1
|
Shaw S, Jiang W, Rush J, Dumont N, Kim J, Singh R, Skepner A, Khodier C, Raffier C, Murphy Z, Yan N, Schluter C, Yu X, Szuchnicki M, Sathappa M, Kahn J, Sperling AS, Wagner F, McKinney DC, Gould AE, Garvie CW, Miller PG. Identification of small molecule inhibitors of PPM1D using an integrated drug discovery platform. iScience 2025; 28:112069. [PMID: 40124519 PMCID: PMC11930361 DOI: 10.1016/j.isci.2025.112069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
PPM1D is a serine/threonine phosphatase recurrently activated in cancer, regulates the DNA damage response (DDR), and suppresses p53. Though PPM1D inhibition impairs tumor growth in cancer models and is the subject of multiple drug discovery efforts, no PPM1D inhibitors with clinical potential have been identified. We screened 600,000 compounds in a displacement assay and generated a hit series with nanomolar activity. We optimized our leads using internally developed assays to interrogate PPM1D, p53, and the DDR and defined important structure-activity relationships. Using an in vivo bioluminescent readout of p53 activation, we compared different DDR and p53 modulators and showed that despite having a distinct chemical structure, our lead compound had comparable in vivo activity to established PPM1D inhibitors. Our approach yielded multiple allosteric inhibitors of PPM1D, deepened our understanding of PPM1D as a drug target, and is highly amenable to studying other modulators of the DDR and p53.
Collapse
Affiliation(s)
- Subrata Shaw
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Wei Jiang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jason Rush
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - John Kim
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ritu Singh
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Carol Khodier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Zachary Murphy
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ni Yan
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cameron Schluter
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Xiao Yu
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Mateusz Szuchnicki
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Adam S. Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Florence Wagner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - David C. McKinney
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexandra E. Gould
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W. Garvie
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Peter G. Miller
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2
|
Jiang W, Shaw S, Rush J, Dumont N, Kim J, Singh R, Skepner A, Khodier C, Raffier C, Yan N, Schluter C, Yu X, Szuchnicki M, Sathappa M, Kahn J, Sperling AS, McKinney DC, Gould AE, Garvie CW, Miller PG. Identification of Small Molecule Inhibitors of PPM1D Using a Novel Drug Discovery Platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595001. [PMID: 38826457 PMCID: PMC11142126 DOI: 10.1101/2024.05.20.595001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Protein phosphatase, Mg2+/Mn2+ dependent 1D (PPM1D), is a serine/threonine phosphatase that is recurrently activated in cancer, regulates the DNA damage response (DDR), and suppresses the activation of p53. Consistent with its oncogenic properties, genetic loss or pharmacologic inhibition of PPM1D impairs tumor growth and sensitizes cancer cells to cytotoxic therapies in a wide range of preclinical models. Given the therapeutic potential of targeting PPM1D specifically and the DDR and p53 pathway more generally, we sought to deepen our biological understanding of PPM1D as a drug target and determine how PPM1D inhibition differs from other therapeutic approaches to activate the DDR. We performed a high throughput screen to identify new allosteric inhibitors of PPM1D, then generated and optimized a suite of enzymatic, cell-based, and in vivo pharmacokinetic and pharmacodynamic assays to drive medicinal chemistry efforts and to further interrogate the biology of PPM1D. Importantly, this drug discovery platform can be readily adapted to broadly study the DDR and p53. We identified compounds distinct from previously reported allosteric inhibitors and showed in vivo on-target activity. Our data suggest that the biological effects of inhibiting PPM1D are distinct from inhibitors of the MDM2-p53 interaction and standard cytotoxic chemotherapies. These differences also highlight the potential therapeutic contexts in which targeting PPM1D would be most valuable. Therefore, our studies have identified a series of new PPM1D inhibitors, generated a suite of in vitro and in vivo assays that can be broadly used to interrogate the DDR, and provided important new insights into PPM1D as a drug target.
Collapse
Affiliation(s)
- Wei Jiang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Subrata Shaw
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jason Rush
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - John Kim
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ritu Singh
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Carol Khodier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ni Yan
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Cameron Schluter
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Xiao Yu
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Mateusz Szuchnicki
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Adam S. Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David C. McKinney
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexandra E. Gould
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W. Garvie
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Peter G. Miller
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
3
|
Robello M, Zheng H, Saha M, George Rosenker KM, Debnath S, Kumar JP, Tagad HD, Mazur SJ, Appella E, Appella DH. Alkyl-substituted N-methylaryl-N'-aryl-4-aminobenzamides: A new series of small molecule inhibitors for Wip1 phosphatase. Eur J Med Chem 2022; 243:114763. [PMID: 36179402 PMCID: PMC9664485 DOI: 10.1016/j.ejmech.2022.114763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 11/03/2022]
Abstract
The wild-type p53 induced phosphatase 1 (Wip1), a member of the serine/threonine-specific PP2C family, is overexpressed in numerous human cancers. Wip1 dephosphorylates p53 as well as several kinases (such as p38 MAPK, ATM, Chk1, and Chk2) in the DNA damage response pathway that are responsible for maintaining genomic stability and preventing oncogenic transformation. As a result, Wip1 is an attractive target for synthetic inhibitors that could be further developed into therapeutics to treat some cancers. In this study, we report a series of alkyl-substituted N-methylaryl-N'-aryl-4-aminobenzamides and their inhibitory activity of the Wip1 phosphatase. A straightforward synthetic route was developed to synthesize the target compounds from commercially available starting materials. Three different portions (R1, R2, R3) of the core scaffold were extensively modified to examine structure-activity relationships. This study revealed interesting trends about a new molecular scaffold to inhibit Wip1.
Collapse
Affiliation(s)
- Marco Robello
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Hongchao Zheng
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Mrinmoy Saha
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Kara M George Rosenker
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Subrata Debnath
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Jay Prakash Kumar
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Harichandra D Tagad
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Ettore Appella
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Daniel H Appella
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States.
| |
Collapse
|
4
|
Clausse V, Fang Y, Tao D, Tagad HD, Sun H, Wang Y, Karavadhi S, Lane K, Shi ZD, Vasalatiy O, LeClair CA, Eells R, Shen M, Patnaik S, Appella E, Coussens NP, Hall MD, Appella DH. Discovery of Novel Small-Molecule Scaffolds for the Inhibition and Activation of WIP1 Phosphatase from a RapidFire Mass Spectrometry High-Throughput Screen. ACS Pharmacol Transl Sci 2022; 5:993-1006. [PMID: 36268125 PMCID: PMC9578142 DOI: 10.1021/acsptsci.2c00147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Indexed: 11/28/2022]
Abstract
Wild-type P53-induced phosphatase 1 (WIP1), also known as PPM1D or PP2Cδ, is a serine/threonine protein phosphatase induced by P53 after genotoxic stress. WIP1 inhibition has been proposed as a therapeutic strategy for P53 wild-type cancers in which it is overexpressed, but this approach would be ineffective in P53-negative cancers. Furthermore, there are several cancers with mutated P53 where WIP1 acts as a tumor suppressor. Therefore, activating WIP1 phosphatase might also be a therapeutic strategy, depending on the P53 status. To date, no specific, potent WIP1 inhibitors with appropriate pharmacokinetic properties have been reported, nor have WIP1-specific activators. Here, we report the discovery of new WIP1 modulators from a high-throughput screen (HTS) using previously described orthogonal biochemical assays suitable for identifying both inhibitors and activators. The primary HTS was performed against a library of 102 277 compounds at a single concentration using a RapidFire mass spectrometry assay. Hits were further evaluated over a range of 11 concentrations with both the RapidFire MS assay and an orthogonal fluorescence-based assay. Further biophysical, biochemical, and cell-based studies of confirmed hits revealed a WIP1 activator and two inhibitors, one competitive and one uncompetitive. These new scaffolds are prime candidates for optimization which might enable inhibitors with improved pharmacokinetics and a first-in-class WIP1 activator.
Collapse
Affiliation(s)
- Victor Clausse
- Synthetic
Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yuhong Fang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Dingyin Tao
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Harichandra D. Tagad
- Laboratory
of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hongmao Sun
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Yuhong Wang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Surendra Karavadhi
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Kelly Lane
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Zhen-Dan Shi
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Olga Vasalatiy
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Christopher A. LeClair
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Rebecca Eells
- Reaction
Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355, United States
| | - Min Shen
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Samarjit Patnaik
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Ettore Appella
- Laboratory
of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nathan P. Coussens
- Molecular
Pharmacology Laboratories, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Matthew D. Hall
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Daniel H. Appella
- Synthetic
Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
5
|
Grimm TM, Herbinger M, Krüger L, Müller S, Mayer TU, Hauck CR. Lockdown, a selective small-molecule inhibitor of the integrin phosphatase PPM1F, blocks cancer cell invasion. Cell Chem Biol 2022; 29:930-946.e9. [PMID: 35443151 DOI: 10.1016/j.chembiol.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 12/18/2022]
Abstract
Phosphatase PPM1F is a regulator of cell adhesion by fine-tuning integrin activity and actin cytoskeleton structures. Elevated expression of this enzyme in human tumors is associated with high invasiveness, enhanced metastasis, and poor prognosis. Thus, PPM1F is a target for pharmacological intervention, yet inhibitors of this enzyme are lacking. Here, we use high-throughput screening to identify Lockdown, a reversible and non-competitive PPM1F inhibitor. Lockdown is selective for PPM1F, because this compound does not inhibit other protein phosphatases in vitro and does not induce additional phenotypes in PPM1F knockout cells. Importantly, Lockdown-treated glioblastoma cells fully re-capitulate the phenotype of PPM1F-deficient cells as assessed by increased phosphorylation of PPM1F substrates and corruption of integrin-dependent cellular processes. Ester modification yields LockdownPro with increased membrane permeability and prodrug-like properties. LockdownPro suppresses tissue invasion by PPM1F-overexpressing human cancer cells, validating PPM1F as a therapeutic target and providing an access point to control tumor cell dissemination.
Collapse
Affiliation(s)
- Tanja M Grimm
- Lehrstuhl Zellbiologie, Department of Biology, University of Konstanz, Maildrop 621, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Marleen Herbinger
- Lehrstuhl Zellbiologie, Department of Biology, University of Konstanz, Maildrop 621, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Lena Krüger
- Department of Chemistry, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Silke Müller
- Lehrstuhl Molekulare Genetik, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Screening Center, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Thomas U Mayer
- Lehrstuhl Molekulare Genetik, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Screening Center, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Department of Biology, University of Konstanz, Maildrop 621, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany.
| |
Collapse
|
6
|
Clonal hematopoiesis: mechanisms driving dominance of stem cell clones. Blood 2021; 136:1590-1598. [PMID: 32746453 DOI: 10.1182/blood.2020006510] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022] Open
Abstract
The discovery of clonal hematopoiesis (CH) in older individuals has changed the way hematologists and stem cell biologists view aging. Somatic mutations accumulate in stem cells over time. While most mutations have no impact, some result in subtle functional differences that ultimately manifest in distinct stem cell behaviors. With a large pool of stem cells and many decades to compete, some of these differences confer advantages under specific contexts. Approximately 20 genes are recurrently found as mutated in CH, indicating they confer some advantage. The impact of these mutations has begun to be analyzed at a molecular level by modeling in cell lines and in mice. Mutations in epigenetic regulators such as DNMT3A and TET2 confer an advantage by enhancing self-renewal of stem and progenitor cells and inhibiting their differentiation. Mutations in other genes involved in the DNA damage response may simply enhance cell survival. Here, we review proposed mechanisms that lead to CH, specifically in the context of stem cell biology, based on our current understanding of the function of some of the CH-associated genes.
Collapse
|
7
|
A conserved allosteric element controls specificity and activity of functionally divergent PP2C phosphatases from Bacillus subtilis. J Biol Chem 2021; 296:100518. [PMID: 33684446 PMCID: PMC8080068 DOI: 10.1016/j.jbc.2021.100518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 01/08/2023] Open
Abstract
Reversible phosphorylation relies on highly regulated kinases and phosphatases that target specific substrates to control diverse cellular processes. Here, we address how protein phosphatase activity is directed to the correct substrates under the correct conditions. The serine/threonine phosphatase SpoIIE from Bacillus subtilis, a member of the widespread protein phosphatase 2C (PP2C) family of phosphatases, is activated by movement of a conserved α-helical element in the phosphatase domain to create the binding site for the metal cofactor. We hypothesized that this conformational switch could provide a general mechanism for control of diverse members of the PP2C family of phosphatases. The B. subtilis phosphatase RsbU responds to different signals, acts on a different substrates, and produces a more graded response than SpoIIE. Using an unbiased genetic screen, we isolated mutants in the α-helical switch region of RsbU that are constitutively active, indicating conservation of the switch mechanism. Using phosphatase activity assays with phosphoprotein substrates, we found that both phosphatases integrate substrate recognition with activating signals to control metal-cofactor binding and substrate dephosphorylation. This integrated control provides a mechanism for PP2C family of phosphatases to produce specific responses by acting on the correct substrates, under the appropriate conditions.
Collapse
|
8
|
Shi L, Tian Q, Feng C, Zhang P, Zhao Y. The biological function and the regulatory roles of wild-type p53-induced phosphatase 1 in immune system. Int Rev Immunol 2020; 39:280-291. [PMID: 32696682 DOI: 10.1080/08830185.2020.1795153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Wild-type p53-induced phosphatase 1 (WIP1) belongs to the protein phosphatase 2C (PP2C) family and is a mammalian serine/threonine specific protein phosphatase to dephosphorylate numerous signaling molecules. Mammalian WIP1 regulates a wide array of targeting molecules and plays key regulatory roles in many cell processes such as DNA damage and repair, cell proliferation, differentiation, apoptosis, and senescence. WIP1 promotes the formation and development of tumors as an oncogene and a negative regulator of p53. It is also involved in the regulation of aging, neurological diseases and immune diseases. Recent studies demonstrated the critical roles of WIP1 in the differentiation and function of immune cells including T cells, neutrophils and macrophages. In the present manuscript, we briefly summarized the expression patterns, biological function and the target molecules and signal pathways of WIP1 and mainly discussed the latest advances on the regulatory effects of WIP1 in the immune system. WIP1 may be a potential target molecule to treat cancers and immune diseases such as allergic asthma.
Collapse
Affiliation(s)
- Lu Shi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qianchuan Tian
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chang Feng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Metal-dependent Ser/Thr protein phosphatase PPM family: Evolution, structures, diseases and inhibitors. Pharmacol Ther 2020; 215:107622. [PMID: 32650009 DOI: 10.1016/j.pharmthera.2020.107622] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Protein phosphatases and kinases control multiple cellular events including proliferation, differentiation, and stress responses through regulating reversible protein phosphorylation, the most important post-translational modification. Members of metal-dependent protein phosphatase (PPM) family, also known as PP2C phosphatases, are Ser/Thr phosphatases that bind manganese/magnesium ions (Mn2+/Mg2+) in their active center and function as single subunit enzymes. In mammals, there are 20 isoforms of PPM phosphatases: PPM1A, PPM1B, PPM1D, PPM1E, PPM1F, PPM1G, PPM1H, PPM1J, PPM1K, PPM1L, PPM1M, PPM1N, ILKAP, PDP1, PDP2, PHLPP1, PHLPP2, PP2D1, PPTC7, and TAB1, whereas there are only 8 in yeast. Phylogenetic analysis of the DNA sequences of vertebrate PPM isoforms revealed that they can be divided into 12 different classes: PPM1A/PPM1B/PPM1N, PPM1D, PPM1E/PPM1F, PPM1G, PPM1H/PPM1J/PPM1M, PPM1K, PPM1L, ILKAP, PDP1/PDP2, PP2D1/PHLPP1/PHLPP2, TAB1, and PPTC7. PPM-family members have a conserved catalytic core region, which contains the metal-chelating residues. The different isoforms also have isoform specific regions within their catalytic core domain and terminal domains, and these regions may be involved in substrate recognition and/or functional regulation of the phosphatases. The twenty mammalian PPM phosphatases are involved in regulating diverse cellular functions, such as cell cycle control, cell differentiation, immune responses, and cell metabolism. Mutation, overexpression, or deletion of the PPM phosphatase gene results in abnormal cellular responses, which lead to various human diseases. This review focuses on the structures and biological functions of the PPM-phosphatase family and their associated diseases. The development of specific inhibitors against the PPM phosphatase family as a therapeutic strategy will also be discussed.
Collapse
|
10
|
Clausse V, Tao D, Debnath S, Fang Y, Tagad HD, Wang Y, Sun H, LeClair CA, Mazur SJ, Lane K, Shi ZD, Vasalatiy O, Eells R, Baker LK, Henderson MJ, Webb MR, Shen M, Hall MD, Appella E, Appella DH, Coussens NP. Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens. J Biol Chem 2019; 294:17654-17668. [PMID: 31481464 PMCID: PMC6873202 DOI: 10.1074/jbc.ra119.010201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/30/2019] [Indexed: 01/07/2023] Open
Abstract
WT P53-Induced Phosphatase 1 (WIP1) is a member of the magnesium-dependent serine/threonine protein phosphatase (PPM) family and is induced by P53 in response to DNA damage. In several human cancers, the WIP1 protein is overexpressed, which is generally associated with a worse prognosis. Although WIP1 is an attractive therapeutic target, no potent, selective, and bioactive small-molecule modulator with favorable pharmacokinetics has been reported. Phosphatase enzymes are among the most challenging targets for small molecules because of the difficulty of achieving both modulator selectivity and bioavailability. Another major obstacle has been the availability of robust and physiologically relevant phosphatase assays that are suitable for high-throughput screening. Here, we describe orthogonal biochemical WIP1 activity assays that utilize phosphopeptides from native WIP1 substrates. We optimized an MS assay to quantify the enzymatically dephosphorylated peptide reaction product in a 384-well format. Additionally, a red-shifted fluorescence assay was optimized in a 1,536-well format to enable real-time WIP1 activity measurements through the detection of the orthogonal reaction product, Pi. We validated these two optimized assays by quantitative high-throughput screening against the National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection and used secondary assays to confirm and evaluate inhibitors identified in the primary screen. Five inhibitors were further tested with an orthogonal WIP1 activity assay and surface plasmon resonance binding studies. Our results validate the application of miniaturized physiologically relevant and orthogonal WIP1 activity assays to discover small-molecule modulators from high-throughput screens.
Collapse
Affiliation(s)
- Victor Clausse
- Synthetic Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Subrata Debnath
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhong Fang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Harichandra D Tagad
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Hongmao Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Kelly Lane
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Zhen-Dan Shi
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Olga Vasalatiy
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Rebecca Eells
- Reaction Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355
| | - Lynn K Baker
- Reaction Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355
| | - Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Martin R Webb
- Francis Crick Institute, 1 Midland Road, London NW1 AT, United Kingdom
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel H Appella
- Synthetic Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| |
Collapse
|