51
|
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: 55] [Impact Index Per Article: 5.5] [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
|
52
|
Woodcock JM, Coolen C, Goodwin KL, Baek DJ, Bittman R, Samuel MS, Pitson SM, Lopez AF. Destabilisation of dimeric 14-3-3 proteins as a novel approach to anti-cancer therapeutics. Oncotarget 2015; 6:14522-36. [PMID: 25971334 PMCID: PMC4546484 DOI: 10.18632/oncotarget.3995] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/11/2015] [Indexed: 12/24/2022] Open
Abstract
14-3-3 proteins play a pivotal role in controlling cell proliferation and survival, two commonly dysregulated hallmarks of cancers. 14-3-3 protein expression is enhanced in many human cancers and correlates with more aggressive tumors and poor prognosis, suggesting a role for 14-3-3 proteins in tumorigenesis and/or progression. We showed previously that the dimeric state of 14-3-3 proteins is regulated by the lipid sphingosine, a physiological inducer of apoptosis. As the functions of 14-3-3 proteins are dependent on their dimeric state, this sphingosine-mediated 14-3-3 regulation provides a possible means to target dimeric 14-3-3 for therapeutic effect. However, sphingosine mimics are needed that are not susceptible to sphingolipid metabolism. We show here the identification and optimization of sphingosine mimetics that render dimeric 14-3-3 susceptible to phosphorylation at a site buried in the dimer interface and induce mitochondrial-mediated apoptosis. Two such compounds, RB-011 and RB-012, disrupt 14-3-3 dimers at low micromolar concentrations and induce rapid down-regulation of Raf-MAPK and PI3K-Akt signaling in Jurkat cells. Importantly, both RB-011 and RB-012 induce apoptosis of human A549 lung cancer cells and RB-012, through disruption of MAPK signaling, reduces xenograft growth in mice. Thus, these compounds provide proof-of-principle for this novel 14-3-3-targeting approach for anti-cancer drug discovery.
Collapse
Affiliation(s)
- Joanna M. Woodcock
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Carl Coolen
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Katy L. Goodwin
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Dong Jae Baek
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY, USA
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY, USA
| | - Michael S. Samuel
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stuart M. Pitson
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Angel F. Lopez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
53
|
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
|
54
|
An S, Yang Y, Ward R, Liu Y, Guo XX, Xu TR. Raf-interactome in tuning the complexity and diversity of Raf function. FEBS J 2014; 282:32-53. [PMID: 25333451 DOI: 10.1111/febs.13113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/06/2014] [Accepted: 10/14/2014] [Indexed: 12/23/2022]
Abstract
Raf kinases have been intensely studied subsequent to their discovery 30 years ago. The Ras-Raf-mitogen-activated protein kinase/extracellular signal-regulated kinase kinase-extracellular signal-regulated kinase/mitogen-activated protein kinase (Ras-Raf-MEK-ERK/MAPK) signaling pathway is at the heart of the signaling networks that control many fundamental cellular processes and Raf kinases takes centre stage in the MAPK pathway, which is now appreciated to be one of the most common sources of the oncogenic mutations in cancer. The dependency of tumors on this pathway has been clearly demonstrated by targeting its key nodes; however, blockade of the central components of the MAPK pathway may have some unexpected side effects. Over recent years, the Raf-interactome or Raf-interacting proteins have emerged as promising targets for protein-directed cancer therapy. This review focuses on the diversity of Raf-interacting proteins and discusses the mechanisms by which these proteins regulate Raf function, as well as the implications of targeting Raf-interacting proteins in the treatment of human cancer.
Collapse
Affiliation(s)
- Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan, China
| | | | | | | | | | | |
Collapse
|
55
|
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
|
56
|
The application of an emerging technique for protein-protein interaction interface mapping: the combination of photo-initiated cross-linking protein nanoprobes with mass spectrometry. Int J Mol Sci 2014; 15:9224-41. [PMID: 24865487 PMCID: PMC4100091 DOI: 10.3390/ijms15069224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 05/06/2014] [Accepted: 05/09/2014] [Indexed: 11/30/2022] Open
Abstract
Protein–protein interaction was investigated using a protein nanoprobe capable of photo-initiated cross-linking in combination with high-resolution and tandem mass spectrometry. This emerging experimental approach introduces photo-analogs of amino acids within a protein sequence during its recombinant expression, preserves native protein structure and is suitable for mapping the contact between two proteins. The contact surface regions involved in the well-characterized interaction between two molecules of human 14-3-3ζ regulatory protein were used as a model. The employed photo-initiated cross-linking techniques extend the number of residues shown to be within interaction distance in the contact surface of the 14-3-3ζ dimer (Gln8–Met78). The results of this study are in agreement with our previously published data from molecular dynamic calculations based on high-resolution chemical cross-linking data and Hydrogen/Deuterium exchange mass spectrometry. The observed contact is also in accord with the 14-3-3ζ X-ray crystal structure (PDB 3dhr). The results of the present work are relevant to the structural biology of transient interaction in the 14-3-3ζ protein, and demonstrate the ability of the chosen methodology (the combination of photo-initiated cross-linking protein nanoprobes and mass spectrometry analysis) to map the protein-protein interface or regions with a flexible structure.
Collapse
|
57
|
Milroy LG, Grossmann TN, Hennig S, Brunsveld L, Ottmann C. Modulators of Protein–Protein Interactions. Chem Rev 2014; 114:4695-748. [DOI: 10.1021/cr400698c] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Lech-Gustav Milroy
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech
2, 5612 AZ Eindhoven, The Netherlands
| | - Tom N. Grossmann
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn Straße 15, 44227 Dortmund, Germany
- Department
of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Sven Hennig
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn Straße 15, 44227 Dortmund, Germany
| | - Luc Brunsveld
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech
2, 5612 AZ Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech
2, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
58
|
Nero TL, Morton CJ, Holien JK, Wielens J, Parker MW. Oncogenic protein interfaces: small molecules, big challenges. Nat Rev Cancer 2014; 14:248-62. [PMID: 24622521 DOI: 10.1038/nrc3690] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Historically, targeting protein-protein interactions with small molecules was not thought possible because the corresponding interfaces were considered mostly flat and featureless and therefore 'undruggable'. Instead, such interactions were targeted with larger molecules, such as peptides and antibodies. However, the past decade has seen encouraging breakthroughs through the refinement of existing techniques and the development of new ones, together with the identification and exploitation of unexpected aspects of protein-protein interaction surfaces. In this Review, we describe some of the latest techniques to discover modulators of protein-protein interactions and how current drug discovery approaches have been adapted to successfully target these interfaces.
Collapse
Affiliation(s)
- Tracy L Nero
- Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Craig J Morton
- Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Jessica K Holien
- Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Jerome Wielens
- 1] Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia. [2] Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy, Victoria 3065, Australia
| | - Michael W Parker
- 1] Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia. [2] Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
59
|
Aeluri M, Chamakuri S, Dasari B, Guduru SKR, Jimmidi R, Jogula S, Arya P. Small Molecule Modulators of Protein–Protein Interactions: Selected Case Studies. Chem Rev 2014; 114:4640-94. [DOI: 10.1021/cr4004049] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Madhu Aeluri
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Srinivas Chamakuri
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Bhanudas Dasari
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Shiva Krishna Reddy Guduru
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Ravikumar Jimmidi
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Srinivas Jogula
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Prabhat Arya
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| |
Collapse
|
60
|
Abstract
Rho GTPases are a family of small GTPases, which play an important role in the regulation of the actin cytoskeleton. Not surprisingly, Rho GTPases are crucial for cell migration and therefore highly important for cancer cell invasion and the formation of metastases. In addition, Rho GTPases are involved in growth and survival of tumor cells, in the interaction of tumor cells with their environment, and they are vital for the cancer supporting functions of the tumor stroma. Recent research has significantly improved our understanding of the regulation of Rho GTPase activity, the specificity of Rho GTPases, and their function in tumor stem cells and tumor stroma. This review summarizes these novel findings and tries to define challenging questions for future research.
Collapse
Affiliation(s)
- Hui Li
- University of Copenhagen, BRIC, BMI, 2200, Copenhagen, Denmark
| | | | | | | |
Collapse
|
61
|
Cheng C, Zhao H, Wang Z, Lu W, Wang L, Wang R, Yao L. The effect of 5'-adenylic acid on hepatic proteome of mice radiated by 60Co γ-ray. Int J Mol Sci 2013; 15:186-202. [PMID: 24368518 PMCID: PMC3907805 DOI: 10.3390/ijms15010186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/15/2013] [Accepted: 12/18/2013] [Indexed: 01/30/2023] Open
Abstract
Understanding the protection mechanism of 5′-AMP requires comprehensive knowledge of the proteins expressed during the period that the body is exposed to irradiation. Proteomics provides the tools for such analyses. Here, the experimental ICR mice were divided into three groups (normal group, model group and 5′-AMP + irradiation group). After different treatment, the hepatic total protein of each animal in three groups was separated by two-dimensional gel electrophoresis (2-DE). 2-DE analysis revealed fifty-eight protein spots were differentially expressed in comparison to three groups. From 58 protein spots, we selected nine spots to identify by MALDI-TOF-MS and received credible results. They were determined to be type I arginase, annexin A5, regucalcin, catalase, Tpm3 protein, Pdia4 protein, 14-3-3 protein epsilon, NAD-Malate dehydrogenase and heat shock protein 90. Considering the characteristic of these proteins, we proposed a possible protection pathway.
Collapse
Affiliation(s)
- Cuilin Cheng
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
- Institute of Extreme Environmental Nutrition and Protection, Harbin Institute of Technology, Harbin 150090, China
| | - Haitian Zhao
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
- Institute of Extreme Environmental Nutrition and Protection, Harbin Institute of Technology, Harbin 150090, China
| | - Zhenyu Wang
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
- Institute of Extreme Environmental Nutrition and Protection, Harbin Institute of Technology, Harbin 150090, China
- School of Forestry, Northeast Forestry University, Harbin 150026, China
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-451-8628-3022; Fax: +86-451-8628-2906
| | - Weihong Lu
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
- Institute of Extreme Environmental Nutrition and Protection, Harbin Institute of Technology, Harbin 150090, China
| | - Lu Wang
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
- Institute of Extreme Environmental Nutrition and Protection, Harbin Institute of Technology, Harbin 150090, China
| | - Rongchun Wang
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
| | - Lei Yao
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (C.C.); (H.Z.); (W.L.); (L.W.); (R.W.); (L.Y.)
- School of Food Science and Engineering, Northeast Agriculture University, Harbin 150030, China
| |
Collapse
|
62
|
Lam T, Thomas LM, White CA, Li G, Pone EJ, Xu Z, Casali P. Scaffold functions of 14-3-3 adaptors in B cell immunoglobulin class switch DNA recombination. PLoS One 2013; 8:e80414. [PMID: 24282540 PMCID: PMC3840166 DOI: 10.1371/journal.pone.0080414] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 10/02/2013] [Indexed: 12/22/2022] Open
Abstract
Class switch DNA recombination (CSR) of the immunoglobulin heavy chain (IgH) locus crucially diversifies antibody biological effector functions. CSR involves the induction of activation-induced cytidine deaminase (AID) expression and AID targeting to switch (S) regions by 14-3-3 adaptors. 14-3-3 adaptors specifically bind to 5'-AGCT-3' repeats, which make up for the core of all IgH locus S regions. They selectively target the upstream and downstream S regions that are set to undergo S-S DNA recombination. We hypothesized that 14-3-3 adaptors function as scaffolds to stabilize CSR enzymatic elements on S regions. Here we demonstrate that all seven 14-3-3β, 14-3-3ε, 14-3-3γ, 14-3-3η, 14-3-3σ, 14-3-3τ and 14-3-3ζ adaptors directly interacted with AID, PKA-Cα (catalytic subunit) and PKA-RIα (regulatory inhibitory subunit) and uracil DNA glycosylase (Ung). 14-3-3 adaptors, however, did not interact with AID C-terminal truncation mutant AIDΔ(180-198) or AIDF193A and AIDL196A point-mutants (which have been shown not to bind to S region DNA and fail to mediate CSR). 14-3-3 adaptors colocalized with AID and replication protein A (RPA) in B cells undergoing CSR. 14-3-3 and AID binding to S region DNA was disrupted by viral protein R (Vpr), an accessory protein of human immunodeficiency virus type-1 (HIV-1), which inhibited CSR without altering AID expression or germline IH-CH transcription. Accordingly, we demonstrated that 14-3-3 directly interact with Vpr, which in turn, also interact with AID, PKA-Cα and Ung. Altogether, our findings suggest that 14-3-3 adaptors play important scaffold functions and nucleate the assembly of multiple CSR factors on S regions. They also show that such assembly can be disrupted by a viral protein, thereby allowing us to hypothesize that small molecule compounds that specifically block 14-3-3 interactions with AID, PKA and/or Ung can be used to inhibit unwanted CSR.
Collapse
Affiliation(s)
- Tonika Lam
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lisa M. Thomas
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
| | - Clayton A. White
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
| | - Guideng Li
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
| | - Egest J. Pone
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
| | - Zhenming Xu
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Paolo Casali
- Institute for Immunology, School of Medicine and School of Biological Sciences, University of California Irvine, Irvine, California, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
63
|
Kleppe R, Ghorbani S, Martinez A, Haavik J. Modelling cellular signal communication mediated by phosphorylation dependent interaction with 14-3-3 proteins. FEBS Lett 2013; 588:92-8. [DOI: 10.1016/j.febslet.2013.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 11/08/2013] [Accepted: 11/12/2013] [Indexed: 11/25/2022]
|
64
|
Abstract
This review focuses on a structure-based analysis of histone posttranslational modification (PTM) readout, where the PTMs serve as docking sites for reader modules as part of larger complexes displaying chromatin modifier and remodeling activities, with the capacity to alter chromatin architecture and templated processes. Individual topics addressed include the diversity of reader-binding pocket architectures and common principles underlying readout of methyl-lysine and methyl-arginine marks, their unmodified counterparts, as well as acetyl-lysine and phosphoserine marks. The review also discusses the impact of multivalent readout of combinations of PTMs localized at specific genomic sites by linked binding modules on processes ranging from gene transcription to repair. Additional topics include cross talk between histone PTMs, histone mimics, epigenetic-based diseases, and drug-based therapeutic intervention. The review ends by highlighting new initiatives and advances, as well as future challenges, toward the promise of enhancing our structural and mechanistic understanding of the readout of histone PTMs at the nucleosomal level.
Collapse
Affiliation(s)
- Dinshaw J Patel
- Structural Biology Department, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA.
| | | |
Collapse
|
65
|
Zhao GY, Ding JY, Lu CL, Lin ZW, Guo J. The overexpression of 14-3-3ζ and Hsp27 promotes non–small cell lung cancer progression. Cancer 2013; 120:652-63. [PMID: 24804299 DOI: 10.1002/cncr.28452] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The 14-3-3ζ protein has been identified as a putative oncoprotein in several cancers, including non–small cell lung cancer (NSCLC). However, the mechanisms underlying its functions have not been well defined. METHODS Proteins that interact with 14-3-3ζ were identified through coimmunoprecipitation and mass spectrometry in NSCLC cells. The interaction of 14-3-3ζ with these molecular partners and their roles in the invasiveness and metastasis of NSCLC cells were assayed through specific disruptions in the 14-3-3ζ signaling network. In addition, the clinical implications of this 14-3-3ζ complex were examined in samples from patients with NSCLC. RESULTS Among the identified proteins that interacted with 14-3-3ζ, there were 230 proteins in 95-D cells, 181 proteins in 95-C cells, and 203 proteins in A549 cells; and 16 interacting proteins were identified that overlapped between all cell lines. Further studies revealed 14-3-3ζ complexes within the heat shock protein 27 (Hsp27) protein and demonstrated that the interference of Hsp27 or 14-3-3ζ inhibited the invasion and metastasis of NSCLC cells. The invasive and metastatic capabilities of cells with both Hsp27 and 14-3-3ζ interference could be completely restored only by Hsp27 and 14-3-3ζ complementary DNA transfection and not by either agent alone. Clinically, the postoperative 5-year overall survival (OS) in patients who had high expression of both 14-3-3ζ and Hsp27 was significantly lower than the 5-year OS in patients who had low expression of both 14-3-3ζ and Hsp27 (26.5% vs 59.7%, respectively). Multivariate analysis revealed that the combined expression of 14-3-3ζ and Hsp27 was an independent prognostic indicator of OS(P = .036). CONCLUSIONS The current data suggest that the combined expression of 14-3-3ζ and Hsp27 may be a biomarker for predicting survival in patients with NSCLC, and this combination may have potential as a therapeutic target for NSCLC.
Collapse
|
66
|
Du Y, Fu RW, Lou B, Zhao J, Qui M, Khuri FR, Fu H. A time-resolved fluorescence resonance energy transfer assay for high-throughput screening of 14-3-3 protein-protein interaction inhibitors. Assay Drug Dev Technol 2013; 11:367-81. [PMID: 23906346 DOI: 10.1089/adt.2013.507] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Protein-protein interaction networks mediate diverse biological processes by regulating various signaling hubs and clusters. 14-3-3 proteins, a family of phosphoserine/threonine-binding molecules, serve as major interaction hubs in eukaryotic cells and have emerged as promising therapeutic targets for various human diseases. In order to identify chemical probes for mechanistic studies and for potential therapeutic development, we have developed highly sensitive bioassays to monitor the interaction of 14-3-3 with a client protein. In this study, we describe a homogenous time-resolved fluorescence resonance energy transfer (TR-FRET) assay to detect the interaction of 14-3-3 with Bad, a proapoptotic member of the Bcl-2 family. Through a series of titration studies in which europium-labeled 14-3-3 serves as an FRET donor and a Dy647-labeled phosphorylated Bad, the peptide acts as an FRET acceptor, we have achieved a robust TR-FRET assay that is suitable for high-throughput screening (HTS) with an excellent signal-to-background ratio of >20 and Z' values >0.7. This assay was further miniaturized to a 1,536-well format for ultra-HTS (uHTS), and exhibited a similar robust performance. The utility and performance of the assay for uHTS were validated by (i) known inhibitors, including peptide R18 and small molecule FOBISIN101, and (ii) screening of a 51,200 compound library. This simple and robust assay is generally applicable to detect the interaction of 14-3-3 with other client proteins. It provides a sensitive and easy-to-use tool to facilitate the discovery of 14-3-3 protein inhibitors as well as to study 14-3-3-mediated protein-protein interactions.
Collapse
Affiliation(s)
- Yuhong Du
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | |
Collapse
|
67
|
Pardo OE, Seckl MJ. S6K2: The Neglected S6 Kinase Family Member. Front Oncol 2013; 3:191. [PMID: 23898460 PMCID: PMC3721059 DOI: 10.3389/fonc.2013.00191] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/08/2013] [Indexed: 01/05/2023] Open
Abstract
S6 kinase 2 (S6K2) is a member of the AGC kinases super-family. Its closest homolog, S6K1, has been extensively studied along the years. However, due to the belief in the community that the high degree of identity between these two isoforms would translate in essentially identical biological functions, S6K2 has been largely neglected. Nevertheless, recent research has clearly highlighted that these two proteins significantly differ in their roles in vitro as well as in vivo. These findings are significant to our understanding of S6 kinase signaling and the development of therapeutic strategies for several diseases including cancer. Here, we will focus on S6K2 and review the protein–protein interactions and specific substrates that determine the selective functions of this kinase.
Collapse
Affiliation(s)
- Olivier E Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital , London , UK
| | | |
Collapse
|
68
|
Molecular tweezers modulate 14-3-3 protein–protein interactions. Nat Chem 2013; 5:234-9. [DOI: 10.1038/nchem.1570] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 01/10/2013] [Indexed: 12/12/2022]
|
69
|
Yin Z, Song Y, Rehse PH. Thymoquinone blocks pSer/pThr recognition by Plk1 Polo-box domain as a phosphate mimic. ACS Chem Biol 2013; 8:303-8. [PMID: 23135290 DOI: 10.1021/cb3004379] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Phosphorylation-dependent protein-protein interaction has rarely been targeted in medicinal chemistry. Thymoquinone, a naturally occurring antitumor agent, disrupts prephosphorylated substrate recognition by the polo-box domain of polo-like kinase 1, a key mitotic regulator responsible for various carcinogenesis when overexpressed. Here, crystallographic studies reveal that the phosphoserine/phosphothreonine recognition site of the polo-box domain is the binding pocket for thymoquinone and its analogue poloxime. Both small molecules displace phosphopeptides bound with the polo-box domain in a slow but noncovalent binding mode. A conserved water bridge and a cation-π interaction were found as their competition strategy against the phosphate group. This mechanism sheds light on small-molecule intervention of phospho-recognition by the polo-box domain of polo-like kinase 1 and other phospho-binding proteins in general.
Collapse
Affiliation(s)
- Zhou Yin
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu 210009, China
| | | | | |
Collapse
|
70
|
Milroy LG, Brunsveld L, Ottmann C. Stabilization and inhibition of protein-protein interactions: the 14-3-3 case study. ACS Chem Biol 2013; 8:27-35. [PMID: 23210482 DOI: 10.1021/cb300599t] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Small-molecule modulation of protein-protein interactions (PPIs) is one of the most exciting but also difficult fields in chemical biology and drug development. As one of the most important "hub" proteins with at least 200-300 interaction partners, the 14-3-3 proteins are an especially fruitful case for PPI intervention. Here, we summarize recent success stories in small-molecule modulation, both inhibition and stabilization, of 14-3-3 PPIs. The chemical breath of modulators includes natural products such as fusicoccin A and derivatives but also compounds identified via high-throughput and in silico screening, which has yielded a toolbox of useful inhibitors and stabilizers for this interesting class of adapter proteins. Protein-protein interactions (PPIs) are involved in almost all biological processes, with any given protein typically engaged in complexes with other proteins for the majority of its lifetime. Hence, proteins function not simply as single, isolated entities but display their roles by interacting with other cellular components. These different interaction patterns are presumably as important as the intrinsic biochemical activity status of the protein itself. The biological role of a protein is therefore decisively dependent on the underlying PPI network that furthermore can show great spatial and temporal variations. A thorough appreciation and understanding of this concept and its regulation mechanisms could help to develop new therapeutic agents and concepts.
Collapse
Affiliation(s)
- Lech-Gustav Milroy
- Laboratory of Chemical
Biology,
Department of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech, 5612 AZ Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical
Biology,
Department of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech, 5612 AZ Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical
Biology,
Department of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech, 5612 AZ Eindhoven, The Netherlands
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn Straße
15, 44227 Dortmund, Germany
| |
Collapse
|
71
|
Maki T, Kawamura A, Kato N, Ohkanda J. Chemical ligation of epoxide-containing fusicoccins and peptide fragments guided by 14-3-3 protein. ACTA ACUST UNITED AC 2013. [DOI: 10.1039/c2mb25388g] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
72
|
Aristizábal-Corrales D, Schwartz S, Cerón J. PAR-5 is a PARty hub in the germline: Multitask proteins in development and disease. WORM 2013; 2:e21834. [PMID: 24058859 PMCID: PMC3670460 DOI: 10.4161/worm.21834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/06/2012] [Accepted: 08/14/2012] [Indexed: 11/19/2022]
Abstract
As our understanding of how molecular machineries work expands, an increasing number of proteins that appear as regulators of different processes have been identified. These proteins are hubs within and among functional networks. The 14-3-3 protein family is involved in multiple cellular pathways and, therefore, influences signaling in several disease processes, from neurobiological disorders to cancer. As a consequence, 14-3-3 proteins are currently being investigated as therapeutic targets. Moreover, 14-3-3 protein levels have been associated with resistance to chemotherapies. There are seven 14-3-3 genes in humans, while Caenorhabditis elegans only possesses two, namely par-5 and ftt-2. Among the C. elegans scientific community, par-5 is mainly recognized as one of the par genes that is essential for the asymmetric first cell division in the embryo. However, a recent study from our laboratory describes roles of par-5 in germ cell proliferation and in the cellular response to DNA damage induced by genotoxic agents. In this review, we explore the broad functionality of 14-3-3 proteins in C. elegans and comment on the potential use of worms for launching a drugs/modifiers discovery platform for the therapeutic regulation of 14-3-3 function in cancer.
Collapse
Affiliation(s)
- David Aristizábal-Corrales
- Drug Delivery and Targeting; CIBBIM-Nanomedicine; Vall d'Hebron Research Institute; Universidad Autónoma de Barcelona; Barcelona, Spain ; Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Barcelona, Spain ; Department of Cancer and Human Molecular Genetics; Bellvitge Biomedical Research Institute (IDIBELL); L'Hospitalet de Llobregat; Barcelona, Spain
| | | | | |
Collapse
|
73
|
Thiel P, Röglin L, Meissner N, Hennig S, Kohlbacher O, Ottmann C. Virtual screening and experimental validation reveal novel small-molecule inhibitors of 14-3-3 protein–protein interactions. Chem Commun (Camb) 2013; 49:8468-70. [DOI: 10.1039/c3cc44612c] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
74
|
Ponterini G. Fluorescence Observables and Enzyme Kinetics in the Investigation of PPI Modulation by Small Molecules: Detection, Mechanistic Insight, and Functional Consequences. DISRUPTION OF PROTEIN-PROTEIN INTERFACES 2013. [PMCID: PMC7123529 DOI: 10.1007/978-3-642-37999-4_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The potential of fluorescence-based methods and kinetic analysis in the screening and molecular-scale mechanistic investigation of PPI modulation by small molecules is discussed through several representative examples collected and commented. These experimental approaches take advantage of a variety of observables. Changes in the protein aggregation pattern have been monitored through fluorescence properties such as spectra, intensities (related to quantum yields), time-decays, and anisotropies of intrinsic protein fluorophores, of extrinsic fluorescent tags and, even, of the same small molecules added to modulate PPIs, as well as through bimolecular excited-state processes such as static and collisional quenching, including electron and excitation-energy transfer, or exciton interaction, whose efficiencies are crucially structure dependent. Besides allowing for qualitative and quantitative information on the small-molecule induced PPI modulation, these approaches can take advantage from the sensitivity of fluorescence observables on fine structural details to shed light on the molecular-scale mechanisms of action and their functional consequences. Direct investigation of the latter by kinetic inhibition analysis represents a useful change in perspective whenever PPI are relevant for enzyme activity. Dissociative inhibition, that is, the ability of some small molecules to inhibit enzymes by disrupting their active oligomeric assembly is shortly reviewed.
Collapse
|
75
|
Ottmann C. Small-molecule modulators of 14-3-3 protein-protein interactions. Bioorg Med Chem 2012; 21:4058-62. [PMID: 23266179 DOI: 10.1016/j.bmc.2012.11.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 11/17/2012] [Accepted: 11/20/2012] [Indexed: 01/07/2023]
Abstract
14-3-3 Proteins are eukaryotic adapter proteins that regulate a plethora of physiological processes by binding to several hundred partner proteins. They play a role in biological activities as diverse as signal transduction, cell cycle regulation, apoptosis, host-pathogen interactions and metabolic control. As such, 14-3-3s are implicated in disease areas like cancer, neurodegeneration, diabetes, pulmonary disease, and obesity. Targeted modulation of 14-3-3 protein-protein interactions (PPIs) by small molecules is therefore an attractive concept for disease intervention. In recent years a number of examples of inhibitors and stabilizers of 14-3-3 PPIs have been reported promising a vivid future in chemical biology and drug development for this remarkable class of proteins.
Collapse
Affiliation(s)
- Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech 2, 5612 AZ Eindhoven, The Netherlands.
| |
Collapse
|
76
|
Kragelund BB, Jensen MK, Skriver K. Order by disorder in plant signaling. TRENDS IN PLANT SCIENCE 2012; 17:625-32. [PMID: 22819467 DOI: 10.1016/j.tplants.2012.06.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 06/19/2012] [Accepted: 06/20/2012] [Indexed: 05/10/2023]
Abstract
Protein intrinsic disorder (ID), referring to the lack of a fixed tertiary structure, is an emerging topic in plant science. Proteins with ID challenge our perception of protein interactions because of their malleable behavior. They are abundant in highly regulated processes such as cellular signaling and transcription, where they exploit the flexibility of ID. In this opinion article we highlight trends in the field of protein ID and discuss its implications for interactions between plant transcription factors (TFs) and the cellular signaling hub protein RADICAL-INDUCED CELL DEATH 1 (RCD1). We envision RCD1-TF interactions as models for translating knowledge of ID-based interactions in vitro to the organismal level in vivo, and urge increased focus on ID in basic plant research and agricultural sciences.
Collapse
Affiliation(s)
- Birthe B Kragelund
- Biomolecular Sciences, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, DK-2200 Copenhagen N, Denmark
| | | | | |
Collapse
|
77
|
Jacobson KA, Balasubramanian R, Deflorian F, Gao ZG. G protein-coupled adenosine (P1) and P2Y receptors: ligand design and receptor interactions. Purinergic Signal 2012; 8:419-36. [PMID: 22371149 PMCID: PMC3360101 DOI: 10.1007/s11302-012-9294-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/30/2012] [Indexed: 12/17/2022] Open
Abstract
The medicinal chemistry and pharmacology of the four subtypes of adenosine receptors (ARs) and the eight subtypes of P2Y receptors (P2YRs, activated by a range of purine and pyrimidine mono- and dinucleotides) has recently advanced significantly leading to selective ligands. X-ray crystallographic structures of both agonist- and antagonist-bound forms of the A(2A)AR have provided unprecedented three-dimensional detail concerning molecular recognition in the binding site and the conformational changes in receptor activation. It is apparent that this ubiquitous cell signaling system has implications for understanding and treating many diseases. ATP and other nucleotides are readily released from intracellular sources under conditions of injury and organ stress, such as hypoxia, ischemia, or mechanical stress, and through channels and vesicular release. Adenosine may be generated extracellularly or by cellular release. Therefore, depending on pathophysiological factors, in a given tissue, there is often a tonic activation of one or more of the ARs or P2YRs that can be modulated by exogenous agents for a beneficial effect. Thus, this field has provided fertile ground for pharmaceutical development, leading to clinical trials of selective receptor ligands as imaging agents or for conditions including cardiac arrhythmias, ischemia/reperfusion injury, diabetes, pain, thrombosis, Parkinson's disease, rheumatoid arthritis, psoriasis, dry eye disease, pulmonary diseases such as cystic fibrosis, glaucoma, cancer, chronic hepatitis C, and other diseases.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
78
|
Molzan M, Ottmann C. Synergistic binding of the phosphorylated S233- and S259-binding sites of C-RAF to one 14-3-3ζ dimer. J Mol Biol 2012; 423:486-95. [PMID: 22922483 DOI: 10.1016/j.jmb.2012.08.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 08/12/2012] [Accepted: 08/14/2012] [Indexed: 01/02/2023]
Abstract
C-RAF kinase is a central component of the Ras-RAF-MEK (mitogen-activated protein kinase/extracellular signal-regulated kinase)-ERK (extracellular signal-regulated kinase) pathway, which has been shown to be activated in 30% of human tumors. 14-3-3 proteins inactivate C-RAF by binding to the two N-terminal phosphorylation-dependent binding sites surrounding S233 and S259. 14-3-3 proteins can bind two target sequences located on one polypeptide chain simultaneously, thereby increasing binding affinity compared to single-site binding and possibly allowing regulated 14-3-3 binding through gatekeeper phosphorylation. To date, it was unclear whether 14-3-3 proteins can bind the two N-terminal phosphorylation-dependent binding sites of C-RAF simultaneously. Fluorescence polarization using phosphorylated peptides demonstrated that S233 is the low-affinity and S259 is the high-affinity binding site, while simultaneous engagement of both sites by 14-3-3ζ enhances affinity compared to single-site binding. Determination of a 1:1 stoichiometry for the di-phosphorylated peptide binding to one 14-3-3ζ dimer with isothermal titration calorimetry was supported by the crystal structure of the 14-3-3ζ/C-RAFpS233,pS259 complex. Cellular localization studies validate the significance of these sites for cytoplasmic retention of C-RAF, suggesting an extended mechanism of RAF regulation by 14-3-3 proteins.
Collapse
Affiliation(s)
- Manuela Molzan
- Chemical Genomics Centre of the Max-Planck-Society, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | | |
Collapse
|
79
|
Haladová K, Mrázek H, Ječmen T, Halada P, Man P, Novák P, Chmelík J, Obšil T, Šulc M. The combination of hydrogen/deuterium exchange or chemical cross-linking techniques with mass spectrometry: mapping of human 14-3-3ζ homodimer interface. J Struct Biol 2012; 179:10-7. [PMID: 22580067 DOI: 10.1016/j.jsb.2012.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 03/27/2012] [Accepted: 04/24/2012] [Indexed: 11/25/2022]
Abstract
Hydrogen/deuterium (H/D) exchange or chemical cross-linking by soluble carbodiimide (EDC) was employed in combination with high-resolution mass spectrometry (MS) to extend our knowledge about contact surface regions involved in the well-characterized model of interaction between two molecules of human 14-3-3ζ regulatory protein. The H/D exchange experiment provided low resolution mapping of interaction in the homodimeric 14-3-3ζ complex. A lower level of deuteration, suggesting structural protection, of two sequential segments has been demonstrated for dimeric 14-3-3ζ wild type relative to the monomeric mutant 14-3-3ζ S58D. The N-terminal sequence (the first 27 residues) from one subunit interacts with region αC'and αD'-helices (residues 45-98) of the other molecule across the dimer interface. To identify interacting amino acid residues within the studied complex, a chemical cross-linking reaction was carried out to produce the covalent homodimer, which was detected by SDS-PAGE. The MS analysis (following tryptic in-gel digestion) employing both high resolution and tandem mass spectrometry revealed cross-linked amino acid residues. Two alternative salt bridges between Glu81 and either Lys9 or the N-terminal amino group have been found to participate in transient interactions of the 14-3-3ζ isotype homodimerization. The data obtained, which have never previously been reported, were used to modify the published 14-3-3 crystal structure using molecular modeling. Based on our findings, utilization of this combination of experimental approaches, which preserve protein native structures, is suitable for mapping the contact between two proteins and also allows for the description of transient interactions or of regions with flexible structure in the studied protein complexes.
Collapse
Affiliation(s)
- Kateřina Haladová
- Institute of Microbiology, v.v.i., Academy of Sciences of the Czech Republic, Vídeňská 1083, CZ-14220 Prague 4, Czech Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Reply to Roglin et al.: Synchrotron radiation-induced covalent modification of 14-3-3 by diazene compounds containing pyridoxal phosphate. Proc Natl Acad Sci U S A 2012. [DOI: 10.1073/pnas.1203341109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
81
|
Covalent attachment of pyridoxal-phosphate derivatives to 14-3-3 proteins. Proc Natl Acad Sci U S A 2012; 109:E1051-3; author reply E1054. [PMID: 22532669 DOI: 10.1073/pnas.1116592109] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
82
|
Richter A, Rose R, Hedberg C, Waldmann H, Ottmann C. An Optimised Small-Molecule Stabiliser of the 14-3-3-PMA2 Protein-Protein Interaction. Chemistry 2012; 18:6520-7. [DOI: 10.1002/chem.201103761] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Indexed: 11/08/2022]
|
83
|
Numata J, Juneja A, Diestler DJ, Knapp EW. Influence of Spacer–Receptor Interactions on the Stability of Bivalent Ligand–Receptor Complexes. J Phys Chem B 2012; 116:2595-604. [DOI: 10.1021/jp211383s] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jorge Numata
- Department of Biology, Chemistry
and Pharmacy, Institute of Chemistry and Biochemistry, Fabeckstrasse 36A, D-14195 Berlin, Germany
| | - Alok Juneja
- Department of Biology, Chemistry
and Pharmacy, Institute of Chemistry and Biochemistry, Fabeckstrasse 36A, D-14195 Berlin, Germany
- Department of Biosciences and
Nutrition, Karolinska Institutet, SE-141
83 Huddinge, Sweden
| | - Dennis J. Diestler
- Department of Biology, Chemistry
and Pharmacy, Institute of Chemistry and Biochemistry, Fabeckstrasse 36A, D-14195 Berlin, Germany
- University of Nebraska-Lincoln, Lincoln, Nebraska 68583,
United States
| | - Ernst-Walter Knapp
- Department of Biology, Chemistry
and Pharmacy, Institute of Chemistry and Biochemistry, Fabeckstrasse 36A, D-14195 Berlin, Germany
| |
Collapse
|
84
|
Johns MA. The international chemical biology community synthesizes a new society. ACS Chem Biol 2012; 7:14-9. [PMID: 22260495 DOI: 10.1021/cb200507f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Margaret A. Johns
- Emory Chemical Biology
Discovery Center, Emory University, Atlanta,
Georgia, United States
| |
Collapse
|
85
|
Abstract
Myeloid leukaemia factor 1 (MLF1) binds to 14-3-3 adapter proteins by a sequence surrounding Ser34 with the functional consequences of this interaction largely unknown. We present here the high-resolution crystal structure of this binding motif [MLF1(29-42)pSer34] in complex with 14-3-3ε and analyse the interaction with isothermal titration calorimetry. Fragment-based ligand discovery employing crystals of the binary 14-3-3ε/MLF1(29-42)pSer34 complex was used to identify a molecule that binds to the interface rim of the two proteins, potentially representing the starting point for the development of a small molecule that stabilizes the MLF1/14-3-3 protein-protein interaction. Such a compound might be used as a chemical biology tool to further analyse the 14-3-3/MLF1 interaction without the use of genetic methods. Database Structural data are available in the Protein Data Bank under the accession number(s) 3UAL [14-3-3ε/MLF1(29-42)pSer34 complex] and 3UBW [14-3-3ε/MLF1(29-42)pSer34/3-pyrrolidinol complex] Structured digital abstract • 14-3-3 epsilon and MLF1 bind by x-ray crystallography (View interaction) • 14-3-3 epsilon and MLF1 bind by isothermal titration calorimetry (View Interaction: 1, 2).
Collapse
Affiliation(s)
- Manuela Molzan
- Chemical Genomics Centre of the Max-Planck-Society, Dortmund, Germany
| | | | | | | |
Collapse
|
86
|
|
87
|
Zhao J, Meyerkord CL, Du Y, Khuri FR, Fu H. 14-3-3 proteins as potential therapeutic targets. Semin Cell Dev Biol 2011; 22:705-12. [PMID: 21983031 DOI: 10.1016/j.semcdb.2011.09.012] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 02/05/2023]
Abstract
The 14-3-3 family of phosphoserine/phosphothreonine-binding proteins dynamically regulates the activity of client proteins in various signaling pathways that control diverse physiological and pathological processes. In response to environmental cues, 14-3-3 proteins orchestrate the highly regulated flow of signals through complex networks of molecular interactions to achieve well-controlled physiological outputs, such as cell proliferation or differentiation. Accumulating evidence now supports the concept that either an abnormal state of 14-3-3 protein expression, or dysregulation of 14-3-3/client protein interactions, contributes to the development of a large number of human diseases. In particular, clinical investigations in the field of oncology have demonstrated a correlation between upregulated 14-3-3 levels and poor survival of cancer patients. These studies highlight the rapid emergence of 14-3-3 proteins as a novel class of molecular target for potential therapeutic intervention. The current status of 14-3-3 modulator discovery is discussed.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | | | |
Collapse
|
88
|
Goc A, Abdalla M, Al-Azayzih A, Somanath PR. Tuning cell cycle regulation with an iron key. PLoS One 2007; 7:e40594. [PMID: 22808202 PMCID: PMC3396618 DOI: 10.1371/journal.pone.0040594] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/11/2012] [Indexed: 01/20/2023] Open
Abstract
14-3-3 proteins are ubiquitously expressed dimeric adaptor proteins that have emerged as key mediators of many cell signaling pathways in multiple cell types. Its effects are mainly mediated by binding to selective phosphoserine/threonine proteins. The importance of 14-3-3 proteins in cancer have only started to become apparent and its exact role in cancer progression as well as the mechanisms by which 14-3-3 proteins mediate cancer cell function remain unknown. While protein 14-3-3σ is widely accepted as a tumor suppressor, 14-3-3ζ, β and γ isoforms have been shown to have tumor promoting effects. Despite the importance of 14-3-3 family in mediating various cell processes, the exact role and mechanism of 14-3-3ζ remain unexplored. In the current study, we investigated the role of protein 14-3-3ζ in prostate cancer cell motility and transendothelial migration using biochemical, molecular biology and electric cell-substrate impedance sensing approaches as well as cell based functional assays. Our study indicated that expression with wild-type protein 14-3-3ζ significantly enhanced Rac activity in PC3 cells. In contrast, expression of dimer-resistant mutant of protein 14-3-3ζ (DM-14-3-3) inhibited Rac activity and associated phosphorylation of p21 activated kinase-1 and 2. Expression with wild-type 14-3-3ζ or constitutively active Rac1 enhanced extracellular matrix recognition, lamellipodia formation, cell migration and trans-endothelial migration by PC3 cells. In contrast, expression with DM 14-3-3ζ or DN-Rac1 in PC3 cells significantly inhibited these cell functions. Our results demonstrate for the first time that 14-3-3ζ enhances prostate cancer cell-matrix interactions, motility and transendothelial migration in vitro via activation of Rac1-GTPase and is an important target for therapeutic interventions for prostate cancer.
Collapse
Affiliation(s)
- Anna Goc
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
| | - Maha Abdalla
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
| | - Ahmad Al-Azayzih
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- Department of Medicine, Georgia Health Sciences University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|