1
|
Wozniak JM, Li W, Parker CG. Chemical proteomic mapping of reversible small molecule binding sites in native systems. Trends Pharmacol Sci 2024; 45:969-981. [PMID: 39406592 DOI: 10.1016/j.tips.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 11/10/2024]
Abstract
The impact of small molecules in human biology are manifold; not only are they critical regulators of physiological processes, but they also serve as probes to investigate biological pathways and leads for therapeutic development. Identifying the protein targets of small molecules, and where they bind, is critical to understanding their functional consequences and potential for pharmacological use. Over the past two decades, chemical proteomics has emerged as a go-to strategy for the comprehensive mapping of small molecule-protein interactions. Recent advancements in this field, particularly innovations of photoaffinity labeling (PAL)-based methods, have enabled the robust identification of small molecule binding sites on protein targets, often in live cells. In this opinion article, we examine these advancements as well as reflect on how their strategic integration with other emerging tools can advance therapeutic development.
Collapse
Affiliation(s)
| | - Weichao Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
2
|
Sheng L, Bhalla R. Biomarkers and Target-Specific Small-Molecule Drugs in Alzheimer's Diagnostic and Therapeutic Research: From Amyloidosis to Tauopathy. Neurochem Res 2024; 49:2273-2302. [PMID: 38844706 PMCID: PMC11310295 DOI: 10.1007/s11064-024-04178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 08/09/2024]
Abstract
Alzheimer's disease (AD) is the most common type of human dementia and is responsible for over 60% of diagnosed dementia cases worldwide. Abnormal deposition of β-amyloid and the accumulation of neurofibrillary tangles have been recognised as the two pathological hallmarks targeted by AD diagnostic imaging as well as therapeutics. With the progression of pathological studies, the two hallmarks and their related pathways have remained the focus of researchers who seek for AD diagnostic and therapeutic strategies in the past decades. In this work, we reviewed the development of the AD biomarkers and their corresponding target-specific small molecule drugs for both diagnostic and therapeutic applications, underlining their success, failure, and future possibilities.
Collapse
Affiliation(s)
- Li Sheng
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
3
|
Krieger IV, Yalamanchili S, Dickson P, Engelhart CA, Zimmerman MD, Wood J, Clary E, Nguyen J, Thornton N, Centrella PA, Chan B, Cuozzo JW, Gengenbacher M, Guié MA, Guilinger JP, Bienstock C, Hartl H, Hupp CD, Jetson R, Satoh T, Yeoman JTS, Zhang Y, Dartois V, Schnappinger D, Keefe AD, Sacchettini JC. Inhibitors of the Thioesterase Activity of Mycobacterium tuberculosis Pks13 Discovered Using DNA-Encoded Chemical Library Screening. ACS Infect Dis 2024; 10:1561-1575. [PMID: 38577994 PMCID: PMC11091879 DOI: 10.1021/acsinfecdis.3c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
DNA-encoded chemical library (DEL) technology provides a time- and cost-efficient method to simultaneously screen billions of compounds for their affinity to a protein target of interest. Here we report its use to identify a novel chemical series of inhibitors of the thioesterase activity of polyketide synthase 13 (Pks13) from Mycobacterium tuberculosis (Mtb). We present three chemically distinct series of inhibitors along with their enzymatic and Mtb whole cell potency, the measure of on-target activity in cells, and the crystal structures of inhibitor-enzyme complexes illuminating their interactions with the active site of the enzyme. One of these inhibitors showed a favorable pharmacokinetic profile and demonstrated efficacy in an acute mouse model of tuberculosis (TB) infection. These findings and assay developments will aid in the advancement of TB drug discovery.
Collapse
Affiliation(s)
- Inna V. Krieger
- Department
of Biochemistry & Biophysics, Texas
A&M University, College
Station, Texas 77843, United States
| | | | - Paige Dickson
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Curtis A. Engelhart
- Department
of Microbiology and Immunology, Weill Cornell
Medicine, New York, New York 10021, United States
| | - Matthew D Zimmerman
- Center for
Discovery and Innovation, Hackensack Meridian
Health, Nutley, New Jersey 07110, United States
| | - Jeremy Wood
- Department
of Biochemistry & Biophysics, Texas
A&M University, College
Station, Texas 77843, United States
| | - Ethan Clary
- Department
of Biochemistry & Biophysics, Texas
A&M University, College
Station, Texas 77843, United States
| | - Jasmine Nguyen
- Department
of Biochemistry & Biophysics, Texas
A&M University, College
Station, Texas 77843, United States
| | - Natalie Thornton
- Department
of Microbiology and Immunology, Weill Cornell
Medicine, New York, New York 10021, United States
| | - Paolo A. Centrella
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Betty Chan
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- Auron
Therapeutics, 55 Chapel
Street, Newton, Massachusetts 02458, United States
| | - John W Cuozzo
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- Relay
Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Martin Gengenbacher
- Center for
Discovery and Innovation, Hackensack Meridian
Health, Nutley, New Jersey 07110, United States
| | - Marie-Aude Guié
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - John P Guilinger
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Corey Bienstock
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Hajnalka Hartl
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- Orogen
Therapeutics, 12 Gill
Street, Woburn, Massachusetts 01801, United States
| | - Christopher D. Hupp
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- Ipsen Bioscience
Inc., 1 Main Street, Cambridge, Massachusetts 02142, United States
| | - Rachael Jetson
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- Valo
Health, 75 Hayden Avenue, Lexington, Massachusetts 02141, United States
| | - Takashi Satoh
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- EXO
Therapeutics, 150 Cambridgepark
Drive, suite 300, Cambridge, Massachusetts 02140, United States
| | - John T. S. Yeoman
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
- Recludix
Pharmaceuticals, 222
Third Street, Cambridge, Massachusetts 02142, United States
| | - Ying Zhang
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Veronique Dartois
- Center for
Discovery and Innovation, Hackensack Meridian
Health, Nutley, New Jersey 07110, United States
- Hackensack
Meridian School of Medicine, Hackensack
Meridian Health, Nutley, New Jersey 07110, United States
| | - Dirk Schnappinger
- Department
of Microbiology and Immunology, Weill Cornell
Medicine, New York, New York 10021, United States
| | - Anthony D. Keefe
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - James C. Sacchettini
- Department
of Biochemistry & Biophysics, Texas
A&M University, College
Station, Texas 77843, United States
| |
Collapse
|
4
|
Ando S, Tanaka K, Matsumoto M, Oyama Y, Tomabechi Y, Yamagata A, Shirouzu M, Nakagawa R, Okimoto N, Taiji M, Sato K, Ohama T. The luciferase-based in vivo protein-protein interaction assay revealed that CHK1 promotes PP2A and PME-1 interaction. J Biol Chem 2024; 300:107277. [PMID: 38588804 PMCID: PMC11098961 DOI: 10.1016/j.jbc.2024.107277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024] Open
Abstract
Protein phosphatase 2A (PP2A) is an essential serine/threonine protein phosphatase, and its dysfunction is involved in the onset of cancer and neurodegenerative disorders. PP2A functions as a trimeric holoenzyme whose composition is regulated by the methyl-esterification (methylation) of the PP2A catalytic subunit (PP2Ac). Protein phosphatase methylesterase-1 (PME-1) is the sole PP2Ac methylesterase, and the higher PME-1 expression is observed in various cancer and neurodegenerative diseases. Apart from serving as a methylesterase, PME-1 acts as a PP2A inhibitory protein, binding directly to PP2Ac and suppressing its activity. The intricate function of PME-1 hinders drug development by targeting the PME-1/PP2Ac axis. This study applied the NanoBiT system, a bioluminescence-based protein interaction assay, to elucidate the molecular mechanism that modulates unknown PME-1/PP2Ac protein-protein interaction (PPI). Compound screening identified that the CHK1 inhibitors inhibited PME-1/PP2Ac association without affecting PP2Ac methylation levels. CHK1 directly phosphorylates PP2Ac to promote PME-1 association. Phospho-mass spectrometry identified multiple phospho-sites on PP2Ac, including the Thr219, that affect PME-1 interaction. An anti-phospho-Thr219 PP2Ac antibody was generated and showed that CHK1 regulates the phosphorylation levels of this site in cells. On the contrary, in vitro phosphatase assay showed that CHK1 is the substrate of PP2A, and PME-1 hindered PP2A-mediated dephosphorylation of CHK1. Our data provides novel insights into the molecular mechanisms governing the PME-1/PP2Ac PPI and the triad relationship between PP2A, PME-1, and CHK1.
Collapse
Affiliation(s)
- Sana Ando
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Keiko Tanaka
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Maharu Matsumoto
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yuki Oyama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yuri Tomabechi
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Atsushi Yamagata
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Reiko Nakagawa
- Laboratory for Cell-Free Protein Synthesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Noriaki Okimoto
- Laboratory for Computational Molecular Design, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan; Drug Discovery Molecular Simulation Platform Unit, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Makoto Taiji
- Laboratory for Computational Molecular Design, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan; Drug Discovery Molecular Simulation Platform Unit, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan; Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan; Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
5
|
Porta EOJ. Mapping the Evolution of Activity-Based Protein Profiling: A Bibliometric Review. Adv Pharm Bull 2023; 13:639-645. [PMID: 38022804 PMCID: PMC10676541 DOI: 10.34172/apb.2023.082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/24/2023] [Accepted: 05/17/2023] [Indexed: 12/01/2023] Open
Abstract
Activity-based protein profiling (ABPP) is a chemoproteomic approach that employs small-molecule probes to directly evaluate protein functionality within complex proteomes. This technology has proven to be a potent strategy for mapping ligandable sites in organisms and has significantly impacted drug discovery processes by enabling the development of highly selective small-molecule inhibitors and the identification of new therapeutic molecular targets. Despite being nearly a quarter of a century old as a chemoproteomic tool, ABPP has yet to undergo a bibliometric analysis. In order to gauge its scholarly impact and evolution, a bibliometric analysis was performed, comparing all 1919 reported articles with the articles published in the last five years. Through a comprehensive data analysis, including a 5-step workflow, the most influential articles were identified, and their bibliometric parameters were determined. The 1919 analyzed articles span from 1999 to 2022, providing a comprehensive overview of the historical and current state of ABPP research. This analysis presents, for the first time, the characteristics of the most influential ABPP articles, offering valuable insight into the research conducted in this field and its potential future directions. The findings underscore the crucial role of ABPP in drug discovery and novel therapeutic target identification, as well as the need for continued advancements in the development of novel chemical probes and proteomic technologies to further expand the utility of ABPP.
Collapse
|
6
|
Haanen TJ, O'Connor CM, Narla G. Biased holoenzyme assembly of protein phosphatase 2A (PP2A): From cancer to small molecules. J Biol Chem 2022; 298:102656. [PMID: 36328247 PMCID: PMC9707111 DOI: 10.1016/j.jbc.2022.102656] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a family of serine threonine phosphatases responsible for regulating protein phosphorylation, thus opposing the activity of cellular kinases. PP2A is composed of a catalytic subunit (PP2A Cα/β) and scaffolding subunit (PP2A Aα/β) and various substrate-directing B regulatory subunits. PP2A biogenesis is regulated at multiple levels. For example, the sequestration of the free catalytic subunit during the process of biogenesis avoids promiscuous phosphatase activity. Posttranslational modifications of PP2A C direct PP2A heterotrimeric formation. Additionally, PP2A functions as a haploinsufficient tumor suppressor, where attenuated PP2A enzymatic activity creates a permissive environment for oncogenic transformation. Recent work studying PP2A in cancer showed that its role in tumorigenesis is more nuanced, with some holoenzymes being tumor suppressive, while others are required for oncogenic transformation. In cancer biology, PP2A function is modulated through various mechanisms including the displacement of specific B regulatory subunits by DNA tumor viral antigens, by recurrent mutations, and through loss of carboxymethyl-sensitive heterotrimeric complexes. In aggregate, these alterations bias PP2A activity away from its tumor suppressive functions and toward oncogenic ones. From a therapeutic perspective, molecular glues and disruptors present opportunities for both the selective stabilization of tumor-suppressive holoenzymes and disruption of holoenzymes that are pro-oncogenic. Collectively, these approaches represent an attractive cancer therapy for a wide range of tumor types. This review will discuss the mechanisms by which PP2A holoenzyme formation is dysregulated in cancer and the current therapies that are aimed at biasing heterotrimer formation of PP2A for the treatment of cancer.
Collapse
Affiliation(s)
- Terrance J Haanen
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Caitlin M O'Connor
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
7
|
Li Y, Balakrishnan VK, Rowse M, Wu CG, Bravos AP, Yadav VK, Ivarsson YI, Strack S, Novikova IV, Xing Y. Coupling to short linear motifs creates versatile PME-1 activities in PP2A holoenzyme demethylation and inhibition. eLife 2022; 11:79736. [PMID: 35924897 PMCID: PMC9398451 DOI: 10.7554/elife.79736] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022] Open
Abstract
Protein phosphatase 2A (PP2A) holoenzymes target broad substrates by recognizing short motifs via regulatory subunits. PP2A methylesterase 1 (PME-1) is a cancer-promoting enzyme and undergoes methylesterase activation upon binding to the PP2A core enzyme. Here, we showed that PME-1 readily demethylates different families of PP2A holoenzymes and blocks substrate recognition in vitro. The high-resolution cryoelectron microscopy structure of a PP2A-B56 holoenzyme–PME-1 complex reveals that PME-1 disordered regions, including a substrate-mimicking motif, tether to the B56 regulatory subunit at remote sites. They occupy the holoenzyme substrate-binding groove and allow large structural shifts in both holoenzyme and PME-1 to enable multipartite contacts at structured cores to activate the methylesterase. B56 interface mutations selectively block PME-1 activity toward PP2A-B56 holoenzymes and affect the methylation of a fraction of total cellular PP2A. The B56 interface mutations allow us to uncover B56-specific PME-1 functions in p53 signaling. Our studies reveal multiple mechanisms of PME-1 in suppressing holoenzyme functions and versatile PME-1 activities derived from coupling substrate-mimicking motifs to dynamic structured cores.
Collapse
Affiliation(s)
- Yitong Li
- Department of Oncology, University of Wisconsin-Madison, Madison, United States
| | | | - Michael Rowse
- Indiana University - Purdue University Columbus, Columbus, United States
| | - Cheng-Guo Wu
- Department of Oncology, University of Wisconsin-Madison, Madison, United States
| | | | - Vikash K Yadav
- 5Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - YIva Ivarsson
- 5Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, United States
| | - Irina V Novikova
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, United States
| | - Yongna Xing
- Department of Oncology, University of Wisconsin-Madison, Madison, United States
| |
Collapse
|
8
|
Suć Sajko J, Jerić I. Synthesis of N β-Substituted 1,2-Diazetidin-3-ones by the Ugi Reaction Comprising Chiral α-Hydrazino Acids. J Org Chem 2022; 87:7076-7084. [PMID: 35549347 DOI: 10.1021/acs.joc.2c00238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report the synthesis of a structurally diverse library of chiral Nβ-substituted 1,2-diazetidin-3-ones by a 4-center 3-component Ugi reaction comprising unprotected α-hydrazino acids. Various isocyanides and carbonyl compounds and both aldehydes and ketones were utilized. In addition, postcondensation modifications at three different sites have been demonstrated.
Collapse
Affiliation(s)
- Josipa Suć Sajko
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Ivanka Jerić
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
9
|
Khan MM, Kalim UU, Khan MH, Lahesmaa R. PP2A and Its Inhibitors in Helper T-Cell Differentiation and Autoimmunity. Front Immunol 2022; 12:786857. [PMID: 35069561 PMCID: PMC8766794 DOI: 10.3389/fimmu.2021.786857] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric Ser/Thr phosphatase that regulates many cellular processes. The role of PP2A as a tumor suppressor has been extensively studied and reviewed. However, emerging evidence suggests PP2A constrains inflammatory responses and is important in autoimmune and neuroinflammatory diseases. Here, we reviewed the existing literature on the role of PP2A in T-cell differentiation and autoimmunity. We have also discussed the modulation of PP2A activity by endogenous inhibitors and its small-molecule activators as potential therapeutic approaches against autoimmunity.
Collapse
Affiliation(s)
- Mohd Moin Khan
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine (TuDMM), University of Turku, Turku, Finland
| | - Ubaid Ullah Kalim
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Meraj H. Khan
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
10
|
Ikeda S, Tsuji S, Ohama T, Sato K. Involvement of PP2A methylation in the adipogenic differentiation of bone marrow-derived mesenchymal stem cell. J Biochem 2021; 168:643-650. [PMID: 32663263 DOI: 10.1093/jb/mvaa077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/20/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent stem cells with ability to self-replicate and differentiate into mesodermal derivatives, such as adipocytes and osteoblasts. BM-MSCs are a critical component of the tumour microenvironment. They support tumour progression by recruiting additional BM-MSCs and by differentiating into myofibroblasts (also called cancer-associated fibroblasts). Protein phosphatase 2A (PP2A) is an essential serine/threonine protein phosphatase that regulates a broad range of cellular signalling. PP2A forms a heterotrimer to dephosphorylate specific substrates. The reversible methylesterification (methylation) of Leu309 in the catalytic subunit of PP2A (PP2Ac) regulates biogenesis of the PP2A holoenzyme. It is unknown whether the methylation of PP2Ac plays a role in BM-MSC differentiation. Our experiments determined that protein levels of PP2A subunits and PP2A methyltransferase (LCMT-1) are significantly altered during differentiation. PP2Ac methylation levels in BM-MSCs decrease over time in response to an adipogenic differentiation stimulus. However, blockage of PP2A demethylation using the PP2A dimethyl-esterase inhibitors enhanced adipocyte differentiation. This suggests that PP2Ac demethylation is involved in adipocyte differentiation resistance. The results of our study provide a greater understanding of the regulation of BM-MSCs differentiation by PP2A holoenzyme.
Collapse
Affiliation(s)
- Shunta Ikeda
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Shunya Tsuji
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| |
Collapse
|
11
|
Meng L, Lu C, Wu B, Lan C, Mo L, Chen C, Wang X, Zhang N, Lan L, Wang Q, Zeng X, Li X, Tang S. Taurine Antagonizes Macrophages M1 Polarization by Mitophagy-Glycolysis Switch Blockage via Dragging SAM-PP2Ac Transmethylation. Front Immunol 2021; 12:648913. [PMID: 33912173 PMCID: PMC8071881 DOI: 10.3389/fimmu.2021.648913] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
The excessive M1 polarization of macrophages drives the occurrence and development of inflammatory diseases. The reprogramming of macrophages from M1 to M2 can be achieved by targeting metabolic events. Taurine promotes for the balance of energy metabolism and the repair of inflammatory injury, preventing chronic diseases and complications. However, little is known about the mechanisms underlying the action of taurine modulating the macrophage polarization phenotype. In this study, we constructed a low-dose LPS/IFN-γ-induced M1 polarization model to simulate a low-grade pro-inflammatory process. Our results indicate that the taurine transporter TauT/SlC6A6 is upregulated at the transcriptional level during M1 macrophage polarization. The nutrient uptake signal on the membrane supports the high abundance of taurine in macrophages after taurine supplementation, which weakens the status of methionine metabolism, resulting in insufficient S-adenosylmethionine (SAM). The low availability of SAM is directly sensed by LCMT-1 and PME-1, hindering PP2Ac methylation. PP2Ac methylation was found to be necessary for M1 polarization, including the positive regulation of VDAC1 and PINK1. Furthermore, its activation was found to promote the elimination of mitochondria by macrophages via the mitophagy pathway for metabolic adaptation. Mechanistically, taurine inhibits SAM-dependent PP2Ac methylation to block PINK1-mediated mitophagy flux, thereby maintaining a high mitochondrial density, which ultimately hinders the conversion of energy metabolism to glycolysis required for M1. Our findings reveal a novel mechanism of taurine-coupled M1 macrophage energy metabolism, providing novel insights into the occurrence and prevention of low-grade inflammation, and propose that the sensing of taurine and SAM availability may allow communication to inflammatory response in macrophages.
Collapse
Affiliation(s)
- Ling Meng
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Cailing Lu
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Bin Wu
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Chunhua Lan
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Laiming Mo
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| | - Chengying Chen
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xinhang Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Ning Zhang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Li Lan
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Qihui Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xia Zeng
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xiyi Li
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Shen Tang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
12
|
Cao WB, Jiang S, Li HY, Xu XP, Ji SJ. Synthesis of strained 1,2-diazetidines via [3 + 1] cycloaddition of C,N-cyclic azomethine imines with isocyanides and synthetic derivation. Org Chem Front 2021. [DOI: 10.1039/d1qo00130b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Strained diazetidines are assembled simply from 1,3-dipolar cycloaddition of isocyanides and C,N-cyclic azomethine imines, and their diversified transformations are presented.
Collapse
Affiliation(s)
- Wen-Bin Cao
- Key Laboratory of Organic Synthesis of Jiangsu Province
- College of Chemistry
- Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Soochow University
- Suzhou 215123
| | - Shuai Jiang
- Key Laboratory of Organic Synthesis of Jiangsu Province
- College of Chemistry
- Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Soochow University
- Suzhou 215123
| | - Hai-Yan Li
- Analysis and Testing Center
- Soochow University
- Suzhou 215123
- China
| | - Xiao-Ping Xu
- Key Laboratory of Organic Synthesis of Jiangsu Province
- College of Chemistry
- Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Soochow University
- Suzhou 215123
| | - Shun-Jun Ji
- Key Laboratory of Organic Synthesis of Jiangsu Province
- College of Chemistry
- Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Soochow University
- Suzhou 215123
| |
Collapse
|
13
|
Nasa I, Kettenbach AN. Effects of carboxyl-terminal methylation on holoenzyme function of the PP2A subfamily. Biochem Soc Trans 2020; 48:2015-2027. [PMID: 33125487 PMCID: PMC8380034 DOI: 10.1042/bst20200177] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 01/07/2023]
Abstract
Phosphoprotein Phosphatases (PPPs) are enzymes highly conserved from yeast and human and catalyze the majority of the serine and threonine dephosphorylation in cells. To achieve substrate specificity and selectivity, PPPs form multimeric holoenzymes consisting of catalytic, structural/scaffolding, and regulatory subunits. For the Protein Phosphatase 2A (PP2A)-subfamily of PPPs, holoenzyme assembly is at least in part regulated by an unusual carboxyl-terminal methyl-esterification, commonly referred to as 'methylation'. Carboxyl-terminal methylation is catalyzed by Leucine carboxyl methyltransferase-1 (LCMT1) that utilizes S-adenosyl-methionine (SAM) as the methyl donor and removed by protein phosphatase methylesterase 1 (PME1). For PP2A, methylation dictates regulatory subunit selection and thereby downstream phosphorylation signaling. Intriguingly, there are four families of PP2A regulatory subunits, each exhibiting different levels of methylation sensitivity. Thus, changes in PP2A methylation stoichiometry alters the complement of PP2A holoenzymes in cells and creates distinct modes of kinase opposition. Importantly, selective inactivation of PP2A signaling through the deregulation of methylation is observed in several diseases, most prominently Alzheimer's disease (AD). In this review, we focus on how carboxyl-terminal methylation of the PP2A subfamily (PP2A, PP4, and PP6) regulates holoenzyme function and thereby phosphorylation signaling, with an emphasis on AD.
Collapse
Affiliation(s)
- Isha Nasa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH, U.S.A
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center at Dartmouth, Lebanon, NH, U.S.A
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH, U.S.A
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center at Dartmouth, Lebanon, NH, U.S.A
| |
Collapse
|
14
|
Wu B, Cai H, Tang S, Xu Y, Shi Q, Wei L, Meng L, Zhang N, Wang X, Xiao D, Zou Y, Yang X, Li X, Lu C. Methionine-Mediated Protein Phosphatase 2A Catalytic Subunit (PP2Ac) Methylation Ameliorates the Tauopathy Induced by Manganese in Cell and Animal Models. Neurotherapeutics 2020; 17:1878-1896. [PMID: 32959271 PMCID: PMC7851222 DOI: 10.1007/s13311-020-00930-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 01/10/2023] Open
Abstract
The molecular mechanism of Alzheimer-like cognitive impairment induced by manganese (Mn) exposure has not yet been fully clarified, and there are currently no effective interventions to treat neurodegenerative lesions related to manganism. Protein phosphatase 2 A (PP2A) is a major tau phosphatase and was recently identified as a potential therapeutic target molecule for neurodegenerative diseases; its activity is directed by the methylation status of the catalytic C subunit. Methionine is an essential amino acid, and its downstream metabolite S-adenosylmethionine (SAM) participates in transmethylation pathways as a methyl donor. In this study, the neurotoxic mechanism of Mn and the protective effect of methionine were evaluated in Mn-exposed cell and rat models. We show that Mn-induced neurotoxicity is characterized by PP2Ac demethylation accompanied by abnormally decreased LCMT-1 and increased PME-1, which are associated with tau hyperphosphorylation and spatial learning and memory deficits, and that the poor availability of SAM in the hippocampus is likely to determine the loss of PP2Ac methylation. Importantly, maintenance of local SAM levels through continuous supplementation with exogenous methionine, or through specific inhibition of PP2Ac demethylation by ABL127 administration in vitro, can effectively prevent tau hyperphosphorylation to reduce cellular oxidative stress, apoptosis, damage to cell viability, and rat memory deficits in cell or animal Mn exposure models. In conclusion, our data suggest that SAM and PP2Ac methylation may be novel targets for the treatment of Mn poisoning and neurotoxic mechanism-related tauopathies.
Collapse
Affiliation(s)
- Bin Wu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Haiqing Cai
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Shen Tang
- School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yilu Xu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Qianqian Shi
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Lancheng Wei
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Ling Meng
- School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Ning Zhang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xinhang Wang
- School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Deqiang Xiao
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yunfeng Zou
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiaobo Yang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiyi Li
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| | - Cailing Lu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
15
|
Reduced Expression of the PP2A Methylesterase, PME-1, or the PP2A Methyltransferase, LCMT-1, Alters Sensitivity to Beta-Amyloid-Induced Cognitive and Electrophysiological Impairments in Mice. J Neurosci 2020; 40:4596-4608. [PMID: 32341098 DOI: 10.1523/jneurosci.2983-19.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/27/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Beta-amyloid (Aβ) is thought to play a critical role in Alzheimer's disease (AD), and application of soluble oligomeric forms of Aβ produces AD-like impairments in cognition and synaptic plasticity in experimental systems. We found previously that transgenic overexpression of the PP2A methylesterase, PME-1, or the PP2A methyltransferase, LCMT-1, altered the sensitivity of mice to Aβ-induced impairments, suggesting that PME-1 inhibition may be an effective approach for preventing or treating these impairments. To explore this possibility, we examined the behavioral and electrophysiological effects of acutely applied synthetic Aβ oligomers in male and female mice heterozygous for either a PME-1 KO or an LCMT-1 gene-trap mutation. We found that heterozygous PME-1 KO mice were resistant to Aβ-induced impairments in cognition and synaptic plasticity, whereas LCMT-1 gene-trap mice showed increased sensitivity to Aβ-induced impairments. The heterozygous PME-1 KO mice produced normal levels of endogenous Aβ and exhibited normal electrophysiological responses to picomolar concentrations of Aβ, suggesting that reduced PME-1 expression in these animals protects against Aβ-induced impairments without impacting normal physiological Aβ functions. Together, these data provide additional support for roles for PME-1 and LCMT-1 in regulating sensitivity to Aβ-induced impairments, and suggest that inhibition of PME-1 may constitute a viable therapeutic approach for selectively protecting against the pathologic actions of Aβ in AD.SIGNIFICANCE STATEMENT Elevated levels of β-amyloid (Aβ) in the brain are thought to contribute to the cognitive impairments observed in Alzheimer's disease patients. Here we show that genetically reducing endogenous levels of the PP2A methylesterase, PME-1, prevents the cognitive and electrophysiological impairments caused by acute exposure to pathologic concentrations of Aβ without impairing normal physiological Aβ function or endogenous Aβ production. Conversely, reducing endogenous levels of the PP2A methyltransferase, LCMT-1, increases sensitivity to Aβ-induced impairments. These data offer additional insights into the molecular factors that control sensitivity to Aβ-induced impairments, and suggest that inhibiting PME-1 may constitute a viable therapeutic avenue for preventing Aβ-related impairments in Alzheimer's disease.
Collapse
|
16
|
Shenoy VM, Thompson BR, Shi J, Zhu HJ, Smith DE, Amidon GL. Chemoproteomic Identification of Serine Hydrolase RBBP9 as a Valacyclovir-Activating Enzyme. Mol Pharm 2020; 17:1706-1714. [DOI: 10.1021/acs.molpharmaceut.0c00131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Vikram M. Shenoy
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| | - Brian R. Thompson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| | - Jian Shi
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | - David E. Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| | - Gordon L. Amidon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| |
Collapse
|
17
|
Dalton SE, Campos S. Covalent Small Molecules as Enabling Platforms for Drug Discovery. Chembiochem 2020; 21:1080-1100. [DOI: 10.1002/cbic.201900674] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Samuel E. Dalton
- Astex Pharmaceuticals 436 Cambridge Science Park Milton Road Cambridge CB4 0QA UK
| | - Sebastien Campos
- PharmaronDrug Discovery Services Europe Hertford Road Hoddesdon Hertfordshire EN11 9BU UK
| |
Collapse
|
18
|
Remmerie M, Janssens V. PP2A: A Promising Biomarker and Therapeutic Target in Endometrial Cancer. Front Oncol 2019; 9:462. [PMID: 31214504 PMCID: PMC6558005 DOI: 10.3389/fonc.2019.00462] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the use of targeted therapies has immensely increased in the treatment of cancer. However, treatment for endometrial carcinomas (ECs) has lagged behind, although potential molecular markers have been identified. This is particularly problematic for the type II ECs, since these aggressive tumors are usually not responsive toward the current standard therapies. Therefore, type II ECs are responsible for most EC-related deaths, indicating the need for new treatment options. Interestingly, molecular analyses of type II ECs have uncovered frequent genetic alterations (up to 40%) in PPP2R1A, encoding the Aα subunit of the tumor suppressive heterotrimeric protein phosphatase type 2A (PP2A). PPP2R1A mutations were also reported in type I ECs and other common gynecologic cancers, albeit at much lower frequencies (0-7%). Nevertheless, PP2A inactivation in the latter cancer types is common via other mechanisms, in particular by increased expression of Cancerous Inhibitor of PP2A (CIP2A) and PP2A Methylesterase-1 (PME-1) proteins. In this review, we discuss the therapeutic potential of direct and indirect PP2A targeting compounds, possibly in combination with other anti-cancer drugs, in EC. Furthermore, we investigate the potential of the PP2A status as a predictive and/or prognostic marker for type I and II ECs.
Collapse
Affiliation(s)
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Javadpour P, Dargahi L, Ahmadiani A, Ghasemi R. To be or not to be: PP2A as a dual player in CNS functions, its role in neurodegeneration, and its interaction with brain insulin signaling. Cell Mol Life Sci 2019; 76:2277-2297. [PMID: 30874837 PMCID: PMC11105459 DOI: 10.1007/s00018-019-03063-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/16/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
Abstract
Accumulating evidence has reached the consensus that the balance of phosphorylation state of signaling molecules is a pivotal point in the regulation of cell signaling. Therefore, characterizing elements (kinases-phosphatases) in the phosphorylation balance are at great importance. However, the role of phosphatase enzymes is less investigated than kinase enzymes. PP2A is a member of serine/threonine protein phosphatase that its imbalance has been reported in neurodegenerative diseases. Therefore, we reviewed the superfamily of phosphatases and more specifically PP2A, its regulation, and physiological functions participate in CNS. Thereafter, we discussed the latest findings about PP2A dysregulation in Alzheimer and Parkinson diseases and possible interplay between this phosphatase and insulin signaling pathways. Finally, activating/inhibitory modulators for PP2A activity as well as experimental methods for PP2A study have been reviewed.
Collapse
Affiliation(s)
- Pegah Javadpour
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Tang S, Lu C, Mo L, Wang X, Liang Z, Qin F, Liu Y, Liu Y, Huang H, Huang Y, Cai H, Xiao D, Guo S, Ouyang Y, Sun B, Li X. Hydrogen peroxide redistributes the localization of protein phosphatase methylesterase 1. Life Sci 2018; 213:166-173. [DOI: 10.1016/j.lfs.2018.10.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/04/2018] [Accepted: 10/15/2018] [Indexed: 10/28/2022]
|
21
|
Tang S, Qin F, Wang X, Liang Z, Cai H, Mo L, Huang Y, Liang B, Wei X, Ao Q, Xu Y, Liu Y, Xiao D, Guo S, Lu C, Li X. H 2 O 2 induces PP2A demethylation to downregulate mTORC1 signaling in HEK293 cells. Cell Biol Int 2018; 42:1182-1191. [PMID: 29752834 DOI: 10.1002/cbin.10987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/05/2018] [Indexed: 12/14/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a Ser/Thr protein kinase that functions as an ATP and amino acid sensor to govern cell growth and proliferation by mediating mitogen- and nutrient-dependent signal transduction. Protein phosphatase 2A (PP2A), a ubiquitously expressed serine/threonine phosphatase, negatively regulates mTOR signaling. Methylation of PP2A is catalyzed by leucine carboxyl methyltransferase-1 (LCMT1) and reversed by protein phosphatase methylesterase 1 (PME-1), which regulates PP2A activity and substrate specificity. However, whether PP2A methylation is related to mTOR signaling is still unknown. In this study, we examined the effect of PP2A methylation on mTOR signaling in HEK293 cells under oxidative stress. Our results show that oxidative stress induces PP2A demethylation and inhibits the mTORC1 signaling pathway. Next, we examined two strategies to block PP2A demethylation under oxidative stress. One strategy was to prevent PP2A demethylation using a PME-1 inhibitor; the other strategy was to activate PP2A methylation via overexpression of LCMT1. The results show that both the PME-1 inhibitor and LCMT1 overexpression prevent the mTORC1 signaling suppression induced by oxidative stress. Additionally, LCMT1 overexpression rescued cell viability and the mitochondrial membrane potential decrease in response to oxidative stress. These results demonstrate that H2 O2 induces PP2A demethylation to downregulate mTORC1 signaling. These findings provide a novel mechanism for the regulation of PP2A demethylation and mTORC1 signaling under oxidative stress.
Collapse
Affiliation(s)
- Shen Tang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Fu Qin
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xinhang Wang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ziwei Liang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Haiqing Cai
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Laiming Mo
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yue Huang
- School of Medicine, University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Boyin Liang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xuejing Wei
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qingqing Ao
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yilu Xu
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuyang Liu
- Hunan Provincial Center for Disease Control and Prevention, Changsha, Hunan, 410005, China
| | - Deqiang Xiao
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Songchao Guo
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Cailing Lu
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiyi Li
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, 530021, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
22
|
Santos MS, Nortcliffe A, Lewis W, Bradshaw TD, Moody CJ. Synthesis of Highly Substituted 1,2-Diazetidin-3-ones, Small-Ring Scaffolds for Drug Discovery. Chemistry 2018; 24:8325-8330. [DOI: 10.1002/chem.201801309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Marilia S. Santos
- School of Chemistry; University of Nottingham; University Park Nottingham NG7 2RD United Kingdom
| | - Andrew Nortcliffe
- School of Chemistry; University of Nottingham; University Park Nottingham NG7 2RD United Kingdom
| | - William Lewis
- School of Chemistry; University of Nottingham; University Park Nottingham NG7 2RD United Kingdom
| | - Tracey D. Bradshaw
- School of Pharmacy, Centre for Biomolecular Science; University of Nottingham; University Park Nottingham NG7 2RD United Kingdom
| | - Christopher J. Moody
- School of Chemistry; University of Nottingham; University Park Nottingham NG7 2RD United Kingdom
| |
Collapse
|
23
|
Gerry CJ, Schreiber SL. Chemical probes and drug leads from advances in synthetic planning and methodology. Nat Rev Drug Discov 2018; 17:333-352. [PMID: 29651105 PMCID: PMC6707071 DOI: 10.1038/nrd.2018.53] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Screening of small-molecule libraries is a productive method for identifying both chemical probes of disease-related targets and potential starting points for drug discovery. In this article, we focus on strategies such as diversity-oriented synthesis that aim to explore novel areas of chemical space efficiently by populating small-molecule libraries with compounds containing structural features that are typically under-represented in commercially available screening collections. Drawing from more than a decade's worth of examples, we highlight how the design and synthesis of such libraries have been enabled by modern synthetic chemistry, and we illustrate the impact of the resultant chemical probes and drug leads in a wide range of diseases.
Collapse
Affiliation(s)
- Christopher J Gerry
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- The Broad Institute of Harvard & MIT, Cambridge, MA, USA
| | - Stuart L Schreiber
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- The Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| |
Collapse
|
24
|
Wang S, Tian Y, Wang M, Wang M, Sun GB, Sun XB. Advanced Activity-Based Protein Profiling Application Strategies for Drug Development. Front Pharmacol 2018; 9:353. [PMID: 29686618 PMCID: PMC5900428 DOI: 10.3389/fphar.2018.00353] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/27/2018] [Indexed: 12/28/2022] Open
Abstract
Drug targets and modes of action remain two of the biggest challenges in drug development. To address these problems, chemical proteomic approaches have been introduced to profile targets in complex proteomes. Activity-based protein profiling (ABPP) is one of a growing number chemical proteomic approaches that uses small-molecule chemical probes to understand the interaction mechanisms between compounds and targets. ABPP can be used to identify the protein targets of small molecules and even the active sites of target proteins. This review focuses on the overall workflow of the ABPP technology and on additional advanced strategies for target identification and/or drug discovery. Herein, we mainly describe the design strategies for small-molecule probes and discuss the ways in which these probes can be used to identify targets and even validate the interactions of small molecules with targets. In addition, we discuss some basic strategies that have been developed to date, such as click chemistry-ABPP, competitive strategies and, recently, more advanced strategies, including isoTOP-ABPP, fluoPol-ABPP, and qNIRF-ABPP. The isoTOP-ABPP strategy has been coupled with quantitative proteomics to identify the active sites of proteins and explore whole proteomes with specific amino acid profiling. FluoPol-ABPP combined with HTS can be used to discover new compounds for some substrate-free enzymes. The qNIRF-ABPP strategy has a number of applications for in vivo imaging. In this review, we will further discuss the applications of these advanced strategies.
Collapse
Affiliation(s)
- Shan Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Tian
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Wang
- Life and Environmental Science Research Center, Harbin University of Commerce, Harbin, China
| | - Gui-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Target Identification of Bioactive Covalently Acting Natural Products. Curr Top Microbiol Immunol 2018; 420:351-374. [PMID: 30105423 DOI: 10.1007/82_2018_121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There are countless natural products that have been isolated from microbes, plants, and other living organisms that have been shown to possess therapeutic activities such as antimicrobial, anticancer, or anti-inflammatory effects. However, developing these bioactive natural products into drugs has remained challenging in part because of their difficulty in isolation, synthesis, mechanistic understanding, and off-target effects. Among the large pool of bioactive natural products lies classes of compounds that contain potential reactive electrophilic centers that can covalently react with nucleophilic amino acid hotspots on proteins and other biological molecules to modulate their biological action. Covalently acting natural products are more amenable to rapid target identification and mapping of specific druggable hotspots within proteins using activity-based protein profiling (ABPP)-based chemoproteomic strategies. In addition, the granular biochemical insights afforded by knowing specific sites of protein modifications of covalently acting natural products enable the pharmacological interrogation of these sites with more synthetically tractable covalently acting small molecules whose structures are more easily tuned. Both discovering binding pockets and targets hit by natural products and exploiting druggable modalities targeted by natural products with simpler molecules may overcome some of the challenges faced with translating natural products into drugs.
Collapse
|
26
|
Therapeutic targeting of PP2A. Int J Biochem Cell Biol 2017; 96:182-193. [PMID: 29107183 DOI: 10.1016/j.biocel.2017.10.008] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022]
Abstract
Protein phosphatase 2A (PP2A) is a major serine/threonine phosphatase that regulates many cellular processes. Given the central role of PP2A in regulating diverse biological functions and its dysregulation in many diseases, including cancer, PP2A directed therapeutics have become of great interest. The main approaches leveraged thus far can be categorized as follows: 1) inhibiting endogenous inhibitors of PP2A, 2) targeted disruption of post translational modifications on PP2A subunits, or 3) direct targeting of PP2A. Additional insight into the structural, molecular, and biological framework driving the efficacy of these therapeutic strategies will provide a foundation for the refinement and development of novel and clinically tractable PP2A targeted therapies.
Collapse
|
27
|
Chen X, Wong YK, Wang J, Zhang J, Lee YM, Shen HM, Lin Q, Hua ZC. Target identification with quantitative activity based protein profiling (ABPP). Proteomics 2016; 17. [PMID: 27723264 DOI: 10.1002/pmic.201600212] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/14/2016] [Accepted: 10/06/2016] [Indexed: 12/20/2022]
Abstract
As many small bioactive molecules fulfill their functions through interacting with protein targets, the identification of such targets is crucial in understanding their mechanisms of action (MOA) and side effects. With technological advancements in target identification, it has become possible to accurately and comprehensively study the MOA and side effects of small molecules. While small molecules with therapeutic potential were derived solely from nature in the past, the remodeling and synthesis of such molecules have now been made possible. Presently, while some small molecules have seen successful application as drugs, the majority remain undeveloped, requiring further understanding of their MOA and side effects to fully tap into their potential. Given the typical promiscuity of many small molecules and the complexity of the cellular proteome, a high-flux and high-accuracy method is necessary. While affinity chromatography approaches combined with MS have had successes in target identification, limitations associated with nonspecific results remain. To overcome these complications, quantitative chemical proteomics approaches have been developed including metabolic labeling, chemical labeling, and label-free methods. These new approaches are adopted in conjunction with activity-based protein profiling (ABPP), allowing for a rapid process and accurate results. This review will briefly introduce the principles involved in ABPP, then summarize current advances in quantitative chemical proteomics approaches as well as illustrate with examples how ABPP coupled with quantitative chemical proteomics has been used to detect the targets of drugs and other bioactive small molecules including natural products.
Collapse
Affiliation(s)
- Xiao Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, P. R., China
| | - Yin Kwan Wong
- Department of Biological Sciences, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jigang Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, P. R., China.,Department of Biological Sciences, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research & Technology (SMART), Singapore
| | - Jianbin Zhang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, P. R., China
| | - Yew-Mun Lee
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, P. R., China
| |
Collapse
|
28
|
Regulation of protein phosphatase 2A (PP2A) tumor suppressor function by PME-1. Biochem Soc Trans 2016; 44:1683-1693. [DOI: 10.1042/bst20160161] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/06/2016] [Accepted: 09/09/2016] [Indexed: 02/06/2023]
Abstract
Protein phosphatase 2A (PP2A) plays a major role in maintaining cellular signaling homeostasis by dephosphorylation of a variety of signaling proteins and acts as a tumor suppressor. Protein phosphatase methylesterase-1 (PME-1) negatively regulates PP2A activity by highly complex mechanisms that are reviewed here. Importantly, recent studies have shown that PME-1 promotes oncogenic MAPK/ERK and AKT pathway activities in various cancer types. In human glioma, high PME-1 expression correlates with tumor progression and kinase inhibitor resistance. We discuss the emerging cancer-associated function of PME-1 and its potential clinical relevance.
Collapse
|
29
|
Kaur A, Denisova OV, Qiao X, Jumppanen M, Peuhu E, Ahmed SU, Raheem O, Haapasalo H, Eriksson J, Chalmers AJ, Laakkonen P, Westermarck J. PP2A Inhibitor PME-1 Drives Kinase Inhibitor Resistance in Glioma Cells. Cancer Res 2016; 76:7001-7011. [DOI: 10.1158/0008-5472.can-16-1134] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/16/2016] [Accepted: 09/08/2016] [Indexed: 11/16/2022]
|
30
|
Ahn K, Boehm M, Brown MF, Calloway J, Che Y, Chen J, Fennell KF, Geoghegan KF, Gilbert AM, Gutierrez JA, Kalgutkar AS, Lanba A, Limberakis C, Magee TV, O’Doherty I, Oliver R, Pabst B, Pandit J, Parris K, Pfefferkorn JA, Rolph TP, Patel R, Schuff B, Shanmugasundaram V, Starr JT, Varghese AH, Vera NB, Vernochet C, Yan J. Discovery of a Selective Covalent Inhibitor of Lysophospholipase-like 1 (LYPLAL1) as a Tool to Evaluate the Role of this Serine Hydrolase in Metabolism. ACS Chem Biol 2016; 11:2529-40. [PMID: 27391855 DOI: 10.1021/acschembio.6b00266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Lysophospholipase-like 1 (LYPLAL1) is an uncharacterized metabolic serine hydrolase. Human genome-wide association studies link variants of the gene encoding this enzyme to fat distribution, waist-to-hip ratio, and nonalcoholic fatty liver disease. We describe the discovery of potent and selective covalent small-molecule inhibitors of LYPLAL1 and their use to investigate its role in hepatic metabolism. In hepatocytes, selective inhibition of LYPLAL1 increased glucose production supporting the inference that LYPLAL1 is a significant actor in hepatic metabolism. The results provide an example of how a selective chemical tool can contribute to evaluating a hypothetical target for therapeutic intervention, even in the absence of complete biochemical characterization.
Collapse
Affiliation(s)
- Kay Ahn
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Markus Boehm
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew F. Brown
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jessica Calloway
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ye Che
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jinshan Chen
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kimberly F. Fennell
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kieran F. Geoghegan
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Adam M. Gilbert
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jemy A. Gutierrez
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amit S. Kalgutkar
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Adhiraj Lanba
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Chris Limberakis
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Thomas V. Magee
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Inish O’Doherty
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Robert Oliver
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Brandon Pabst
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jayvardhan Pandit
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin Parris
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jeffrey A. Pfefferkorn
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Timothy P. Rolph
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Rushi Patel
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Brandon Schuff
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Veerabahu Shanmugasundaram
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jeremy T. Starr
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Alison H. Varghese
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nicholas B. Vera
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Cecile Vernochet
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jiangli Yan
- Cardiovascular, Metabolic, and Endocrine Diseases (CVMED) Research
Unit, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
- Worldwide Medicinal Chemistry and §Pharmacokinetics, Dynamics, & Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
31
|
Roberts AM, Ward CC, Nomura DK. Activity-based protein profiling for mapping and pharmacologically interrogating proteome-wide ligandable hotspots. Curr Opin Biotechnol 2016; 43:25-33. [PMID: 27568596 DOI: 10.1016/j.copbio.2016.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/04/2016] [Accepted: 08/11/2016] [Indexed: 11/19/2022]
Abstract
Despite the completion of human genome sequencing efforts nearly 15 years ago that brought with it the promise of genome-based discoveries that would cure human diseases, most protein targets that control human diseases have remained largely untranslated, in-part because they represent difficult protein targets to drug. In addition, many of these protein targets lack screening assays or accessible binding pockets, making the development of small-molecule modulators very challenging. Here, we discuss modern methods for activity-based protein profiling-based chemoproteomic strategies to map 'ligandable' hotspots in proteomes using activity and reactivity-based chemical probes to allow for pharmacological interrogation of these previously difficult targets. We will showcase several recent examples of how these technologies have been used to develop highly selective small-molecule inhibitors against disease-related protein targets.
Collapse
Affiliation(s)
- Allison M Roberts
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, United States
| | - Carl C Ward
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, United States
| | - Daniel K Nomura
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, United States.
| |
Collapse
|
32
|
Grech G, Baldacchino S, Saliba C, Grixti MP, Gauci R, Petroni V, Fenech AG, Scerri C. Deregulation of the protein phosphatase 2A, PP2A in cancer: complexity and therapeutic options. Tumour Biol 2016; 37:11691-11700. [PMID: 27444275 DOI: 10.1007/s13277-016-5145-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/11/2016] [Indexed: 01/26/2023] Open
Abstract
The complexity of the phosphatase, PP2A, is being unravelled and current research is increasingly providing information on the association of deregulated PP2A function with cancer initiation and progression. It has been reported that decreased activity of PP2A is a recurrent observation in many types of cancer, including colorectal and breast cancer (Baldacchino et al. EPMA J. 5:3, 2014; Cristobal et al. Mol Cancer Ther. 13:938-947, 2014). Since deregulation of PP2A and its regulatory subunits is a common event in cancer, PP2A is a potential target for therapy (Baldacchino et al. EPMA J. 5:3, 2014). In this review, the structural components of the PP2A complex are described, giving an in depth overview of the diversity of regulatory subunits. Regulation of the active PP2A trimeric complex, through phosphorylation and methylation, can be targeted using known compounds, to reactivate the complex. The endogenous inhibitors of the PP2A complex are highly deregulated in cancer, representing cases that are eligible to PP2A-activating drugs. Pharmacological opportunities to target low PP2A activity are available and preclinical data support the efficacy of these drugs, but clinical trials are lacking. We highlight the importance of PP2A deregulation in cancer and the current trends in targeting the phosphatase.
Collapse
Affiliation(s)
- Godfrey Grech
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta.
| | - Shawn Baldacchino
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta
| | - Christian Saliba
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Maria Pia Grixti
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta
| | - Robert Gauci
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta
| | - Vanessa Petroni
- Department of Anatomy, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Anthony G Fenech
- Department of Clinical Pharmacology & Therapeutics, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Christian Scerri
- Department of Physiology and Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, Malta.,Molecular Genetics Clinic, Mater Dei Hospital, Msida, Malta
| |
Collapse
|
33
|
Hall MD, Yasgar A, Peryea T, Braisted JC, Jadhav A, Simeonov A, Coussens NP. Fluorescence polarization assays in high-throughput screening and drug discovery: a review. Methods Appl Fluoresc 2016; 4:022001. [PMID: 28809163 DOI: 10.1088/2050-6120/4/2/022001] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The sensitivity of fluorescence polarization (FP) and fluorescence anisotropy (FA) to molecular weight changes has enabled the interrogation of diverse biological mechanisms, ranging from molecular interactions to enzymatic activity. Assays based on FP/FA technology have been widely utilized in high-throughput screening (HTS) and drug discovery due to the homogenous format, robust performance and relative insensitivity to some types of interferences, such as inner filter effects. Advancements in assay design, fluorescent probes, and technology have enabled the application of FP assays to increasingly complex biological processes. Herein we discuss different types of FP/FA assays developed for HTS, with examples to emphasize the diversity of applicable targets. Furthermore, trends in target and fluorophore selection, as well as assay type and format, are examined using annotated HTS assays within the PubChem database. Finally, practical considerations for the successful development and implementation of FP/FA assays for HTS are provided based on experience at our center and examples from the literature, including strategies for flagging interference compounds among a list of hits.
Collapse
Affiliation(s)
- Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Sarma BK, Liu X, Kodadek T. Identification of selective covalent inhibitors of platelet activating factor acetylhydrolase 1B2 from the screening of an oxadiazolone-capped peptoid-azapeptoid hybrid library. Bioorg Med Chem 2016; 24:3953-3963. [PMID: 27160052 DOI: 10.1016/j.bmc.2016.04.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/21/2022]
Abstract
A potent and selective inhibitor of platelet-activating factor acetylhydrolase 1B2 (PAFAH1B2) is described. The compound was derived by improvement of a modest affinity primary hit isolated from the screening of a bead-displayed peptoid-azapeptoid hybrid library tethered to an oxadiazolone 'warhead'. The oxadiazolone moiety of the inhibitors was found to react covalently with the active site serine residue of PAFAH1B2. This screening strategy may be useful for the identification of many selective, covalent inhibitors of serine hydrolases.
Collapse
Affiliation(s)
- Bani Kanta Sarma
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh-201314, India
| | - Xiaodan Liu
- Departments of Chemistry, Scripps Research Institute, Scripps Florida, 130 Scripps Way, #3A2, Jupiter, Fl 33458, USA
| | - Thomas Kodadek
- Departments of Chemistry, Scripps Research Institute, Scripps Florida, 130 Scripps Way, #3A2, Jupiter, Fl 33458, USA
| |
Collapse
|
35
|
Relationships of human α/β hydrolase fold proteins and other organophosphate-interacting proteins. Chem Biol Interact 2016; 259:343-351. [PMID: 27109753 DOI: 10.1016/j.cbi.2016.04.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/29/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
Organophosphates (OPs) are either found in nature or synthetized for use as pesticides, flame retardants, neurotoxic warfare agents or drugs (cholinergic enhancers in Alzheimer's disease and myasthenia gravis, or inhibitors of lipases in metabolic diseases). Because of the central role of acetylcholinesterase cholinergic neurotransmission in humans, one of the main purposes for using OPs is inactivation of the enzyme by phosphorylation of the nucleophilic serine residue in the active center. However, hundreds of serine hydrolases are expressed in the human proteome, and many of them are potential targets for OP adduction. In this review, we first situate the α/β hydrolase fold proteins among the distinctively folded proteins known to interact with OPs, in particular the different lipases, peptidases, and enzymes hydrolyzing OPs. Second, we compile the human α/β hydrolases and review those that have been experimentally shown to interact with OPs. Among the 120 human α/β hydrolase fold proteins, 102 have a serine in the consensus GXSXG pentapeptide compatible with an active site, 6 have an aspartate or a cysteine as the active site nucleophile residue, and 12 evidently lack an active site. 76 of the 120 have been experimentally shown to bind an OP.
Collapse
|
36
|
Xia X, Gholkar A, Senese S, Torres JZ. A LCMT1-PME-1 methylation equilibrium controls mitotic spindle size. Cell Cycle 2016; 14:1938-47. [PMID: 25839665 PMCID: PMC4614068 DOI: 10.1080/15384101.2015.1026487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Leucine carboxyl methyltransferase-1 (LCMT1) and protein phosphatase methylesterase-1 (PME-1) are essential enzymes that regulate the methylation of the protein phosphatase 2A catalytic subunit (PP2AC). LCMT1 and PME-1 have been linked to the regulation of cell growth and proliferation, but the underlying mechanisms have remained elusive. We show here an important role for an LCMT1-PME-1 methylation equilibrium in controlling mitotic spindle size. Depletion of LCMT1 or overexpression of PME-1 led to long spindles. In contrast, depletion of PME-1, pharmacological inhibition of PME-1 or overexpression of LCMT1 led to short spindles. Furthermore, perturbation of the LCMT1-PME-1 methylation equilibrium led to mitotic arrest, spindle assembly checkpoint activation, defective cell divisions, induction of apoptosis and reduced cell viability. Thus, we propose that the LCMT1-PME-1 methylation equilibrium is critical for regulating mitotic spindle size and thereby proper cell division.
Collapse
Affiliation(s)
- Xiaoyu Xia
- a Department of Chemistry and Biochemistry; University of California ; Los Angeles , CA , USA
| | | | | | | |
Collapse
|
37
|
Pusey M, Bail S, Xu Y, Buiakova O, Nestor M, Yang JJ, Rice LM. Inhibition of protein methylesterase 1 decreased cancerous phenotypes in endometrial adenocarcinoma cell lines and xenograft tumor models. Tumour Biol 2016; 37:11835-11842. [PMID: 27048286 DOI: 10.1007/s13277-016-5036-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/28/2016] [Indexed: 12/15/2022] Open
Abstract
Protein methylesterase 1 (PME-1) promotes cancerous phenotypes through the demethylation and inactivation of protein phosphatase 2A. We previously demonstrated that PME-1 overexpression promotes Akt, ERK, and may promote Wnt signaling and increases tumor burden in a xenograft model of endometrial cancer. Here, we show that covalent PME-1 inhibitors decrease cell proliferation and invasive growth in vitro but have no effect in vivo at the concentrations tested; however, depletion of PME-1 with shRNA in an endometrial cancer xenograft model significantly reduced tumor growth. Thus, discovery of more potent PME-1 inhibitors may be beneficial for the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Michelle Pusey
- Oncoveda, Cancer Signaling and Cell Cycle Team, Medical Diagnostic Laboratories, LLC, 1000 Waterview Drive, Room 345, Hamilton, NJ, 08691, USA
| | - Sophie Bail
- Oncoveda, Cancer Signaling and Cell Cycle Team, Medical Diagnostic Laboratories, LLC, 1000 Waterview Drive, Room 345, Hamilton, NJ, 08691, USA
| | - Yan Xu
- Invivotek, LLC, 16 Black Forest Road, Hamilton, NJ, 08691, USA
| | - Olesia Buiakova
- Invivotek, LLC, 16 Black Forest Road, Hamilton, NJ, 08691, USA
| | - Mariya Nestor
- Pathology Department, Members of Genesis Biotechnology Group, LLC, Medical Diagnostic Laboratories LLC, 2439 Kuser Road, Hamilton, NJ, 08690, USA
| | - Jing-Jing Yang
- Pathology Department, Members of Genesis Biotechnology Group, LLC, Medical Diagnostic Laboratories LLC, 2439 Kuser Road, Hamilton, NJ, 08690, USA
| | - Lyndi M Rice
- Oncoveda, Cancer Signaling and Cell Cycle Team, Medical Diagnostic Laboratories, LLC, 1000 Waterview Drive, Room 345, Hamilton, NJ, 08691, USA.
| |
Collapse
|
38
|
Sangodkar J, Farrington C, McClinch K, Galsky MD, Kastrinsky DB, Narla G. All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J 2016; 283:1004-24. [PMID: 26507691 PMCID: PMC4803620 DOI: 10.1111/febs.13573] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/29/2015] [Accepted: 10/21/2015] [Indexed: 12/22/2022]
Abstract
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in the regulation of many cellular processes. A confirmed tumor suppressor protein, PP2A is genetically altered or functionally inactivated in many cancers highlighting a need for its therapeutic reactivation. In this review we discuss recent literature on PP2A: the elucidation of its structure and the functions of its subunits, and the identification of molecular lesions and post-translational modifications leading to its dysregulation in cancer. A final section will discuss the proteins and small molecules that modulate PP2A and how these might be used to target dysregulated forms of PP2A to treat cancers and other diseases.
Collapse
Affiliation(s)
- Jaya Sangodkar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Caroline Farrington
- Department of Medicine and Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kimberly McClinch
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D. Galsky
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David B. Kastrinsky
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goutham Narla
- Department of Medicine and Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
39
|
Li Y, Du S, Du Z, Chen C. A theoretical study of DABCO and PPh3 catalyzed annulations of allenoates with azodicarboxylate. RSC Adv 2016. [DOI: 10.1039/c6ra19308k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Previous experiments have shown that DABCO-catalyzed annulation of 2,3-butadienoate and diethylazodicarboxylate leads to 1,2-diazetidine (reaction (1)), whereas PPh3-catalyzed 2-benzyl-2,3-butadienoate and diethylazodicarboxylate gives pyrazoline (reaction (2)).
Collapse
Affiliation(s)
- Yan Li
- School of Petroleum and Chemical Engineering
- Dalian University of Technology
- Panjin
- China
| | - Shiwen Du
- School of Petroleum and Chemical Engineering
- Dalian University of Technology
- Panjin
- China
| | - Zheng Du
- National Supercomputing Center in Shenzhen (Shenzhen Cloud Computing Center)
- Shenzhen
- China
| | - Congmei Chen
- National Supercomputing Center in Shenzhen (Shenzhen Cloud Computing Center)
- Shenzhen
- China
| |
Collapse
|
40
|
Chen B, Ge SS, Zhao YC, Chen C, Yang S. Activity-based protein profiling: an efficient approach to study serine hydrolases and their inhibitors in mammals and microbes. RSC Adv 2016. [DOI: 10.1039/c6ra20006k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
This review focuses on the identification of serine hydrolases and their inhibitors in mammals and microbes with activity-based protein profiling (ABPP).
Collapse
Affiliation(s)
- Biao Chen
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering
- Key Laboratory of Green Pesticide and Agricultural Bioengineering
- Ministry of Education
- Center for R&D of Fine Chemicals of Guizhou University
- Guiyang
| | - Sha-Sha Ge
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering
- Key Laboratory of Green Pesticide and Agricultural Bioengineering
- Ministry of Education
- Center for R&D of Fine Chemicals of Guizhou University
- Guiyang
| | - Yuan-Chao Zhao
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering
- Key Laboratory of Green Pesticide and Agricultural Bioengineering
- Ministry of Education
- Center for R&D of Fine Chemicals of Guizhou University
- Guiyang
| | - Chong Chen
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering
- Key Laboratory of Green Pesticide and Agricultural Bioengineering
- Ministry of Education
- Center for R&D of Fine Chemicals of Guizhou University
- Guiyang
| | - Song Yang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering
- Key Laboratory of Green Pesticide and Agricultural Bioengineering
- Ministry of Education
- Center for R&D of Fine Chemicals of Guizhou University
- Guiyang
| |
Collapse
|
41
|
Yabe R, Miura A, Usui T, Mudrak I, Ogris E, Ohama T, Sato K. Protein Phosphatase Methyl-Esterase PME-1 Protects Protein Phosphatase 2A from Ubiquitin/Proteasome Degradation. PLoS One 2015; 10:e0145226. [PMID: 26678046 PMCID: PMC4683032 DOI: 10.1371/journal.pone.0145226] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/01/2015] [Indexed: 11/30/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a conserved essential enzyme that is implicated as a tumor suppressor based on its central role in phosphorylation-dependent signaling pathways. Protein phosphatase methyl esterase (PME-1) catalyzes specifically the demethylation of the C-terminal Leu309 residue of PP2A catalytic subunit (PP2Ac). It has been shown that PME-1 affects the activity of PP2A by demethylating PP2Ac, but also by directly binding to the phosphatase active site, suggesting loss of PME-1 in cells would enhance PP2A activity. However, here we show that PME-1 knockout mouse embryonic fibroblasts (MEFs) exhibit lower PP2A activity than wild type MEFs. Loss of PME-1 enhanced poly-ubiquitination of PP2Ac and shortened the half-life of PP2Ac protein resulting in reduced PP2Ac levels. Chemical inhibition of PME-1 and rescue experiments with wild type and mutated PME-1 revealed methyl-esterase activity was necessary to maintain PP2Ac protein levels. Our data demonstrate that PME-1 methyl-esterase activity protects PP2Ac from ubiquitin/proteasome degradation.
Collapse
Affiliation(s)
- Ryotaro Yabe
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753–8515, Japan
| | - Akane Miura
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753–8515, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Toxicology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753–8515, Japan
| | - Ingrid Mudrak
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, 1030, Austria
| | - Egon Ogris
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, 1030, Austria
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753–8515, Japan
- * E-mail:
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753–8515, Japan
| |
Collapse
|
42
|
Mapping proteome-wide interactions of reactive chemicals using chemoproteomic platforms. Curr Opin Chem Biol 2015; 30:68-76. [PMID: 26647369 DOI: 10.1016/j.cbpa.2015.11.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 11/09/2015] [Indexed: 12/11/2022]
Abstract
A large number of pharmaceuticals, endogenous metabolites, and environmental chemicals act through covalent mechanisms with protein targets. Yet, their specific interactions with the proteome still remain poorly defined for most of these reactive chemicals. Deciphering direct protein targets of reactive small-molecules is critical in understanding their biological action, off-target effects, potential toxicological liabilities, and development of safer and more selective agents. Chemoproteomic technologies have arisen as a powerful strategy that enable the assessment of proteome-wide interactions of these irreversible agents directly in complex biological systems. We review here several chemoproteomic strategies that have facilitated our understanding of specific protein interactions of irreversibly-acting pharmaceuticals, endogenous metabolites, and environmental electrophiles to reveal novel pharmacological, biological, and toxicological mechanisms.
Collapse
|
43
|
Dudiki T, Kadunganattil S, Ferrara JK, Kline DW, Vijayaraghavan S. Changes in Carboxy Methylation and Tyrosine Phosphorylation of Protein Phosphatase PP2A Are Associated with Epididymal Sperm Maturation and Motility. PLoS One 2015; 10:e0141961. [PMID: 26569399 PMCID: PMC4646675 DOI: 10.1371/journal.pone.0141961] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/15/2015] [Indexed: 01/21/2023] Open
Abstract
Mammalian sperm contain the serine/threonine phosphatases PP1γ2 and PP2A. The role of sperm PP1γ2 is relatively well studied. Here we confirm the presence of PP2A in sperm and show that it undergoes marked changes in methylation (leucine 309), tyrosine phosphorylation (tyrosine 307) and catalytic activity during epididymal sperm maturation. Spermatozoa isolated from proximal caput, distal caput and caudal regions of the epididymis contain equal immuno-reactive amounts of PP2A. Using demethyl sensitive antibodies we show that PP2A is methylated at its carboxy terminus in sperm from the distal caput and caudal regions but not in sperm from the proximal caput region of the epididymis. The methylation status of PP2A was confirmed by isolation of PP2A with microcystin agarose followed by alkali treatment, which causes hydrolysis of protein carboxy methyl esters. Tyrosine phosphorylation of sperm PP2A varied inversely with methylation. That is, PP2A was tyrosine phosphorylated when it was demethylated but not when methylated. PP2A demethylation and its reciprocal tyrosine phosphorylation were also affected by treatment of sperm with L-homocysteine and adenosine, which are known to elevate intracellular S-adenosylhomocysteine, a feedback inhibitor of methyltransferases. Catalytic activity of PP2A declined during epididymal sperm maturation. Inhibition of PP2A by okadaic acid or by incubation of caudal epididymal spermatozoa with L-homocysteine and adenosine resulted in increase of sperm motility parameters including percent motility, velocity, and lateral head amplitude. Demethylation or pharmacological inhibition of PP2A also leads to an increase in phosphorylation of glycogen synthase kinase-3 (GSK3). Our results show for the first time that changes in PP2A activity due to methylation and tyrosine phosphorylation occur in sperm and that these changes may play an important role in the regulation of sperm function.
Collapse
Affiliation(s)
- Tejasvi Dudiki
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Suraj Kadunganattil
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - John K. Ferrara
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Douglas W. Kline
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | | |
Collapse
|
44
|
Xu S, Chen J, Shang J, Qing Z, Zhang J, Tang Y. Divergent amine-catalyzed [2+2] annulation of allenoates with azodicarboxylates: facile synthesis of 1,2-diazetidines. Tetrahedron Lett 2015. [DOI: 10.1016/j.tetlet.2015.09.151] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
45
|
Iglesias J, Lamontagne J, Erb H, Gezzar S, Zhao S, Joly E, Truong VL, Skorey K, Crane S, Madiraju SRM, Prentki M. Simplified assays of lipolysis enzymes for drug discovery and specificity assessment of known inhibitors. J Lipid Res 2015; 57:131-41. [PMID: 26423520 DOI: 10.1194/jlr.d058438] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Indexed: 12/25/2022] Open
Abstract
Lipids are used as cellular building blocks and condensed energy stores and also act as signaling molecules. The glycerolipid/ fatty acid cycle, encompassing lipolysis and lipogenesis, generates many lipid signals. Reliable procedures are not available for measuring activities of several lipolytic enzymes for the purposes of drug screening, and this resulted in questionable selectivity of various known lipase inhibitors. We now describe simple assays for lipolytic enzymes, including adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL), sn-1-diacylglycerol lipase (DAGL), monoacylglycerol lipase, α/β-hydrolase domain 6, and carboxylesterase 1 (CES1) using recombinant human and mouse enzymes either in cell extracts or using purified enzymes. We observed that many of the reported inhibitors lack specificity. Thus, Cay10499 (HSL inhibitor) and RHC20867 (DAGL inhibitor) also inhibit other lipases. Marked differences in the inhibitor sensitivities of human ATGL and HSL compared with the corresponding mouse enzymes was noticed. Thus, ATGListatin inhibited mouse ATGL but not human ATGL, and the HSL inhibitors WWL11 and Compound 13f were effective against mouse enzyme but much less potent against human enzyme. Many of these lipase inhibitors also inhibited human CES1. Results describe reliable assays for measuring lipase activities that are amenable for drug screening and also caution about the specificity of the many earlier described lipase inhibitors.
Collapse
Affiliation(s)
- Jose Iglesias
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Julien Lamontagne
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Heidi Erb
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Sari Gezzar
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Shangang Zhao
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Erik Joly
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | | | | | - Sheldon Crane
- NuChem Therapeutics, Montréal, Québec, Canada, H4P 2R2
| | - S R Murthy Madiraju
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Marc Prentki
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| |
Collapse
|
46
|
Kohnz RA, Mulvihill MM, Chang JW, Hsu KL, Sorrentino A, Cravatt BF, Bandyopadhyay S, Goga A, Nomura DK. Activity-Based Protein Profiling of Oncogene-Driven Changes in Metabolism Reveals Broad Dysregulation of PAFAH1B2 and 1B3 in Cancer. ACS Chem Biol 2015; 10:1624-30. [PMID: 25945974 DOI: 10.1021/acschembio.5b00053] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Targeting dysregulated metabolic pathways is a promising therapeutic strategy for eradicating cancer. Understanding how frequently altered oncogenes regulate metabolic enzyme targets would be useful in identifying both broad-spectrum and targeted metabolic therapies for cancer. Here, we used activity-based protein profiling to identify serine hydrolase activities that were consistently upregulated by various human oncogenes. Through this profiling effort, we found oncogenic regulatory mechanisms for several cancer-relevant serine hydrolases and discovered that platelet activating factor acetylhydrolase 1B2 and 1B3 (PAFAH1B2 and PAFAH1B3) activities were consistently upregulated by several oncogenes, alongside previously discovered cancer-relevant hydrolases fatty acid synthase and monoacylglycerol lipase. While we previously showed that PAFAH1B2 and 1B3 were important in breast cancer, our most recent profiling studies have revealed that these enzymes may be dysregulated broadly across many types of cancers. Here, we find that pharmacological blockade of both enzymes impairs cancer pathogenicity across multiple different types of cancer cells, including breast, ovarian, melanoma, and prostate cancer. We also show that pharmacological blockade of PAFAH1B2 and 1B3 causes unique changes in lipid metabolism, including heightened levels of tumor-suppressing lipids. Our results reveal oncogenic regulatory mechanisms of several cancer-relevant serine hydrolases using activity-based protein profiling, and we show that PAFAH1B2 and 1B3 are important in maintaining cancer pathogenicity across a wide spectrum of cancer types.
Collapse
Affiliation(s)
- Rebecca A. Kohnz
- Program in Metabolic Biology, Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, 127 Morgan Hall, Berkeley, California 94720, United States
| | - Melinda M. Mulvihill
- Program in Metabolic Biology, Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, 127 Morgan Hall, Berkeley, California 94720, United States
| | - Jae Won Chang
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Rd. SR107, La Jolla, California 92037, United States
| | - Ku-Lung Hsu
- Department
of Chemistry, University of Virginia, McCormick Road,
P.O. Box 400319, Charlottesville, Virginia 22904-4319, United States
| | | | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Rd. SR107, La Jolla, California 92037, United States
| | | | - Andrei Goga
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, Box 0128, San Francisco, California 94143, United States
| | - Daniel K. Nomura
- Program in Metabolic Biology, Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, 127 Morgan Hall, Berkeley, California 94720, United States
| |
Collapse
|
47
|
Schreiber SL, Kotz JD, Li M, Aubé J, Austin CP, Reed JC, Rosen H, White EL, Sklar LA, Lindsley CW, Alexander BR, Bittker JA, Clemons PA, de Souza A, Foley MA, Palmer M, Shamji AF, Wawer MJ, McManus O, Wu M, Zou B, Yu H, Golden JE, Schoenen FJ, Simeonov A, Jadhav A, Jackson MR, Pinkerton AB, Chung TDY, Griffin PR, Cravatt BF, Hodder PS, Roush WR, Roberts E, Chung DH, Jonsson CB, Noah JW, Severson WE, Ananthan S, Edwards B, Oprea TI, Conn PJ, Hopkins CR, Wood MR, Stauffer SR, Emmitte KA. Advancing Biological Understanding and Therapeutics Discovery with Small-Molecule Probes. Cell 2015; 161:1252-65. [PMID: 26046436 PMCID: PMC4564295 DOI: 10.1016/j.cell.2015.05.023] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Indexed: 02/06/2023]
Abstract
Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.
Collapse
Affiliation(s)
- Stuart L Schreiber
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Joanne D Kotz
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Min Li
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Jeffrey Aubé
- University of Kansas Specialized Chemistry Center, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, 66045, USA
| | - Christopher P Austin
- NIH Chemical Genomics Center, National Institutes of Health, Rockville, MD 20850, USA; National Center for Advancing Translational Sciences, Bethesda, MD 20892, USA
| | - John C Reed
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Hugh Rosen
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - E Lucile White
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Larry A Sklar
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM 87131, USA; Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Craig W Lindsley
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Benjamin R Alexander
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Joshua A Bittker
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Development of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Paul A Clemons
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Andrea de Souza
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michael A Foley
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michelle Palmer
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Alykhan F Shamji
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Mathias J Wawer
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Owen McManus
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Meng Wu
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Beiyan Zou
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Haibo Yu
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Jennifer E Golden
- University of Kansas Specialized Chemistry Center, Lawrence, KS 66045, USA
| | - Frank J Schoenen
- University of Kansas Specialized Chemistry Center, Lawrence, KS 66045, USA
| | - Anton Simeonov
- NIH Chemical Genomics Center, National Institutes of Health, Rockville, MD 20850, USA; National Center for Advancing Translational Sciences, Bethesda, MD 20892, USA
| | - Ajit Jadhav
- NIH Chemical Genomics Center, National Institutes of Health, Rockville, MD 20850, USA; National Center for Advancing Translational Sciences, Bethesda, MD 20892, USA
| | - Michael R Jackson
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Anthony B Pinkerton
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Thomas D Y Chung
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Patrick R Griffin
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Benjamin F Cravatt
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Peter S Hodder
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA
| | - William R Roush
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Chemistry, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Edward Roberts
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA
| | - Dong-Hoon Chung
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Colleen B Jonsson
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - James W Noah
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - William E Severson
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Subramaniam Ananthan
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Bruce Edwards
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM 87131, USA; Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Tudor I Oprea
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM 87131, USA; Department of Internal Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - P Jeffrey Conn
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Corey R Hopkins
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Michael R Wood
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Shaun R Stauffer
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kyle A Emmitte
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
48
|
Medina-Cleghorn D, Nomura DK. Exploring metabolic pathways and regulation through functional chemoproteomic and metabolomic platforms. ACTA ACUST UNITED AC 2015; 21:1171-84. [PMID: 25237861 DOI: 10.1016/j.chembiol.2014.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 07/15/2014] [Accepted: 07/21/2014] [Indexed: 02/07/2023]
Abstract
Genome sequencing efforts have revealed a strikingly large number of uncharacterized genes, including poorly or uncharacterized metabolic enzymes, metabolites, and metabolic networks that operate in normal physiology, and those enzymes and pathways that may be rewired under pathological conditions. Although deciphering the functions of the uncharacterized metabolic genome is a challenging prospect, it also presents an opportunity for identifying novel metabolic nodes that may be important in disease therapy. In this review, we will discuss the chemoproteomic and metabolomic platforms used in identifying, characterizing, and targeting nodal metabolic pathways important in physiology and disease, describing an integrated workflow for functional mapping of metabolic enzymes.
Collapse
Affiliation(s)
- Daniel Medina-Cleghorn
- Program in Metabolic Biology and Molecular Toxicology, Department of Nutritional Sciences and Toxicology, 127 Morgan Hall, Berkeley, CA 94720, USA
| | - Daniel K Nomura
- Program in Metabolic Biology and Molecular Toxicology, Department of Nutritional Sciences and Toxicology, 127 Morgan Hall, Berkeley, CA 94720, USA.
| |
Collapse
|
49
|
Chang JW, Zuhl AM, Speers AE, Niessen S, Brown SJ, Mulvihill MM, Fan YC, Spicer TP, Southern M, Scampavia L, Fernandez-Vega V, Dix MM, Cameron MD, Hodder PS, Rosen H, Nomura DK, Kwon O, Hsu KL, Cravatt BF. Selective inhibitor of platelet-activating factor acetylhydrolases 1b2 and 1b3 that impairs cancer cell survival. ACS Chem Biol 2015; 10:925-32. [PMID: 25602368 DOI: 10.1021/cb500893q] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Platelet-activating factor acetylhydrolases (PAFAHs) 1b2 and 1b3 are poorly characterized serine hydrolases that form a complex with a noncatalytic protein (1b1) to regulate brain development, spermatogenesis, and cancer pathogenesis. Determining physiological substrates and biochemical functions for the PAFAH1b complex would benefit from selective chemical probes that can perturb its activity in living systems. Here, we report a class of tetrahydropyridine reversible inhibitors of PAFAH1b2/3 discovered using a fluorescence polarization-activity-based protein profiling (fluopol-ABPP) screen of the NIH 300,000+ compound library. The most potent of these agents, P11, exhibited IC50 values of ∼40 and 900 nM for PAFAH1b2 and 1b3, respectively. We confirm selective inhibition of PAFAH1b2/3 in cancer cells by P11 using an ABPP protocol adapted for in situ analysis of reversible inhibitors and show that this compound impairs tumor cell survival, supporting a role for PAFAH1b2/3 in cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Melinda M. Mulvihill
- Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, 127 Morgan Hall, Berkeley, California 94720, United States
| | - Yi Chiao Fan
- Department
of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | | | | | | | | | | | | | | | | | - Daniel K. Nomura
- Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, 127 Morgan Hall, Berkeley, California 94720, United States
| | - Ohyun Kwon
- Department
of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | | | | |
Collapse
|
50
|
Adachi S, Cognetta AB, Niphakis MJ, He Z, Zajdlik A, St Denis JD, Scully CCG, Cravatt BF, Yudin AK. Facile synthesis of borofragments and their evaluation in activity-based protein profiling. Chem Commun (Camb) 2015; 51:3608-11. [PMID: 25633248 PMCID: PMC4330092 DOI: 10.1039/c4cc09107h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The discovery of enzyme inhibitors relies on synthetic methods that enable rapid and modular construction of small molecules. Heterocyclic fragments designed to maximize enthalpic interactions with their protein targets represent a particularly desirable class of molecules. Here we describe a reagent that enables straightforward construction of "borofragments", in which a heterocycle is separated from the boron center by two or three rotatable bonds. The stability of these molecules depends on the MIDA group which likely acts as a slow-release element under biological conditions. Borofragments can be used to discover inhibitors of enzymes that use catalytic oxygen nucleophiles. We have employed this method to identify inhibitors of ABHD10 and the predicted carboxypeptidase CPVL. This technique should be applicable to other classes of targets.
Collapse
Affiliation(s)
- Shinya Adachi
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON M5S3H6, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|