibitors and stabilizers of 14-3-3 PPI.
Collapse
|
46
|
Uversky VN, Davé V, Iakoucheva LM, Malaney P, Metallo SJ, Pathak RR, Joerger AC. Pathological unfoldomics of uncontrolled chaos: intrinsically disordered proteins and human diseases. Chem Rev 2014; 114:6844-79. [PMID: 24830552 PMCID: PMC4100540 DOI: 10.1021/cr400713r] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute University of South Florida, Tampa, Florida 33612, United States
- Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Vrushank Davé
- Department of Pathology and Cell Biology , Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Lilia M. Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093, United States
| | - Prerna Malaney
- Department of Pathology and Cell Biology , Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Steven J. Metallo
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Ravi Ramesh Pathak
- Department of Pathology and Cell Biology , Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Andreas C. Joerger
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
47
|
Nguyen TA, Menendez D, Resnick MA, Anderson CW. Mutant TP53 posttranslational modifications: challenges and opportunities. Hum Mutat 2014; 35:738-55. [PMID: 24395704 DOI: 10.1002/humu.22506] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022]
Abstract
The wild-type (WT) human p53 (TP53) tumor suppressor can be posttranslationally modified at over 60 of its 393 residues. These modifications contribute to changes in TP53 stability and in its activity as a transcription factor in response to a wide variety of intrinsic and extrinsic stresses in part through regulation of protein-protein and protein-DNA interactions. The TP53 gene frequently is mutated in cancers, and in contrast to most other tumor suppressors, the mutations are mostly missense often resulting in the accumulation of mutant (MUT) protein, which may have novel or altered functions. Most MUT TP53s can be posttranslationally modified at the same residues as in WT TP53. Strikingly, however, codons for modified residues are rarely mutated in human tumors, suggesting that TP53 modifications are not essential for tumor suppression activity. Nevertheless, these modifications might alter MUT TP53 activity and contribute to a gain-of-function leading to increased metastasis and tumor progression. Furthermore, many of the signal transduction pathways that result in TP53 modifications are altered or disrupted in cancers. Understanding the signaling pathways that result in TP53 modification and the functions of these modifications in both WT TP53 and its many MUT forms may contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Thuy-Ai Nguyen
- Chromosome Stability Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | |
Collapse
|
48
|
Ono W, Hayashi Y, Yokoyama W, Kuroda T, Kishimoto H, Ito I, Kimura K, Akaogi K, Waku T, Yanagisawa J. The nucleolar protein Myb-binding protein 1A (MYBBP1A) enhances p53 tetramerization and acetylation in response to nucleolar disruption. J Biol Chem 2013; 289:4928-40. [PMID: 24375404 DOI: 10.1074/jbc.m113.474049] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Tetramerization of p53 is crucial to exert its biological activity, and nucleolar disruption is sufficient to activate p53. We previously demonstrated that nucleolar stress induces translocation of the nucleolar protein MYBBP1A from the nucleolus to the nucleoplasm and enhances p53 activity. However, whether and how MYBBP1A regulates p53 tetramerization in response to nucleolar stress remain unclear. In this study, we demonstrated that MYBBP1A enhances p53 tetramerization, followed by acetylation under nucleolar stress. We found that MYBBP1A has two regions that directly bind to lysine residues of the p53 C-terminal regulatory domain. MYBBP1A formed a self-assembled complex that provided a molecular platform for p53 tetramerization and enhanced p300-mediated acetylation of the p53 tetramer. Moreover, our results show that MYBBP1A functions to enhance p53 tetramerization that is necessary for p53 activation, followed by cell death with actinomycin D treatment. Thus, we suggest that MYBBP1A plays a pivotal role in the cellular stress response.
Collapse
Affiliation(s)
- Wakana Ono
- From the Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki 305-8577, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Activation and control of p53 tetramerization in individual living cells. Proc Natl Acad Sci U S A 2013; 110:15497-501. [PMID: 24006363 DOI: 10.1073/pnas.1311126110] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Homo-oligomerization is found in many biological systems and has been extensively studied in vitro. However, our ability to quantify and understand oligomerization processes in cells is still limited. We used fluorescence correlation spectroscopy and mathematical modeling to measure the dynamics of the tetramers formed by the tumor suppressor protein p53 in single living cells. Previous in vitro studies suggested that in basal conditions all p53 molecules are bound in dimers. We found that in resting cells p53 is present in a mix of oligomeric states with a large cell-to-cell variation. After DNA damage, p53 molecules in all cells rapidly assemble into tetramers before p53 protein levels increase. We developed a model to understand the connection between p53 accumulation and tetramerization. We found that the rapid increase in p53 tetramers requires a combination of active tetramerization and protein stabilization, however tetramerization alone is sufficient to activate p53 transcriptional targets. This suggests triggering tetramerization as a mechanism for activating the p53 pathway in cancer cells. Many other transcription factors homo-oligomerize, and our approach provides a unique way for probing the dynamics and functional consequences of oligomerization.
Collapse
|
50
|
Zakharchenko O, Cojoc M, Dubrovska A, Souchelnytskyi S. A role of TGFß1 dependent 14-3-3σ phosphorylation at Ser69 and Ser74 in the regulation of gene transcription, stemness and radioresistance. PLoS One 2013; 8:e65163. [PMID: 23741479 PMCID: PMC3669286 DOI: 10.1371/journal.pone.0065163] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/23/2013] [Indexed: 01/16/2023] Open
Abstract
Transforming growth factor-β (TGFβ) is a potent regulator of tumorigenesis, although mechanisms defining its tumor suppressing and tumor promoting activities are not understood. Here we describe phosphoproteome profiling of TGFβ signaling in mammary epithelial cells, and show that 60 identified TGFβ-regulated phosphoproteins form a network with scale-free characteristics. The network highlighted interactions, which may distribute signaling inputs to regulation of cell proliferation, metabolism, differentiation and cell organization. In this report, we identified two novel and TGFβ-dependent phosphorylation sites of 14-3-3σ, i.e. Ser69 and Ser74. We observed that 14-3-3σ phosphorylation is a feed-forward mechanism in TGFβ/Smad3-dependent transcription. TGFβ-dependent 14-3-3σ phosphorylation may provide a scaffold for the formation of the protein complexes which include Smad3 and p53 at the Smad3-specific CAGA element. Furthermore, breast tumor xenograft studies in mice and radiobiological assays showed that phosphorylation of 14-3-3σ at Ser69 and Ser74 is involved in regulation of cancer progenitor population and radioresistance in breast cancer MCF7 cells. Our data suggest that TGFβ-dependent phosphorylation of 14-3-3σ orchestrates a functional interaction of TGFβ/Smad3 with p53, plays a role in the maintenance of cancer stem cells and could provide a new potential target for intervention in breast cancer.
Collapse
Affiliation(s)
- Olena Zakharchenko
- Karolinska Biomics Center, Dept. of Oncology-Pathology, Karolinska Institute, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | | | | | |
Collapse
|