1
|
Stickland BA, Greulich F, Uhlenhaut NH. The specificity of CTBP dehydrogenase inhibitors MTOB and 4-Cl-HIPP. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001415. [PMID: 39967797 PMCID: PMC11833458 DOI: 10.17912/micropub.biology.001415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
C-terminal binding proteins (CTBPs) are conserved transcriptional repressors important in cancer and inflammation. Uniquely amongst transcriptional co-regulators, CTBPs possess a functional dehydrogenase domain. Since multiple malignancies display elevated CTBP levels, CTBP inhibitors targeting this dehydrogenase domain have been developed. While the importance of CTBPs dehydrogenase function for transcriptional regulation remains unclear, several studies have relied on CTBP inhibitors. In vitro experiments have confirmed binding of these compounds to CTBP's active site, however evidence for specificity is lacking. To address this, we treated wildtype and Ctbp1 , 2 double knockout J774.1 cells with MTOB or 4-Cl-HIPP and performed RNA-seq. We observed that both inhibitors elicit distinct transcriptional changes indicating non-overlapping modes of action. Moreover, the majority of changes induced by either inhibitor are observed in Ctbp1/2 double knockout cells suggesting off-target effects. We hypothesize that those CTBP dehydrogenase inhibitors lack specificity to CTBPs and emphasise careful revaluation of findings inferred from studies using those inhibitors.
Collapse
Affiliation(s)
- Benjamin A. Stickland
- Metabolic Programming, TUM School of Life Sciences, Technical University of Munich, Munich, Bavaria, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Munich, Bavaria, Germany
| | - Franziska Greulich
- Metabolic Programming, TUM School of Life Sciences, Technical University of Munich, Munich, Bavaria, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Munich, Bavaria, Germany
| | - Nina Henriette Uhlenhaut
- Metabolic Programming, TUM School of Life Sciences, Technical University of Munich, Munich, Bavaria, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Munich, Bavaria, Germany
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Zentrum München, Munich, Bavaria, Germany
- German Center for Diabetes Research, Munich, Bavaria, Germany
| |
Collapse
|
2
|
Craig O, Lee S, Pilcher C, Saoud R, Abdirahman S, Salazar C, Williams N, Ascher D, Vary R, Luu J, Cowley K, Ramm S, Li MX, Thio N, Li J, Semple T, Simpson K, Gorringe K, Holien J. A new method for network bioinformatics identifies novel drug targets for mucinous ovarian carcinoma. NAR Genom Bioinform 2024; 6:lqae096. [PMID: 39184376 PMCID: PMC11344246 DOI: 10.1093/nargab/lqae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Mucinous ovarian carcinoma (MOC) is a subtype of ovarian cancer that is distinct from all other ovarian cancer subtypes and currently has no targeted therapies. To identify novel therapeutic targets, we developed and applied a new method of differential network analysis comparing MOC to benign mucinous tumours (in the absence of a known normal tissue of origin). This method mapped the protein-protein network in MOC and then utilised structural bioinformatics to prioritise the proteins identified as upregulated in the MOC network for their likelihood of being successfully drugged. Using this protein-protein interaction modelling, we identified the strongest 5 candidates, CDK1, CDC20, PRC1, CCNA2 and TRIP13, as structurally tractable to therapeutic targeting by small molecules. siRNA knockdown of these candidates performed in MOC and control normal fibroblast cell lines identified CDK1, CCNA2, PRC1 and CDC20, as potential drug targets in MOC. Three targets (TRIP13, CDC20, CDK1) were validated using known small molecule inhibitors. Our findings demonstrate the utility of our pipeline for identifying new targets and highlight potential new therapeutic options for MOC patients.
Collapse
Affiliation(s)
- Olivia Craig
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Samuel Lee
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Carlton, VIC 3010, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Courtney Pilcher
- School of Science, STEM College, RMIT University, Bundoora, VIC 3082, Australia
| | - Rita Saoud
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Suad Abdirahman
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Carolina Salazar
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Nathan Williams
- St Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- School of Science, STEM College, RMIT University, Bundoora, VIC 3082, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4067, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robert Vary
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Jennii Luu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Karla J Cowley
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Susanne Ramm
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Mark Xiang Li
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Niko Thio
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
| | - Jason Li
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
| | - Tim Semple
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
| | - Kaylene J Simpson
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Kylie L Gorringe
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jessica K Holien
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Carlton, VIC 3010, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- School of Science, STEM College, RMIT University, Bundoora, VIC 3082, Australia
| |
Collapse
|
3
|
Huang M, Li Y, Li Y, Liu S. C-Terminal Binding Protein: Regulator between Viral Infection and Tumorigenesis. Viruses 2024; 16:988. [PMID: 38932279 PMCID: PMC11209466 DOI: 10.3390/v16060988] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
C-terminal binding protein (CtBP), a transcriptional co-repressor, significantly influences cellular signaling, impacting various biological processes including cell proliferation, differentiation, apoptosis, and immune responses. The CtBP family comprises two highly conserved proteins, CtBP1 and CtBP2, which have been shown to play critical roles in both tumorigenesis and the regulation of viral infections. Elevated CtBP expression is noted in various tumor tissues, promoting tumorigenesis, invasiveness, and metastasis through multiple pathways. Additionally, CtBP's role in viral infections varies, exhibiting differing or even opposing effects depending on the virus. This review synthesizes the advances in CtBP's function research in viral infections and virus-associated tumorigenesis, offering new insights into potential antiviral and anticancer strategies.
Collapse
Affiliation(s)
- Meihui Huang
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yucong Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yuxiao Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Shuiping Liu
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| |
Collapse
|
4
|
Filograna A, De Tito S, Monte ML, Oliva R, Bruzzese F, Roca MS, Zannetti A, Greco A, Spano D, Ayala I, Liberti A, Petraccone L, Dathan N, Catara G, Schembri L, Colanzi A, Budillon A, Beccari AR, Del Vecchio P, Luini A, Corda D, Valente C. Identification and characterization of a new potent inhibitor targeting CtBP1/BARS in melanoma cells. J Exp Clin Cancer Res 2024; 43:137. [PMID: 38711119 PMCID: PMC11071220 DOI: 10.1186/s13046-024-03044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The C-terminal-binding protein 1/brefeldin A ADP-ribosylation substrate (CtBP1/BARS) acts both as an oncogenic transcriptional co-repressor and as a fission inducing protein required for membrane trafficking and Golgi complex partitioning during mitosis, hence for mitotic entry. CtBP1/BARS overexpression, in multiple cancers, has pro-tumorigenic functions regulating gene networks associated with "cancer hallmarks" and malignant behavior including: increased cell survival, proliferation, migration/invasion, epithelial-mesenchymal transition (EMT). Structurally, CtBP1/BARS belongs to the hydroxyacid-dehydrogenase family and possesses a NAD(H)-binding Rossmann fold, which, depending on ligands bound, controls the oligomerization of CtBP1/BARS and, in turn, its cellular functions. Here, we proposed to target the CtBP1/BARS Rossmann fold with small molecules as selective inhibitors of mitotic entry and pro-tumoral transcriptional activities. METHODS Structured-based screening of drug databases at different development stages was applied to discover novel ligands targeting the Rossmann fold. Among these identified ligands, N-(3,4-dichlorophenyl)-4-{[(4-nitrophenyl)carbamoyl]amino}benzenesulfonamide, called Comp.11, was selected for further analysis. Fluorescence spectroscopy, isothermal calorimetry, computational modelling and site-directed mutagenesis were employed to define the binding of Comp.11 to the Rossmann fold. Effects of Comp.11 on the oligomerization state, protein partners binding and pro-tumoral activities were evaluated by size-exclusion chromatography, pull-down, membrane transport and mitotic entry assays, Flow cytometry, quantitative real-time PCR, motility/invasion, and colony assays in A375MM and B16F10 melanoma cell lines. Effects of Comp.11 on tumor growth in vivo were analyzed in mouse tumor model. RESULTS We identify Comp.11 as a new, potent and selective inhibitor of CtBP1/BARS (but not CtBP2). Comp.11 directly binds to the CtBP1/BARS Rossmann fold affecting the oligomerization state of the protein (unlike other known CtBPs inhibitors), which, in turn, hinders interactions with relevant partners, resulting in the inhibition of both CtBP1/BARS cellular functions: i) membrane fission, with block of mitotic entry and cellular secretion; and ii) transcriptional pro-tumoral effects with significantly hampered proliferation, EMT, migration/invasion, and colony-forming capabilities. The combination of these effects impairs melanoma tumor growth in mouse models. CONCLUSIONS: This study identifies a potent and selective inhibitor of CtBP1/BARS active in cellular and melanoma animal models revealing new opportunities to study the role of CtBP1/BARS in tumor biology and to develop novel melanoma treatments.
Collapse
Affiliation(s)
- Angela Filograna
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Stefano De Tito
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK. The Study Has Been Previously Performed at IEOS-CNR, Naples, Italy
| | - Matteo Lo Monte
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Rosario Oliva
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Francesca Bruzzese
- Animal Facility Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), Naples, 80145, Italy
| | - Adelaide Greco
- Interdepartmental Service Center of Veterinary Radiology, University of Naples Federico II, 80137, Naples, Italy
| | - Daniela Spano
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Inmaculada Ayala
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Assunta Liberti
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Luigi Petraccone
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Nina Dathan
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), 80131, Naples, Italy
| | - Laura Schembri
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | | | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Alberto Luini
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Daniela Corda
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Carmen Valente
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
- Present address: Dompé Farmaceutici S.P.A, L'Aquila, Italy.
| |
Collapse
|
5
|
Dcona MM, Chougoni KK, Dcona DT, West JL, Singh SJ, Ellis KC, Grossman SR. Combined Targeting of NAD Biosynthesis and the NAD-dependent Transcription Factor C-terminal Binding Protein as a Promising Novel Therapy for Pancreatic Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:2003-2013. [PMID: 37707363 PMCID: PMC10549224 DOI: 10.1158/2767-9764.crc-22-0521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/07/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Cancer therapies targeting metabolic derangements unique to cancer cells are emerging as a key strategy to address refractory solid tumors such as pancreatic ductal adenocarcinomas (PDAC) that exhibit resistance to extreme nutrient deprivation in the tumor microenvironment. Nicotinamide adenine dinucleotide (NAD) participates in multiple metabolic pathways and nicotinamide phosphoribosyl transferase (NAMPT) is one of the key intracellular enzymes that facilitate the synthesis of NAD. C-terminal binding proteins 1 and 2 (CtBP) are paralogous NAD-dependent oncogenic transcription factors and dehydrogenases that nucleate an epigenetic complex regulating a cohort of genes responsible for cancer proliferation and metastasis. As adequate intracellular NAD is required for CtBP to oligomerize and execute its oncogenic transcriptional coregulatory activities, we hypothesized that NAD depletion would synergize with CtBP inhibition, improving cell inhibitory efficacy. Indeed, depletion of cellular NAD via the NAMPT inhibitor GMX1778 enhanced growth inhibition induced by either RNAi-mediated CtBP1/2 knockdown or the CtBP dehydrogenase inhibitor 4-chlorophenyl-2-hydroxyimino propanoic acid as much as 10-fold in PDAC cells, while untransformed pancreatic ductal cells were unaffected. The growth inhibitory effects of the NAMPT/CtBP inhibitor combination correlated pharmacodynamically with on-target disruption of CtBP1/2 dimerization, CtBP2 interaction with the CoREST epigenetic regulator, and transcriptional activation of the oncogenic target gene TIAM1. Moreover, this same therapeutic combination strongly attenuated growth of PDAC cell line xenografts in immunodeficient mice, with no observable toxicity. Collectively, our data demonstrate that targeting CtBP in combination with NAD depletion represents a promising therapeutic strategy for PDAC. SIGNIFICANCE Effective precision therapies are lacking in PDAC. We demonstrate that simultaneous inhibition of NAD metabolism and the oncoprotein CtBP is potently effective at blocking growth of both PDAC cells in culture and human PDAC-derived tumors in mice and should be explored further as a potential therapy for patients with PDAC.
Collapse
Affiliation(s)
- M. Michael Dcona
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kranthi Kumar Chougoni
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Diana T. Dcona
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jacqueline L. West
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
| | - Sahib J. Singh
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Keith C. Ellis
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Steven R. Grossman
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
6
|
Kellogg GE, Cen Y, Dukat M, Ellis KC, Guo Y, Li J, May AE, Safo MK, Zhang S, Zhang Y, Desai UR. Merging cultures and disciplines to create a drug discovery ecosystem at Virginia commonwealth university: Medicinal chemistry, structural biology, molecular and behavioral pharmacology and computational chemistry. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:255-269. [PMID: 36863508 PMCID: PMC10619687 DOI: 10.1016/j.slasd.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
The Department of Medicinal Chemistry, together with the Institute for Structural Biology, Drug Discovery and Development, at Virginia Commonwealth University (VCU) has evolved, organically with quite a bit of bootstrapping, into a unique drug discovery ecosystem in response to the environment and culture of the university and the wider research enterprise. Each faculty member that joined the department and/or institute added a layer of expertise, technology and most importantly, innovation, that fertilized numerous collaborations within the University and with outside partners. Despite moderate institutional support with respect to a typical drug discovery enterprise, the VCU drug discovery ecosystem has built and maintained an impressive array of facilities and instrumentation for drug synthesis, drug characterization, biomolecular structural analysis and biophysical analysis, and pharmacological studies. Altogether, this ecosystem has had major impacts on numerous therapeutic areas, such as neurology, psychiatry, drugs of abuse, cancer, sickle cell disease, coagulopathy, inflammation, aging disorders and others. Novel tools and strategies for drug discovery, design and development have been developed at VCU in the last five decades; e.g., fundamental rational structure-activity relationship (SAR)-based drug design, structure-based drug design, orthosteric and allosteric drug design, design of multi-functional agents towards polypharmacy outcomes, principles on designing glycosaminoglycans as drugs, and computational tools and algorithms for quantitative SAR (QSAR) and understanding the roles of water and the hydrophobic effect.
Collapse
Affiliation(s)
- Glen E Kellogg
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA.
| | - Yana Cen
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Malgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Keith C Ellis
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Youzhong Guo
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Jiong Li
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Aaron E May
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Martin K Safo
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Umesh R Desai
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA.
| |
Collapse
|
7
|
Simulation-guided relationships and interaction characteristics of human CtBP1 in complex with protocatechualdehyde. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Erlandsen H, Jecrois AM, Nichols JC, Cole JL, Royer WE. NADH/NAD + binding and linked tetrameric assembly of the oncogenic transcription factors CtBP1 and CtBP2. FEBS Lett 2022; 596:479-490. [PMID: 34997967 DOI: 10.1002/1873-3468.14276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022]
Abstract
The activation of oncogenic C-terminal binding Protein (CtBP) transcriptional activity is coupled with NAD(H) binding and homo-oligomeric assembly, although the level of CtBP assembly and nucleotide binding affinity continues to be debated. Here, we apply biophysical techniques to address these fundamental issues for CtBP1 and CtBP2. Our ultracentrifugation results unambiguously demonstrate that CtBP assembles into tetramers in the presence of saturating NAD+ or NADH with tetramer to dimer dissociation constants about 100 nm. Isothermal titration calorimetry measurements of NAD(H) binding to CtBP show dissociation constants between 30 and 500 nm, depending on the nucleotide and paralog. Given cellular levels of NAD+ , CtBP is likely to be fully saturated with NAD under physiological concentrations suggesting that CtBP is unable to act as a sensor for NADH levels.
Collapse
Affiliation(s)
- Heidi Erlandsen
- Center for Open Research Resources & Equipment, University of Connecticut, Storrs, CT, USA
| | - Anne M Jecrois
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA
| | - Jeffry C Nichols
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA.,Chemistry Department, Worcester State University, MA, USA
| | - James L Cole
- Department of Molecular and Cell Biology, Department of Chemistry, University of Connecticut, CT, USA
| | - William E Royer
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
9
|
Transcriptomics and Metabolomics Integration Reveals Redox-Dependent Metabolic Rewiring in Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13205058. [PMID: 34680207 PMCID: PMC8534001 DOI: 10.3390/cancers13205058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
Rewiring glucose metabolism toward aerobic glycolysis provides cancer cells with a rapid generation of pyruvate, ATP, and NADH, while pyruvate oxidation to lactate guarantees refueling of oxidized NAD+ to sustain glycolysis. CtPB2, an NADH-dependent transcriptional co-regulator, has been proposed to work as an NADH sensor, linking metabolism to epigenetic transcriptional reprogramming. By integrating metabolomics and transcriptomics in a triple-negative human breast cancer cell line, we show that genetic and pharmacological down-regulation of CtBP2 strongly reduces cell proliferation by modulating the redox balance, nucleotide synthesis, ROS generation, and scavenging. Our data highlight the critical role of NADH in controlling the oncogene-dependent crosstalk between metabolism and the epigenetically mediated transcriptional program that sustains energetic and anabolic demands in cancer cells.
Collapse
|
10
|
The transrepression and transactivation roles of CtBPs in the pathogenesis of different diseases. J Mol Med (Berl) 2021; 99:1335-1347. [PMID: 34196767 DOI: 10.1007/s00109-021-02107-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Gene transcription is strictly controlled by transcriptional complexes, which are assemblies of transcription factors, transcriptional regulators, and co-regulators. Mammalian genomes encode two C-terminal-binding proteins (CtBPs), CtBP1 and CtBP2, which are both well-known transcriptional corepressors of oncogenic processes. Their overexpression in tumors is associated with malignant behavior, such as uncontrolled cell proliferation, migration, and invasion, as well as with an increase in the epithelial-mesenchymal transition. CtBPs coordinate with other transcriptional regulators, such as histone deacetylases (HDACs) and histone acetyltransferases (p300 and CBP [CREBP-binding protein]) that contain the PXDLS motif, and with transcription factors to assemble transcriptional complexes that dock onto the promoters of genes to initiate gene transcription. Emerging evidence suggests that CtBPs function as both corepressors and coactivators in different biological processes ranging from apoptosis to inflammation and osteogenesis. Therapeutic targeting of CtBPs or the interactions required to form transcriptional complexes has also shown promising effects in preventing disease progression. This review summarizes the most recent progress in the study of CtBP functions and therapeutic inhibitors in different biological processes. This knowledge may enable a better understanding of the complexity of the roles of CtBPs, while providing new insights into therapeutic strategies that target CtBPs.
Collapse
|
11
|
Ohashi M, Hayes M, McChesney K, Johannsen E. Epstein-Barr virus nuclear antigen 3C (EBNA3C) interacts with the metabolism sensing C-terminal binding protein (CtBP) repressor to upregulate host genes. PLoS Pathog 2021; 17:e1009419. [PMID: 33720992 PMCID: PMC7993866 DOI: 10.1371/journal.ppat.1009419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/25/2021] [Accepted: 02/22/2021] [Indexed: 12/04/2022] Open
Abstract
Epstein-Barr virus (EBV) infection is associated with the development of specific types of lymphoma and some epithelial cancers. EBV infection of resting B-lymphocytes in vitro drives them to proliferate as lymphoblastoid cell lines (LCLs) and serves as a model for studying EBV lymphomagenesis. EBV nuclear antigen 3C (EBNA3C) is one of the genes required for LCL growth and previous work has suggested that suppression of the CDKN2A encoded tumor suppressor p16INK4A and possibly p14ARF is central to EBNA3C’s role in this growth transformation. To directly assess whether loss of p16 and/or p14 was sufficient to explain EBNA3C growth effects, we used CRISPR/Cas9 to disrupt specific CDKN2A exons in EBV transformed LCLs. Disruption of p16 specific exon 1α and the p16/p14 shared exon 2 were each sufficient to restore growth in the absence of EBNA3C. Using EBNA3C conditional LCLs knocked out for either exon 1α or 2, we identified EBNA3C induced and repressed genes. By trans-complementing with EBNA3C mutants, we determined specific genes that require EBNA3C interaction with RBPJ or CtBP for their regulation. Unexpectedly, interaction with the CtBP repressor was required not only for repression, but also for EBNA3C induction of many host genes. Contrary to previously proposed models, we found that EBNA3C does not recruit CtBP to the promoters of these genes. Instead, our results suggest that CtBP is bound to these promoters in the absence of EBNA3C and that EBNA3C interaction with CtBP interferes with the repressive function of CtBP, leading to EBNA3C mediated upregulation. Epstein-Barr virus (EBV) is a gammaherpesvirus that establishes lifelong infection in about 95% of adult humans. EBV infection is usually benign, but can rarely result in several different malignancies, particularly lymphomas. EBV infection of resting B-lymphocytes in the laboratory drives them to proliferate as lymphoblastoid cell lines (LCLs), a model for EBV lymphomagenesis. In this manuscript we study how one EBV protein expressed in LCLs, EBNA3C, contributes to B lymphocyte transformation. Prior work has established that EBNA3C turns off the CDKN2A gene, but there is disagreement regarding the relative importance of silencing the two CDKN2A gene products: p14 and p16. Using a CRISPR/Cas9 gene editing strategy we confirm that p16 knock-out rescues LCL growth in the absence of EBNA3C even in the presence of wildtype p14. We then use these knock-out LCLs to identify EBNA3C regulated genes and uncover extensive growth-independent changes in B lymphocytes due to the EBNA3C transcription factor. We also discover an unexpected role for the CtBP repressor protein in EBNA3C gene upregulation. Contrary to prior models, we do not observe CtBP recruitment to target genes by EBNA3C. Instead, our data are consistent with EBNA3C interfering with the ability of pre-bound CtBP to repress genes.
Collapse
Affiliation(s)
- Makoto Ohashi
- Department of Medicine, Division of Infectious Diseases, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mitchell Hayes
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kyle McChesney
- Department of Medicine, Division of Infectious Diseases, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eric Johannsen
- Department of Medicine, Division of Infectious Diseases, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
12
|
Nichols JC, Schiffer CA, Royer WE. NAD(H) phosphates mediate tetramer assembly of human C-terminal binding protein (CtBP). J Biol Chem 2021; 296:100351. [PMID: 33524397 PMCID: PMC7949142 DOI: 10.1016/j.jbc.2021.100351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
C-terminal binding proteins (CtBPs) are cotranscriptional factors that play key roles in cell fate. We have previously shown that NAD(H) promotes the assembly of similar tetramers from either human CtBP1 and CtBP2 and that CtBP2 tetramer destabilizing mutants are defective for oncogenic activity. To assist structure-based design efforts for compounds that disrupt CtBP tetramerization, it is essential to understand how NAD(H) triggers tetramer assembly. Here, we investigate the moieties within NAD(H) that are responsible for triggering tetramer formation. Using multiangle light scattering (MALS), we show that ADP is able to promote tetramer formation of both CtBP1 and CtBP2, whereas AMP promotes tetramer assembly of CtBP1, but not CtBP2. Other NAD(H) moieties that lack the adenosine phosphate, including adenosine and those incorporating nicotinamide, all fail to promote tetramer assembly. Our crystal structures of CtBP1 with AMP reveal participation of the adenosine phosphate in the tetrameric interface, pinpointing its central role in NAD(H)-linked assembly. CtBP1 and CtBP2 have overlapping but unique roles, suggesting that a detailed understanding of their unique structural properties might have utility in the design of paralog-specific inhibitors. We investigated the different responses to AMP through a series of site-directed mutants at 13 positions. These mutations reveal a central role for a hinge segment, which we term the 120s hinge that connects the substrate with coenzyme-binding domains and influences nucleotide binding and tetramer assembly. Our results provide insight into suitable pockets to explore in structure-based drug design to interfere with cotranscriptional activity of CtBP in cancer.
Collapse
Affiliation(s)
- Jeffry C Nichols
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA; Chemistry Department, Worcester State University, Worcester, Massachusetts, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - William E Royer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| |
Collapse
|
13
|
Identification of Host Cellular Protein Substrates of SARS-COV-2 Main Protease. Int J Mol Sci 2020; 21:ijms21249523. [PMID: 33333742 PMCID: PMC7765187 DOI: 10.3390/ijms21249523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/20/2022] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease-19 (COVID-19) being associated with severe pneumonia. Like with other viruses, the interaction of SARS-CoV-2 with host cell proteins is necessary for successful replication, and cleavage of cellular targets by the viral protease also may contribute to the pathogenesis, but knowledge about the human proteins that are processed by the main protease (3CLpro) of SARS-CoV-2 is still limited. We tested the prediction potentials of two different in silico methods for the identification of SARS-CoV-2 3CLpro cleavage sites in human proteins. Short stretches of homologous host-pathogen protein sequences (SSHHPS) that are present in SARS-CoV-2 polyprotein and human proteins were identified using BLAST analysis, and the NetCorona 1.0 webserver was used to successfully predict cleavage sites, although this method was primarily developed for SARS-CoV. Human C-terminal-binding protein 1 (CTBP1) was found to be cleaved in vitro by SARS-CoV-2 3CLpro, the existence of the cleavage site was proved experimentally by using a His6-MBP-mEYFP recombinant substrate containing the predicted target sequence. Our results highlight both potentials and limitations of the tested algorithms. The identification of candidate host substrates of 3CLpro may help better develop an understanding of the molecular mechanisms behind the replication and pathogenesis of SARS-CoV-2.
Collapse
|
14
|
Jecrois AM, Dcona MM, Deng X, Bandyopadhyay D, Grossman SR, Schiffer CA, Royer WE. Cryo-EM structure of CtBP2 confirms tetrameric architecture. Structure 2020; 29:310-319.e5. [PMID: 33264605 DOI: 10.1016/j.str.2020.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/10/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
C-terminal binding proteins 1 and 2 (CtBP1 and CtBP2) are transcriptional regulators that activate or repress many genes involved in cellular development, apoptosis, and metastasis. NADH-dependent CtBP activation has been implicated in multiple types of cancer and poor patient prognosis. Central to understanding activation of CtBP in oncogenesis is uncovering how NADH triggers protein assembly, what level of assembly occurs, and if oncogenic activity depends upon such assembly. Here, we present the cryoelectron microscopic structures of two different constructs of CtBP2 corroborating that the native state of CtBP2 in the presence of NADH is tetrameric. The physiological relevance of the observed tetramer was demonstrated in cell culture, showing that CtBP tetramer-destabilizing mutants are defective for cell migration, transcriptional repression of E-cadherin, and activation of TIAM1. Together with our cryoelectron microscopy studies, these results highlight the tetramer as the functional oligomeric form of CtBP2.
Collapse
Affiliation(s)
- Anne M Jecrois
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - M Michael Dcona
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiaoyan Deng
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Dipankar Bandyopadhyay
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Steven R Grossman
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - William E Royer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
15
|
Paredes R, Kelly JR, Geary B, Almarzouq B, Schneider M, Pearson S, Narayanan P, Williamson A, Lovell SC, Wiseman DH, Chadwick JA, Jones NJ, Kustikova O, Schambach A, Garner T, Amaral FMR, Pierce A, Stevens A, Somervaille TCP, Whetton AD, Meyer S. EVI1 phosphorylation at S436 regulates interactions with CtBP1 and DNMT3A and promotes self-renewal. Cell Death Dis 2020; 11:878. [PMID: 33082307 PMCID: PMC7576810 DOI: 10.1038/s41419-020-03099-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 11/22/2022]
Abstract
The transcriptional regulator EVI1 has an essential role in early development and haematopoiesis. However, acute myeloid leukaemia (AML) driven by aberrantly high EVI1 expression has very poor prognosis. To investigate the effects of post-translational modifications on EVI1 function, we carried out a mass spectrometry (MS) analysis of EVI1 in AML and detected dynamic phosphorylation at serine 436 (S436). Wild-type EVI1 (EVI1-WT) with S436 available for phosphorylation, but not non-phosphorylatable EVI1-S436A, conferred haematopoietic progenitor cell self-renewal and was associated with significantly higher organised transcriptional patterns. In silico modelling of EVI1-S436 phosphorylation showed reduced affinity to CtBP1, and CtBP1 showed reduced interaction with EVI1-WT compared with EVI1-S436A. The motif harbouring S436 is a target of CDK2 and CDK3 kinases, which interacted with EVI1-WT. The methyltransferase DNMT3A bound preferentially to EVI1-WT compared with EVI1-S436A, and a hypomethylated cell population associated by EVI1-WT expression in murine haematopoietic progenitors is not maintained with EVI1-S436A. These data point to EVI1-S436 phosphorylation directing functional protein interactions for haematopoietic self-renewal. Targeting EVI1-S436 phosphorylation may be of therapeutic benefit when treating EVI1-driven leukaemia.
Collapse
Affiliation(s)
- Roberto Paredes
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - James R Kelly
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Bethany Geary
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Batool Almarzouq
- Department of Biochemistry, Institute of Integrative Biology/School of Life Sciences, University of Liverpool, Liverpool, UK
| | - Marion Schneider
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Stella Pearson
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Prakrithi Narayanan
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Andrew Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Simon C Lovell
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Daniel H Wiseman
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Epigenetics of Haematopoiesis Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - John A Chadwick
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Leukaemia Biology Laboratory, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Nigel J Jones
- Department of Biochemistry, Institute of Integrative Biology/School of Life Sciences, University of Liverpool, Liverpool, UK
| | - Olga Kustikova
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Terence Garner
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Fabio M R Amaral
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Leukaemia Biology Laboratory, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Andrew Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
| | - Adam Stevens
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Tim C P Somervaille
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Leukaemia Biology Laboratory, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Anthony D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK
- Stoller Biomarker Discovery Centre, University of Manchester, Manchester, UK
| | - Stefan Meyer
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester, UK.
- Department of Paediatric Haematology and Oncology, Royal Manchester Children's Hospital, Manchester, UK.
- Young Oncology Unit, The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
16
|
Deng Y, Guo W, Li G, Li S, Li H, Li X, Niu B, Song M, Zhang Y, Xu Z, Li F. Protocatechuic Aldehyde Represses Proliferation and Migration of Breast Cancer Cells through Targeting C-terminal Binding Protein 1. J Breast Cancer 2020; 23:20-35. [PMID: 32140267 PMCID: PMC7043946 DOI: 10.4048/jbc.2020.23.e7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 12/02/2019] [Indexed: 11/30/2022] Open
Abstract
Purpose C-terminal binding protein 1 (CtBP1) is a transcriptional co-repressor that is overexpressed in many cancers. CtBP1 transcriptionally represses a broad array of tumor suppressors, which promotes cancer cell proliferation, migration, invasion, and resistance to apoptosis. Recent studies have demonstrated that CtBP1 is a potential target for cancer therapy. This study was designed to screen for compounds that potentially target CtBP1. Methods Using a structure-based virtual screening for CtBP1 inhibitors, we found protocatechuic aldehyde (PA), a natural compound found in the root of a traditional Chinese herb, Salvia miltiorrhiza, that directly binds to CtBP1. Microscale thermophoresis assay was performed to determine whether PA and CtBP1 directly bind to each other. Further, clustered regularly interspaced short palindromic repeats associated Cas9 nuclease-mediated CtBP1 knockout in breast cancer cells was used to validate the CtBP1 targeting specificity of PA. Results Functional studies showed that PA repressed the proliferation and migration of breast cancer cells. Furthermore, PA elevated the expression of the downstream targets of CtBP1, p21 and E-cadherin, and decreased CtBP1 binding affinity for the promoter regions of p21 and E-cadherin in breast cancer cells. However, PA did not affect the expression of p21 and E-cadherin in the CtBP1 knockout breast cancer cells. In addition, the CtBP1 knockout breast cancer cells showed resistance to PA-induced repression of proliferation and migration. Conclusion Our findings demonstrated that PA directly bound to CtBP1 and inhibited the growth and migration of breast cancer cells through CtBP1 inhibition. Structural modifications of PA are further required to enhance its binding affinity and selectivity for CtBP1.
Collapse
Affiliation(s)
- Yu Deng
- School of Medicine, Chengdu University, Chengdu, China.,Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, China
| | - Wanjun Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guancheng Li
- The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuang Li
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Hong Li
- Department of Dermatology, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Xinyan Li
- Department of Epidemiology and Biostatistics, College for Public Health and Social Justice, Saint Louis University, St. Louis, MO, USA
| | - Bei Niu
- School of Medicine, Chengdu University, Chengdu, China
| | - Mingzhu Song
- School of Medicine, Chengdu University, Chengdu, China
| | - Yamei Zhang
- Central Lab, Chengdu Univerisity Hospital, Chengdu, China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Jia Y, Burbidge CA, Sweetman C, Schutz E, Soole K, Jenkins C, Hancock RD, Bruning JB, Ford CM. An aldo-keto reductase with 2-keto-l-gulonate reductase activity functions in l-tartaric acid biosynthesis from vitamin C in Vitis vinifera. J Biol Chem 2019; 294:15932-15946. [PMID: 31488549 PMCID: PMC6827314 DOI: 10.1074/jbc.ra119.010196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/22/2019] [Indexed: 12/25/2022] Open
Abstract
Tartaric acid has high economic value as an antioxidant and flavorant in food and wine industries. l-Tartaric acid biosynthesis in wine grape (Vitis vinifera) uses ascorbic acid (vitamin C) as precursor, representing an unusual metabolic fate for ascorbic acid degradation. Reduction of the ascorbate breakdown product 2-keto-l-gulonic acid to l-idonic acid constitutes a critical step in this l-tartaric acid biosynthetic pathway. However, the underlying enzymatic mechanisms remain obscure. Here, we identified a V. vinifera aldo-keto reductase, Vv2KGR, with 2-keto-l-gulonic acid reductase activity. Vv2KGR belongs to the d-isomer-specific 2-hydroxyacid dehydrogenase superfamily and displayed the highest similarity to the hydroxyl pyruvate reductase isoform 2 in Arabidopsis thaliana Enzymatic analyses revealed that Vv2KGR efficiently reduces 2-keto-l-gulonic acid to l-idonic acid and uses NADPH as preferred coenzyme. Moreover, Vv2KGR exhibited broad substrate specificity toward glyoxylate, pyruvate, and hydroxypyruvate, having the highest catalytic efficiency for glyoxylate. We further determined the X-ray crystal structure of Vv2KGR at 1.58 Å resolution. Comparison of the Vv2KGR structure with those of d-isomer-specific 2-hydroxyacid dehydrogenases from animals and microorganisms revealed several unique structural features of this plant hydroxyl pyruvate reductase. Substrate structural analysis indicated that Vv2KGR uses two modes (A and B) to bind different substrates. 2-Keto-l-gulonic acid displayed the lowest predicted free-energy binding to Vv2KGR among all docked substrates. Hence, we propose that Vv2KGR functions in l-tartaric acid biosynthesis. To the best of our knowledge, this is the first report of a d-isomer-specific 2-hydroxyacid dehydrogenase that reduces 2-keto-l-gulonic acid to l-idonic acid in plants.
Collapse
Affiliation(s)
- Yong Jia
- Waite Research Institute, School of Agriculture, Food, and Wine, University of Adelaide, Adelaide 5064, Australia
| | - Crista A Burbidge
- College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide 5001, Australia
| | - Crystal Sweetman
- College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide 5001, Australia
| | - Emi Schutz
- Waite Research Institute, School of Agriculture, Food, and Wine, University of Adelaide, Adelaide 5064, Australia
| | - Kathy Soole
- College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide 5001, Australia
| | - Colin Jenkins
- College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide 5001, Australia
| | - Robert D Hancock
- Cell and Molecular Sciences, James Hutton Institute, Invergowrie, Dundee DD2 5DA, United Kingdom
| | - John B Bruning
- Institute of Photonics and Advanced Sensing, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Christopher M Ford
- Waite Research Institute, School of Agriculture, Food, and Wine, University of Adelaide, Adelaide 5064, Australia
| |
Collapse
|
18
|
Byun JS, Park S, Yi DI, Shin JH, Hernandez SG, Hewitt SM, Nicklaus MC, Peach ML, Guasch L, Tang B, Wakefield LM, Yan T, Caban A, Jones A, Kabbout M, Vohra N, Nápoles AM, Singhal S, Yancey R, De Siervi A, Gardner K. Epigenetic re-wiring of breast cancer by pharmacological targeting of C-terminal binding protein. Cell Death Dis 2019; 10:689. [PMID: 31534138 PMCID: PMC6751206 DOI: 10.1038/s41419-019-1892-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/17/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
The C-terminal binding protein (CtBP) is an NADH-dependent dimeric family of nuclear proteins that scaffold interactions between transcriptional regulators and chromatin-modifying complexes. Its association with poor survival in several cancers implicates CtBP as a promising target for pharmacological intervention. We employed computer-assisted drug design to search for CtBP inhibitors, using quantitative structure-activity relationship (QSAR) modeling and docking. Functional screening of these drugs identified 4 compounds with low toxicity and high water solubility. Micro molar concentrations of these CtBP inhibitors produces significant de-repression of epigenetically silenced pro-epithelial genes, preferentially in the triple-negative breast cancer cell line MDA-MB-231. This epigenetic reprogramming occurs through eviction of CtBP from gene promoters; disrupted recruitment of chromatin-modifying protein complexes containing LSD1, and HDAC1; and re-wiring of activating histone marks at targeted genes. In functional assays, CtBP inhibition disrupts CtBP dimerization, decreases cell migration, abolishes cellular invasion, and improves DNA repair. Combinatorial use of CtBP inhibitors with the LSD1 inhibitor pargyline has synergistic influence. Finally, integrated correlation of gene expression in breast cancer patients with nuclear levels of CtBP1 and LSD1, reveals new potential therapeutic vulnerabilities. These findings implicate a broad role for this class of compounds in strategies for epigenetically targeted therapeutic intervention.
Collapse
Affiliation(s)
- Jung S Byun
- National Institute on Minority Health and Health Disparities, Bethesda, MD, 20892, USA
| | - Samson Park
- Genetics Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Dae Ik Yi
- Genetics Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Jee-Hye Shin
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | | | - Stephen M Hewitt
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Marc C Nicklaus
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 20892, USA
| | - Megan L Peach
- Basic Science Program, Chemical Biology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Laura Guasch
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 20892, USA
| | - Binwu Tang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Lalage M Wakefield
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Tingfen Yan
- National Human Genome Institute, Bethesda, MD, 20892, USA
| | - Ambar Caban
- National Institute on Minority Health and Health Disparities, Bethesda, MD, 20892, USA
| | - Alana Jones
- National Institute on Minority Health and Health Disparities, Bethesda, MD, 20892, USA
| | - Mohamed Kabbout
- National Institute on Minority Health and Health Disparities, Bethesda, MD, 20892, USA
| | - Nasreen Vohra
- Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA
| | - Anna María Nápoles
- National Institute on Minority Health and Health Disparities, Bethesda, MD, 20892, USA
| | - Sandeep Singhal
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Ryan Yancey
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Adriana De Siervi
- Laboratorio de Oncologıa Molecular y Nuevos Blancos Terapeuticos, Instituto de Biologıa y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Kevin Gardner
- National Institute on Minority Health and Health Disparities, Bethesda, MD, 20892, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
19
|
Dcona MM, Damle PK, Zarate-Perez F, Morris BL, Nawaz Z, Dennis MJ, Deng X, Korwar S, Singh SJ, Ellis KC, Royer WE, Bandyopadhyay D, Escalante C, Grossman SR. Active-Site Tryptophan, the Target of Antineoplastic C-Terminal Binding Protein Inhibitors, Mediates Inhibitor Disruption of CtBP Oligomerization and Transcription Coregulatory Activities. Mol Pharmacol 2019; 96:99-108. [PMID: 31036695 PMCID: PMC6560337 DOI: 10.1124/mol.118.114363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 04/21/2019] [Indexed: 01/20/2023] Open
Abstract
C-terminal binding proteins (CtBP1/2) are oncogenic transcriptional coregulators and dehydrogenases often overexpressed in multiple solid tumors, including breast, colon, and ovarian cancer, and associated with poor survival. CtBPs act by repressing expression of genes responsible for apoptosis (e.g., PUMA, BIK) and metastasis-associated epithelial-mesenchymal transition (e.g., CDH1), and by activating expression of genes that promote migratory and invasive properties of cancer cells (e.g., TIAM1) and genes responsible for enhanced drug resistance (e.g., MDR1). CtBP's transcriptional functions are also critically dependent on oligomerization and nucleation of transcriptional complexes. Recently, we have developed a family of CtBP dehydrogenase inhibitors, based on the parent 2-hydroxyimino-3-phenylpropanoic acid (HIPP), that specifically disrupt cancer cell viability, abrogate CtBP's transcriptional function, and block polyp formation in a mouse model of intestinal polyposis that depends on CtBP's oncogenic functions. Crystallographic analysis revealed that HIPP interacts with CtBP1/2 at a conserved active site tryptophan (W318/324; CtBP1/2) that is unique among eukaryotic D2-dehydrogenases. To better understand the mechanism of action of HIPP-class inhibitors, we investigated the contribution of W324 to CtBP2's biochemical and physiologic activities utilizing mutational analysis. Indeed, W324 was necessary for CtBP2 self-association, as shown by analytical ultracentrifugation and in vivo cross-linking. Additionally, W324 supported CtBP's association with the transcriptional corepressor CoREST, and was critical for CtBP2 induction of cell motility. Notably, the HIPP derivative 4-chloro-HIPP biochemically and biologically phenocopied mutational inactivation of CtBP2 W324. Our data support further optimization of W318/W324-interacting CtBP dehydrogenase inhibitors that are emerging as a novel class of cancer cell-specific therapeutic.
Collapse
Affiliation(s)
- M Michael Dcona
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Priyadarshan K Damle
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Francisco Zarate-Perez
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Benjamin L Morris
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Zaid Nawaz
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Michael J Dennis
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Xiaoyan Deng
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Sudha Korwar
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Sahib J Singh
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Keith C Ellis
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - William E Royer
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Dipankar Bandyopadhyay
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Carlos Escalante
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Steven R Grossman
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| |
Collapse
|
20
|
Blevins MA, Zhang C, Zhang L, Li H, Li X, Norris DA, Huang M, Zhao R. CPP-E1A fusion peptides inhibit CtBP-mediated transcriptional repression. Mol Oncol 2018; 12:1358-1373. [PMID: 29879296 PMCID: PMC6068344 DOI: 10.1002/1878-0261.12330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
The carboxyl‐terminal binding proteins (CtBP) are transcriptional corepressors that regulate the expression of multiple epithelial‐specific and pro‐apoptotic genes. Overexpression of CtBP occurs in many human cancers where they promote the epithelial‐to‐mesenchymal transition, stem cell‐like features, and cell survival, while knockdown of CtBP in tumor cells results in p53‐independent apoptosis. CtBPs are recruited to their target genes by binding to a conserved PXDLS peptide motif present in multiple DNA‐binding transcription factors. Disrupting the interaction between CtBP and its transcription factor partners may be a means of altering CtBP‐mediated transcriptional repression and a potential approach for cancer therapies. However, small molecules targeting protein–protein interactions have traditionally been difficult to identify. In this study, we took advantage of the fact that CtBP binds to a conserved peptide motif to explore the feasibility of using peptides containing the PXDLS motif fused to cell‐penetrating peptides (CPP) to inhibit CtBP function. We demonstrate that these peptides disrupt the ability of CtBP to interact with its protein partner, E1A, in an AlphaScreen assay. Moreover, these peptides can enter both lung carcinoma and melanoma cells, disrupt the interaction between CtBP and a transcription factor partner, and inhibit CtBP‐mediated transcriptional repression. Finally, the constitutive expression of one such peptide, Pep1‐E1A‐WT, in a melanoma cell line reverses CtBP‐mediated oncogenic phenotypes including proliferation, migration, and sphere formation and limits tumor growth in vivo. Together, our results suggest that CPP‐fused PXDLS‐containing peptides can potentially be developed into a research tool or therapeutic agent targeting CtBP‐mediated transcriptional events in various biological pathways.
Collapse
Affiliation(s)
- Melanie A Blevins
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Caiguo Zhang
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Lingdi Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Hong Li
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Xueni Li
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - David A Norris
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Mingxia Huang
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
21
|
Bellesis AG, Jecrois AM, Hayes JA, Schiffer CA, Royer WE. Assembly of human C-terminal binding protein (CtBP) into tetramers. J Biol Chem 2018; 293:9101-9112. [PMID: 29700119 DOI: 10.1074/jbc.ra118.002514] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/24/2018] [Indexed: 11/06/2022] Open
Abstract
C-terminal binding protein 1 (CtBP1) and CtBP2 are transcriptional coregulators that repress numerous cellular processes, such as apoptosis, by binding transcription factors and recruiting chromatin-remodeling enzymes to gene promoters. The NAD(H)-linked oligomerization of human CtBP is coupled to its co-transcriptional activity, which is implicated in cancer progression. However, the biologically relevant level of CtBP assembly has not been firmly established; nor has the stereochemical arrangement of the subunits above that of a dimer. Here, multi-angle light scattering (MALS) data established the NAD+- and NADH-dependent assembly of CtBP1 and CtBP2 into tetramers. An examination of subunit interactions within CtBP1 and CtBP2 crystal lattices revealed that both share a very similar tetrameric arrangement resulting from assembly of two dimeric pairs, with specific interactions probably being sensitive to NAD(H) binding. Creating a series of mutants of both CtBP1 and CtBP2, we tested the hypothesis that the crystallographically observed interdimer pairing stabilizes the solution tetramer. MALS data confirmed that these mutants disrupt both CtBP1 and CtBP2 tetramers, with the dimer generally remaining intact, providing the first stereochemical models for tetrameric assemblies of CtBP1 and CtBP2. The crystal structure of a subtle destabilizing mutant suggested that small structural perturbations of the hinge region linking the substrate- and NAD-binding domains are sufficient to weaken the CtBP1 tetramer. These results strongly suggest that the tetramer is important in CtBP function, and the series of CtBP mutants reported here can be used to investigate the physiological role of the tetramer.
Collapse
Affiliation(s)
- Andrew G Bellesis
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and.,the Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610
| | - Anne M Jecrois
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Janelle A Hayes
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Celia A Schiffer
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - William E Royer
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| |
Collapse
|
22
|
Dcona MM, Morris BL, Ellis KC, Grossman SR. CtBP- an emerging oncogene and novel small molecule drug target: Advances in the understanding of its oncogenic action and identification of therapeutic inhibitors. Cancer Biol Ther 2017; 18:379-391. [PMID: 28532298 PMCID: PMC5536941 DOI: 10.1080/15384047.2017.1323586] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
C-terminal Binding Proteins (CtBP) 1 and 2 are oncogenic transcriptional co-regulators overexpressed in many cancer types, with their expression level correlating to worse prognostic outcomes and aggressive tumor features. CtBP negatively regulates the expression of many tumor suppressor genes, while coactivating genes that promote proliferation, epithelial-mesenchymal transition, and cancer stem cell self-renewal activity. In light of this evidence, the development of novel inhibitors that mitigate CtBP function may provide clinically actionable therapeutic tools. This review article focuses on the progress made in understanding CtBP structure, role in tumor progression, and discovery and development of CtBP inhibitors that target CtBP's dehydrogenase activity and other functions, with a focus on the theory and rationale behind the designs of current inhibitors. We provide insight into the future development and use of rational combination therapy that may further augment the efficacy of CtBP inhibitors, specifically addressing metastasis and cancer stem cell populations within tumors.
Collapse
Affiliation(s)
- M Michael Dcona
- a Department of Internal Medicine , Virginia Commonwealth University , Richmond , VA , USA
| | - Benjamin L Morris
- b Department of Human and Molecular Genetics , Virginia Commonwealth University , Richmond , VA , USA
| | - Keith C Ellis
- c Department of Medicinal Chemistry , Virginia Commonwealth University , Richmond , VA , USA.,d Institute for Structural Biology , Drug Discovery and Development, Virginia Commonwealth University , Richmond , VA , USA.,e VCU Massey Cancer Center , Virginia Commonwealth University , Richmond , VA , USA
| | - Steven R Grossman
- a Department of Internal Medicine , Virginia Commonwealth University , Richmond , VA , USA.,b Department of Human and Molecular Genetics , Virginia Commonwealth University , Richmond , VA , USA.,d Institute for Structural Biology , Drug Discovery and Development, Virginia Commonwealth University , Richmond , VA , USA.,e VCU Massey Cancer Center , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
23
|
C-Terminal Binding Protein is Involved in Promoting to the Carcinogenesis of Human Glioma. Mol Neurobiol 2016; 54:6121-6132. [PMID: 27699603 DOI: 10.1007/s12035-016-0159-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022]
Abstract
C-terminal binding protein (CtBP) is responsible for regulating the pathogenesis of a lot of cancer types. However, whether CtBP1/2 is involved in regulating the growth and development of human glioma is still obscure. In the present study presented here, our results firstly reveal that CtBP1/2 deficiency, induced by siRNA interference, disrupts the functional integrity of the MRN complex that is responsible for DNA repair in human glioma cells. The dysfunction of the MRN complex further contributes to the up-regulation of ATM and Rad3-related kinase (ATR) and Chk1 signaling pathway, which inhibits cell cycle progression mediated by CDK2, preparing for the initiation of DNA repair. Under the condition of hypoxia, hypoxia-inducible factor-1α (HIF-1α) can be directly regulated by CDK2 on protein level, playing coordinately regulatory role in the carcinogenesis of human glioma cells. Overall, our findings reveal that CtBP1/2 is essential to promote to human glioma cell growth through maintaining the DNA stability regulated by the MRN/ATR/Chk1/CDK2/HIF-1α signaling pathway.
Collapse
|
24
|
Korwar S, Morris BL, Parikh HI, Coover RA, Doughty TW, Love IM, Hilbert BJ, Royer WE, Kellogg GE, Grossman SR, Ellis KC. Design, synthesis, and biological evaluation of substrate-competitive inhibitors of C-terminal Binding Protein (CtBP). Bioorg Med Chem 2016; 24:2707-15. [PMID: 27156192 DOI: 10.1016/j.bmc.2016.04.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/14/2016] [Accepted: 04/19/2016] [Indexed: 11/30/2022]
Abstract
C-terminal Binding Protein (CtBP) is a transcriptional co-regulator that downregulates the expression of many tumor-suppressor genes. Utilizing a crystal structure of CtBP with its substrate 4-methylthio-2-oxobutyric acid (MTOB) and NAD(+) as a guide, we have designed, synthesized, and tested a series of small molecule inhibitors of CtBP. From our first round of compounds, we identified 2-(hydroxyimino)-3-phenylpropanoic acid as a potent CtBP inhibitor (IC50=0.24μM). A structure-activity relationship study of this compound further identified the 4-chloro- (IC50=0.18μM) and 3-chloro- (IC50=0.17μM) analogues as additional potent CtBP inhibitors. Evaluation of the hydroxyimine analogues in a short-term cell growth/viability assay showed that the 4-chloro- and 3-chloro-analogues are 2-fold and 4-fold more potent, respectively, than the MTOB control. A functional cellular assay using a CtBP-specific transcriptional readout revealed that the 4-chloro- and 3-chloro-hydroxyimine analogues were able to block CtBP transcriptional repression activity. This data suggests that substrate-competitive inhibition of CtBP dehydrogenase activity is a potential mechanism to reactivate tumor-suppressor gene expression as a therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Sudha Korwar
- Department of Medicinal Chemistry, School of Pharmacy, The Institute for Structural Biology, Drug Discovery, and Development, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Benjamin L Morris
- Division of Hematology, Oncology, & Palliative Care, Department of Human and Molecular Genetics, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Hardik I Parikh
- Department of Medicinal Chemistry, School of Pharmacy, The Institute for Structural Biology, Drug Discovery, and Development, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Robert A Coover
- Department of Medicinal Chemistry, School of Pharmacy, The Institute for Structural Biology, Drug Discovery, and Development, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Tyler W Doughty
- Department of Molecular, Cell, & Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Ian M Love
- Division of Hematology, Oncology, & Palliative Care, Department of Human and Molecular Genetics, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Brendan J Hilbert
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - William E Royer
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Glen E Kellogg
- Department of Medicinal Chemistry, School of Pharmacy, The Institute for Structural Biology, Drug Discovery, and Development, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Steven R Grossman
- Division of Hematology, Oncology, & Palliative Care, Department of Human and Molecular Genetics, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States.
| | - Keith C Ellis
- Department of Medicinal Chemistry, School of Pharmacy, The Institute for Structural Biology, Drug Discovery, and Development, and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|