1
|
Approaches to Measuring the Activity of Major Lipolytic and Lipogenic Enzymes In Vitro and Ex Vivo. Int J Mol Sci 2022; 23:ijms231911093. [PMID: 36232405 PMCID: PMC9570359 DOI: 10.3390/ijms231911093] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Since the 1950s, one of the goals of adipose tissue research has been to determine lipolytic and lipogenic activity as the primary metabolic pathways affecting adipocyte health and size and thus representing potential therapeutic targets for the treatment of obesity and associated diseases. Nowadays, there is a relatively large number of methods to measure the activity of these pathways and involved enzymes, but their applicability to different biological samples is variable. Here, we review the characteristics of mean lipogenic and lipolytic enzymes, their inhibitors, and available methodologies for assessing their activity, and comment on the advantages and disadvantages of these methodologies and their applicability in vivo, ex vivo, and in vitro, i.e., in cells, organs and their respective extracts, with the emphasis on adipocytes and adipose tissue.
Collapse
|
2
|
O’Farrell M, Duke G, Crowley R, Buckley D, Martins EB, Bhattacharya D, Friedman SL, Kemble G. FASN inhibition targets multiple drivers of NASH by reducing steatosis, inflammation and fibrosis in preclinical models. Sci Rep 2022; 12:15661. [PMID: 36123383 PMCID: PMC9485253 DOI: 10.1038/s41598-022-19459-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 08/30/2022] [Indexed: 01/05/2023] Open
Abstract
Fatty acid synthase (FASN) is an attractive therapeutic target in non-alcoholic steatohepatitis (NASH) because it drives de novo lipogenesis and mediates pro-inflammatory and fibrogenic signaling. We therefore tested pharmacological inhibition of FASN in human cell culture and in three diet induced mouse models of NASH. Three related FASN inhibitors were used; TVB-3664, TVB-3166 and clinical stage TVB-2640 (denifanstat). In human primary liver microtissues, FASN inhibiton (FASNi) decreased triglyceride (TG) content, consistent with direct anti-steatotic activity. In human hepatic stellate cells, FASNi reduced markers of fibrosis including collagen1α (COL1α1) and α-smooth muscle actin (αSMA). In CD4+ T cells exposed to NASH-related cytokines, FASNi decreased production of Th17 cells, and reduced IL-1β release in LPS-stimulated PBMCs. In mice with diet induced NASH l, FASNi prevented development of hepatic steatosis and fibrosis, and reduced circulating IL-1β. In mice with established diet-induced NASH, FASNi reduced NAFLD activity score, fibrosis score, ALT and TG levels. In the CCl4-induced FAT-NASH mouse model, FASN inhibition decreased hepatic fibrosis and fibrosis markers, and development of hepatocellular carcinoma (HCC) tumors by 85%. These results demonstrate that FASN inhibition attenuates inflammatory and fibrotic drivers of NASH by direct inhibition of immune and stellate cells, beyond decreasing fat accumulation in hepatocytes. FASN inhibition therefore provides an opportunity to target three key hallmarks of NASH.
Collapse
Affiliation(s)
- Marie O’Farrell
- Sagimet Biosciences Inc., 155 Bovet Rd, San Mateo, CA 94402 USA
| | - Greg Duke
- Sagimet Biosciences Inc., 155 Bovet Rd, San Mateo, CA 94402 USA
| | - Richard Crowley
- Sagimet Biosciences Inc., 155 Bovet Rd, San Mateo, CA 94402 USA
| | - Douglas Buckley
- Sagimet Biosciences Inc., 155 Bovet Rd, San Mateo, CA 94402 USA
| | | | - Dipankar Bhattacharya
- grid.59734.3c0000 0001 0670 2351Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Scott L. Friedman
- grid.59734.3c0000 0001 0670 2351Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - George Kemble
- Sagimet Biosciences Inc., 155 Bovet Rd, San Mateo, CA 94402 USA
| |
Collapse
|
3
|
Chen G, Harwood JL, Lemieux MJ, Stone SJ, Weselake RJ. Acyl-CoA:diacylglycerol acyltransferase: Properties, physiological roles, metabolic engineering and intentional control. Prog Lipid Res 2022; 88:101181. [PMID: 35820474 DOI: 10.1016/j.plipres.2022.101181] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022]
Abstract
Acyl-CoA:diacylglycerol acyltransferase (DGAT, EC 2.3.1.20) catalyzes the last reaction in the acyl-CoA-dependent biosynthesis of triacylglycerol (TAG). DGAT activity resides mainly in membrane-bound DGAT1 and DGAT2 in eukaryotes and bifunctional wax ester synthase-diacylglycerol acyltransferase (WSD) in bacteria, which are all membrane-bound proteins but exhibit no sequence homology to each other. Recent studies also identified other DGAT enzymes such as the soluble DGAT3 and diacylglycerol acetyltransferase (EaDAcT), as well as enzymes with DGAT activities including defective in cuticular ridges (DCR) and steryl and phytyl ester synthases (PESs). This review comprehensively discusses research advances on DGATs in prokaryotes and eukaryotes with a focus on their biochemical properties, physiological roles, and biotechnological and therapeutic applications. The review begins with a discussion of DGAT assay methods, followed by a systematic discussion of TAG biosynthesis and the properties and physiological role of DGATs. Thereafter, the review discusses the three-dimensional structure and insights into mechanism of action of human DGAT1, and the modeled DGAT1 from Brassica napus. The review then examines metabolic engineering strategies involving manipulation of DGAT, followed by a discussion of its therapeutic applications. DGAT in relation to improvement of livestock traits is also discussed along with DGATs in various other eukaryotic organisms.
Collapse
Affiliation(s)
- Guanqun Chen
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada.
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Membrane Protein Disease Research Group, Edmonton T6G 2H7, Canada
| | - Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| | - Randall J Weselake
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada
| |
Collapse
|
4
|
Yue Y, Yang WS, Zhang L, Liu CP, Xu RM. Topography of histone H3-H4 interaction with the Hat1-Hat2 acetyltransferase complex. Genes Dev 2022; 36:408-413. [PMID: 35393344 PMCID: PMC9067401 DOI: 10.1101/gad.349099.121] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/14/2022] [Indexed: 01/04/2023]
Abstract
In this study, Yue et al. present the structure of the Hat1–Hat2 acetyltransferase complex bound to Asf1–H3–H4, which shows that the core domains of H3 and H4 are involved in binding Hat1 and Hat2, and the N-terminal tail of H3 makes extensive interaction with Hat2. These findings extend our knowledge of histone–protein interaction and implicate a function of Hat2/RbAp46/48 in the passing of histones between chaperones. Chaperones influence histone conformation and intermolecular interaction in multiprotein complexes, and the structures obtained with full-length histones often provide more accurate and comprehensive views. Here, our structure of the Hat1–Hat2 acetyltransferase complex bound to Asf1–H3–H4 shows that the core domains of H3 and H4 are involved in binding Hat1 and Hat2, and the N-terminal tail of H3 makes extensive interaction with Hat2. These findings expand the knowledge about histone–protein interaction and implicate a function of Hat2/RbAp46/48, which is a versatile histone chaperone found in many chromatin-associated complexes, in the passing of histones between chaperones.
Collapse
Affiliation(s)
- Ye Yue
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Si Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chao-Pei Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui-Ming Xu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Nešuta O, Thomas AG, Alt J, Hin N, Neužilová A, Long S, Tsukamoto T, Rojas C, Wei H, Slusher BS. High Throughput Screening Cascade To Identify Human Aspartate N-Acetyltransferase (ANAT) Inhibitors for Canavan Disease. ACS Chem Neurosci 2021; 12:3445-3455. [PMID: 34477360 DOI: 10.1021/acschemneuro.1c00455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Canavan disease (CD) is a progressive, fatal neurological disorder that begins in infancy resulting from a mutation in aspartoacyclase (ASPA), an enzyme that catalyzes the deacetylation of N-acetyl aspartate (NAA) into acetate and aspartate. Increased NAA levels in the brains of affected children are one of the hallmarks of CD. Interestingly, genetic deletion of N-acetyltransferase-8-like (NAT8L), which encodes aspartate N-aceyltransferase (ANAT), an enzyme responsible for the synthesis of NAA from l-aspartate and acetyl-CoA, leads to normalization of NAA levels and improvement of symptoms in several genetically engineered mouse models of CD. Therefore, pharmacological inhibition of ANAT presents a promising therapeutic strategy for treating CD. Currently, however, there are no clinically viable ANAT inhibitors. Herein we describe the development of fluorescence-based high throughput screening (HTS) and radioactive-based orthogonal assays using recombinant human ANAT expressed in E. coli. In the fluorescence-based assay, ANAT activity was linear with respect to time of incubation up to 30 min and protein concentration up to 97.5 ng/μL with Km values for l-aspartate and acetyl-CoA of 237 μM and 11 μM, respectively. Using this optimized assay, we conducted a pilot screening of a 10 000-compound library. Hits from the fluorescence-based assay were subjected to an orthogonal radioactive-based assay using L-[U-14C] aspartate as a substrate. Two compounds were confirmed to have dose-dependent inhibition in both assays. Inhibitory kinetics studies of the most potent compound revealed an uncompetitive inhibitory mechanism with respect to l-aspartate and a noncompetitive inhibitory mechanism against acetyl-CoA. The screening cascade developed herein will enable large-scale compound library screening to identify novel ANAT inhibitors as leads for further medicinal chemistry optimization.
Collapse
Affiliation(s)
- Ondřej Nešuta
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Niyada Hin
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Anna Neužilová
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Shunyou Long
- ChemBioCORE, High Throughput Screening Facility, Johns Hopkins University, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Huijun Wei
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| |
Collapse
|
6
|
Karatas H, Akbarzadeh M, Adihou H, Hahne G, Pobbati AV, Yihui Ng E, Guéret SM, Sievers S, Pahl A, Metz M, Zinken S, Dötsch L, Nowak C, Thavam S, Friese A, Kang C, Hong W, Waldmann H. Discovery of Covalent Inhibitors Targeting the Transcriptional Enhanced Associate Domain Central Pocket. J Med Chem 2020; 63:11972-11989. [PMID: 32907324 PMCID: PMC7586386 DOI: 10.1021/acs.jmedchem.0c01275] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Transcriptional enhanced associate
domain (TEAD) transcription
factors together with coactivators and corepressors modulate the expression
of genes that regulate fundamental processes, such as organogenesis
and cell growth, and elevated TEAD activity is associated with tumorigenesis.
Hence, novel modulators of TEAD and methods for their identification
are in high demand. We describe the development of a new “thiol
conjugation assay” for identification of novel small molecules
that bind to the TEAD central pocket. The assay monitors prevention
of covalent binding of a fluorescence turn-on probe to a cysteine
in the central pocket by small molecules. Screening of a collection
of compounds revealed kojic acid analogues as TEAD inhibitors, which
covalently target the cysteine in the central pocket, block the interaction
with coactivator yes-associated protein with nanomolar apparent IC50 values, and reduce TEAD target gene expression. This methodology
promises to enable new medicinal chemistry programs aimed at the modulation
of TEAD activity.
Collapse
Affiliation(s)
- Hacer Karatas
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Mohammad Akbarzadeh
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Hélène Adihou
- Department of Chemical Biology, AstraZeneca-Max Planck Institute Satellite Unit, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.,Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, SE-431 83 Gothenburg, Sweden
| | - Gernot Hahne
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Ajaybabu V Pobbati
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, 138673 Singapore, Singapore
| | - Elizabeth Yihui Ng
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), 10 Biopolis Road, Chromos, #05-01, 138670, Singapore
| | - Stéphanie M Guéret
- Department of Chemical Biology, AstraZeneca-Max Planck Institute Satellite Unit, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.,Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, SE-431 83 Gothenburg, Sweden
| | - Sonja Sievers
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Axel Pahl
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Malte Metz
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Sarah Zinken
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| | - Lara Dötsch
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| | - Christine Nowak
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Sasikala Thavam
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - Alexandra Friese
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany
| | - CongBao Kang
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), 10 Biopolis Road, Chromos, #05-01, 138670, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, 138673 Singapore, Singapore
| | - Herbert Waldmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, 44227 Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| |
Collapse
|
7
|
Valastyan JS, Tota MR, Taylor IR, Stergioula V, Hone GAB, Smith CD, Henke BR, Carson KG, Bassler BL. Discovery of PqsE Thioesterase Inhibitors for Pseudomonas aeruginosa Using DNA-Encoded Small Molecule Library Screening. ACS Chem Biol 2020; 15:446-456. [PMID: 31880429 DOI: 10.1021/acschembio.9b00905] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pseudomonas aeruginosa is a leading cause of hospital-acquired infections in the United States. PqsE, a thioesterase enzyme, is vital for virulence of P. aeruginosa, making PqsE an attractive target for inhibition. Neither the substrate nor the product of PqsE catalysis has been identified. A library of 550 million DNA-encoded drug-like small molecules was screened for those that bind to the purified PqsE protein. The structures of the bound molecules were identified by high throughput sequencing of the attached DNA barcodes. Putative PqsE binders with the strongest affinity features were examined for inhibition of PqsE thioesterase activity in vitro. The most potent inhibitors were resynthesized off DNA and examined for the ability to alter PqsE thermal melting and for PqsE thioesterase inhibition. Here, we report the synthesis, biological activity, mechanism of action, and early structure-activity relationships of a series of 2-(phenylcarbamoyl)benzoic acids that noncompetitively inhibit PqsE. A small set of analogs designed to probe initial structure-activity relationships showed increases in potency relative to the original hits, the best of which has an IC50 = 5 μM. Compound refinement is required to assess their in vivo activities as the current compounds do not accumulate in the P. aeruginosa cytosol. Our strategy validates DNA-encoded compound library screening as a rapid and effective method to identify catalytic inhibitors of the PqsE protein, and more generally, for discovering binders to bacterial proteins revealed by genetic screening to have crucial in vivo activities but whose biological functions have not been well-defined.
Collapse
Affiliation(s)
- Julie S. Valastyan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| | - Michael R. Tota
- Macroceutics Incorporated, (now HotSpot Therapeutics), Monmouth Junction, New Jersey 08852, United States
| | - Isabelle R. Taylor
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Vasiliki Stergioula
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Graham A. B. Hone
- Macroceutics Incorporated, (now HotSpot Therapeutics), Monmouth Junction, New Jersey 08852, United States
| | - Chari D. Smith
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Brad R. Henke
- Opti-Mol Consulting, LLC, Cary, North Carolina 27513, United States
| | - Kenneth G. Carson
- Macroceutics Incorporated, (now HotSpot Therapeutics), Monmouth Junction, New Jersey 08852, United States
| | - Bonnie L. Bassler
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| |
Collapse
|
8
|
Nguyen TH, Waldrop GL, Gilman SD. Capillary electrophoretic assay of human acetyl-coenzyme A carboxylase 2. Electrophoresis 2019; 40:1558-1564. [PMID: 30828828 DOI: 10.1002/elps.201800514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 11/09/2022]
Abstract
Human acetyl-coenzyme A carboxylase 2 catalyzes the carboxylation of acetyl coenzyme A to form malonyl coenzyme A, along with the conversion of magnesium-adenosine triphosphate complex to magnesium-adenosine diphosphate complex. A simple off-column capillary electrophoresis assay for human acetyl-coenzyme A carboxylase 2 was developed based on the separation of magnesium-adenosine triphosphate complex, magnesium-adenosine diphosphate complex, acetyl coenzyme A and malonyl coenzyme A with detection by ultraviolet absorption at 256 nm. When Mg2+ was absent from the separation buffer, the zones due to magnesium-adenosine triphosphate complex and magnesium-adenosine diphosphate complex both split and migrated as two separate peaks. With Mg2+ added to the separation buffer, magnesium-adenosine triphosphate complex and magnesium-adenosine diphosphate complex produced single peaks, and the reproducibility of peak shape and area improved for human acetyl-coenzyme A carboxylase 2 assay components. The final separation buffer used was 30.0 mM HEPES, 3.0 mM MgCl2 , 2.5 mM KHCO3 , and 2.5 mM potassium citrate at pH 7.50. The same buffer was used for the enzyme-catalyzed reaction (off-column). Inhibition of human acetyl-coenzyme A carboxylase 2 by CP-640186, a known inhibitor, was detected using the capillary electrophoresis assay.
Collapse
Affiliation(s)
- Thu H Nguyen
- Department of Chemistry, Louisiana State University, Baton Rouge, LA, USA
| | - Grover L Waldrop
- Division of Biochemistry and Molecular Biology, Louisiana State University, Baton Rouge, LA, USA
| | - S Douglass Gilman
- Department of Chemistry, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
9
|
Ling X, Zhang S, Liu Y, Bai M. Light-activatable cannabinoid prodrug for combined and target-specific photodynamic and cannabinoid therapy. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-9. [PMID: 30334393 DOI: 10.1117/1.jbo.23.10.108001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/19/2018] [Indexed: 06/08/2023]
Abstract
Cannabinoids are emerging as promising antitumor drugs. However, complete tumor eradication solely by cannabinoid therapy remains challenging. In this study, we developed a far-red light activatable cannabinoid prodrug, which allows for tumor-specific and combinatory cannabinoid and photodynamic therapy. This prodrug consists of a phthalocyanine photosensitizer (PS), reactive oxygen species (ROS)-sensitive linker, and cannabinoid. It targets the type-2 cannabinoid receptor (CB2R) overexpressed in various types of cancers. Upon the 690-nm light irradiation, the PS produces cytotoxic ROS, which simultaneously cleaves the ROS-sensitive linker and subsequently releases the cannabinoid drug. We found that this unique multifunctional prodrug design offered dramatically improved therapeutic efficacy, and therefore provided a new strategy for targeted, controlled, and effective antitumor cannabinoid therapy.
Collapse
Affiliation(s)
- Xiaoxi Ling
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States
| | - Shaojuan Zhang
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States
| | - Yang Liu
- Vanderbilt University Institute of Imaging Sciences, Nashville, Tennessee, United States
| | - Mingfeng Bai
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States
- University of Pittsburgh, Department of Medicine, Pittsburgh Pennsylvania, United States
- University of Pittsburgh, Department of Bioengineering, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
10
|
Renault K, Fredy JW, Renard PY, Sabot C. Covalent Modification of Biomolecules through Maleimide-Based Labeling Strategies. Bioconjug Chem 2018; 29:2497-2513. [PMID: 29954169 DOI: 10.1021/acs.bioconjchem.8b00252] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Since their first use in bioconjugation more than 50 years ago, maleimides have become privileged chemical partners for the site-selective modification of proteins via thio-Michael addition of biothiols and, to a lesser extent, via Diels-Alder (DA) reactions with biocompatible dienes. Prominent examples include immunotoxins and marketed maleimide-based antibody-drug conjugates (ADCs) such as Adcetris, which are used in cancer therapies. Among the key factors in the success of these groups is the availability of several maleimides that can be N-functionalized by fluorophores, affinity tags, spin labels, and pharmacophores, as well as their unique reactivities in terms of selectivity and kinetics. However, maleimide conjugate reactions have long been considered irreversible, and only recently have systematic studies regarding their reversibility and stability toward hydrolysis been reported. This review provides an overview of the diverse applications for maleimides in bioconjugation, highlighting their strengths and weaknesses, which are being overcome by recent strategies. Finally, the fluorescence quenching ability of maleimides was leveraged for the preparation of fluorogenic probes, which are mainly used for the specific detection of thiol analytes. A summary of the reported structures, their photophysical features, and their relative efficiencies is discussed in the last part of the review.
Collapse
Affiliation(s)
- Kévin Renault
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| | - Jean Wilfried Fredy
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| | - Pierre-Yves Renard
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| | - Cyrille Sabot
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| |
Collapse
|
11
|
Jackson PA, Widen JC, Harki DA, Brummond KM. Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition Reactions. J Med Chem 2017; 60:839-885. [PMID: 27996267 PMCID: PMC5308545 DOI: 10.1021/acs.jmedchem.6b00788] [Citation(s) in RCA: 332] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although Michael acceptors display a potent and broad spectrum of bioactivity, they have largely been ignored in drug discovery because of their presumed indiscriminate reactivity. As such, a dearth of information exists relevant to the thiol reactivity of natural products and their analogues possessing this moiety. In the midst of recently approved acrylamide-containing drugs, it is clear that a good understanding of the hetero-Michael addition reaction and the relative reactivities of biological thiols with Michael acceptors under physiological conditions is needed for the design and use of these compounds as biological tools and potential therapeutics. This Perspective provides information that will contribute to this understanding, such as kinetics of thiol addition reactions, bioactivities, as well as steric and electronic factors that influence the electrophilicity and reversibility of Michael acceptors. This Perspective is focused on α,β-unsaturated carbonyls given their preponderance in bioactive natural products.
Collapse
Affiliation(s)
- Paul A. Jackson
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - John C. Widen
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Daniel A. Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kay M. Brummond
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
12
|
Goosen R, Strauss E. Simultaneous quantification of coenzyme A and its salvage pathway intermediates in in vitro and whole cell-sourced samples. RSC Adv 2017. [DOI: 10.1039/c7ra00192d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
A method for the quantitative analysis of CoA and its thiolated precursors was developed, addressing the analytical shortcomings of previous methods. Its utility was showcased by analysis ofin vitroenzyme reactions and samples extracted from various bacterial strains.
Collapse
Affiliation(s)
- R. Goosen
- Department of Biochemistry
- Stellenbosch University
- Stellenbosch
- South Africa
| | - E. Strauss
- Department of Biochemistry
- Stellenbosch University
- Stellenbosch
- South Africa
| |
Collapse
|
13
|
Takamiya M, Sakurai M, Teranishi F, Ikeda T, Kamiyama T, Asai A. Lead discovery for mammalian elongation of long chain fatty acids family 6 using a combination of high-throughput fluorescent-based assay and RapidFire mass spectrometry assay. Biochem Biophys Res Commun 2016; 480:721-726. [DOI: 10.1016/j.bbrc.2016.10.103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022]
|
14
|
Marcella AM, Barb AW. A rapid fluorometric assay for the S-malonyltransacylase FabD and other sulfhydryl utilizing enzymes. J Biol Methods 2016; 3. [PMID: 27642613 PMCID: PMC5023282 DOI: 10.14440/jbm.2016.144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The development of biorenewable chemicals will support green chemistry initiatives and supplement the catalog of starting materials available to the chemical industry. Bacterial fatty acid biosynthesis is being pursued as a source of protein catalysts to synthesize novel reduced carbon molecules in fermentation systems. The availability of methods to measure enzyme catalysis for native and engineered enzymes from this pathway remains a bottleneck because a simple quantitative enzyme assay for numerous enzymes does not exist. Here we present two variations of a fluorescence assay that is readily extendable to high-throughput screening and is appropriate for thiol consuming and generating enzymes including the E. coli enzymes malonyl-coenzyme A transacylase (FabD) and keto-acylsynthase III (FabH). We measured KM values of 60 ± 20 µM (acetyl-CoA) and 20 ± 10 µM (malonyl-ACP) and a kcat of 7.4–9.0 s-1 with FabH. Assays of FabD included a precipitation step to remove the thiol-containing substrate holo-ACP from the reaction product coenzyme-A to estimate reaction rates. Analysis of initial velocity measurements revealed KM values of 60 ± 20 µM (malonyl-CoA) and 40 ± 10 µM (holo-ACP) and a kcat of 2100–2600 s-1 for the FabD enzyme. Our data show similar results when compared to existing radioactive and continuous coupled assays in terms of sensitivity while providing the benefit of simplicity, scalability and repeatability. Fluorescence detection also eliminates the need for radioactive substrates traditionally used to assay these enzymes.
Collapse
Affiliation(s)
- Aaron M Marcella
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Molecular Biology Building, Room 4210, 2437 Pammel Drive, Iowa State University, Ames, IA 50011, USA
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Molecular Biology Building, Room 4210, 2437 Pammel Drive, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
15
|
Dahlin JL, Nissink JWM, Francis S, Strasser JM, John K, Zhang Z, Walters MA. Post-HTS case report and structural alert: Promiscuous 4-aroyl-1,5-disubstituted-3-hydroxy-2H-pyrrol-2-one actives verified by ALARM NMR. Bioorg Med Chem Lett 2015; 25:4740-4752. [PMID: 26318992 PMCID: PMC6002837 DOI: 10.1016/j.bmcl.2015.08.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/29/2015] [Accepted: 08/06/2015] [Indexed: 12/19/2022]
Abstract
Despite its wide use, not every high-throughput screen (HTS) yields chemical matter suitable for drug development campaigns, and seldom are 'go/no-go' decisions in drug discovery described in detail. This case report describes the follow-up of a 4-aroyl-1,5-disubstituted-3-hydroxy-2H-pyrrol-2-one active from a cell-free HTS to identify small-molecule inhibitors of Rtt109-catalyzed histone acetylation. While this compound and structural analogs inhibited Rtt109-catalyzed histone acetylation in vitro, further work on this series was halted after several risk mitigation strategies were performed. Compounds with this chemotype had a poor structure-activity relationship, exhibited poor selectivity among other histone acetyltransferases, and tested positive in a β-lactamase counter-screen for chemical aggregates. Furthermore, ALARM NMR demonstrated compounds with this chemotype grossly perturbed the conformation of the La protein. In retrospect, this chemotype was flagged as a 'frequent hitter' in an analysis of a large corporate screening deck, yet similar compounds have been published as screening actives or chemical probes versus unrelated biological targets. This report-including the decision-making process behind the 'no-go' decision-should be informative for groups engaged in post-HTS triage and highlight the importance of considering physicochemical properties in early drug discovery.
Collapse
Affiliation(s)
- Jayme L Dahlin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Medical Scientist Training Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | - Subhashree Francis
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN 55414, USA
| | - Jessica M Strasser
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN 55414, USA
| | - Kristen John
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN 55414, USA
| | - Zhiguo Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Michael A Walters
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN 55414, USA.
| |
Collapse
|
16
|
Horiuchi KY. Challenges in profiling and lead optimization of drug discovery for methyltransferases. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 18:62-68. [PMID: 26723894 DOI: 10.1016/j.ddtec.2015.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 06/05/2023]
Abstract
The importance of epigenetics in the initiation and progression of disease has attracted many investigators to incorporate this novel and exciting field in drug development. Protein methyltransferases are one of the target classes which have gained attention as potential therapeutic targets after promising results of inhibitors for EZH2 and DOT1L in clinical trials. There are many technologies developed in order to find small molecule inhibitors for protein methyltransferases. However, in contrast to high throughput screening, profiling against different methyltransferases is challenging since each enzyme has a different substrate preference so that it is hard to profile in one assay format. Here, different technologies for methyltransferase assays will be overviewed, and the advantages and disadvantages of each will be discussed.
Collapse
Affiliation(s)
- Kurumi Y Horiuchi
- Reaction Biology Corporation, One Great Valley Parkway, Suite 2, Malvern, PA 19355, USA.
| |
Collapse
|
17
|
Song XS, Zhang J, Chen X, Palyha O, Chung C, Sonatore LM, Wilsie L, Stout S, McLaren DG, Taggart A, Imbriglio JE, Pinto S, Garcia-Calvo M, Addona GH. Identification of DGAT2 Inhibitors Using Mass Spectrometry. ACTA ACUST UNITED AC 2015; 21:117-26. [DOI: 10.1177/1087057115607463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/23/2015] [Indexed: 11/17/2022]
Abstract
Mass spectrometry offers significant advantages over other detection technologies in the areas of hit finding, hit validation, and medicinal chemistry compound optimization. The foremost obvious advantage is the ability to directly measure enzymatic product formation. In addition, the inherent sensitivity of the liquid chromatography/mass spectrometry (LC/MS) approach allows the execution of enzymatic assays at substrate concentrations typically at or below substrate Km. Another advantage of the LC/MS approach is the ability to assay impure enzyme systems that would otherwise be difficult to prosecute with traditional labeled methods. This approach was used to identify inhibitors of diacylglycerol O-acyltransferase-2 (DGAT2), a transmembrane enzyme involved in the triglyceride (TG) production pathway. The LC/MS approach was employed because of its increased assay window (compared with control membranes) of more than sevenfold compared with less than twofold with a conventional fluorogenic assay. The ability to generate thousands of dose-dependent IC50 data was made possible by the use of a staggered parallel LC/MS system with fast elution gradients. From the hit-deconvolution efforts, several structural scaffold series were identified that inhibit DGAT2 activity. Additional profiling of one chemotype in particular identified two promising reversible and selective compounds (compound 15 and compound 16) that effectively inhibit TG production in mouse primary hepatocytes.
Collapse
Affiliation(s)
- Xuelei S. Song
- Department of Pharmacology, Merck Research Laboratories, Boston, MA, USA
| | - Jiaping Zhang
- Department of Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Xun Chen
- Department of Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Oksana Palyha
- Atherosclerosis, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Christine Chung
- Department of Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Lisa M. Sonatore
- Department of Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Larissa Wilsie
- Atherosclerosis, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Steven Stout
- Department of Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - David G. McLaren
- Department of Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Andrew Taggart
- Atherosclerosis, Merck Research Laboratories, Kenilworth, NJ, USA
| | | | - Shirly Pinto
- Atherosclerosis, Merck Research Laboratories, Kenilworth, NJ, USA
| | | | - George H. Addona
- Department of Pharmacology, Merck Research Laboratories, Boston, MA, USA
| |
Collapse
|
18
|
Ung T, Mason JL, Robinson RG, Spais CM, Ator MA, Angeles TS. A Cellular Assay for Inhibitors of the Fatty Acid Biosynthetic Pathway Using Scintillating Microplates. Assay Drug Dev Technol 2015; 13:285-92. [PMID: 26125659 DOI: 10.1089/adt.2015.644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A simplified method for monitoring the incorporation of radiolabeled acetate into lipids in a cellular system is described. The assay eliminates the commonly employed labor-intensive organic extraction step by plating the cells in 96-well tissue culture-treated ScintiPlates(®) that enable direct measurement of radiolabeled cell membrane-embedded lipids. Since the scintillant is entrenched in the plates, radioactivity in close proximity to the scintillant is measured without the need for liquid scintillation cocktail. The utility of this method for evaluating inhibitors of the de novo fatty acid synthetic pathway is demonstrated here with fatty acid synthase (FASN). Due to the upregulation of FASN activity in many tumor types, development of inhibitors to block the FASN activity in cells shows promise as an attractive and tractable approach for therapeutic intervention.
Collapse
Affiliation(s)
- Thao Ung
- 1 Lead Discovery and Profiling, Teva Branded Pharmaceutical Products R&D, Inc. , West Chester, Pennsylvania
| | - Jennifer L Mason
- 1 Lead Discovery and Profiling, Teva Branded Pharmaceutical Products R&D, Inc. , West Chester, Pennsylvania
| | - Ron G Robinson
- 1 Lead Discovery and Profiling, Teva Branded Pharmaceutical Products R&D, Inc. , West Chester, Pennsylvania
| | - Chrysanthe M Spais
- 2 Global Bioassay and Technology, Teva Branded Pharmaceutical Products R&D, Inc. , West Chester, Pennsylvania
| | - Mark A Ator
- 1 Lead Discovery and Profiling, Teva Branded Pharmaceutical Products R&D, Inc. , West Chester, Pennsylvania
| | - Thelma S Angeles
- 1 Lead Discovery and Profiling, Teva Branded Pharmaceutical Products R&D, Inc. , West Chester, Pennsylvania
| |
Collapse
|
19
|
Marcella AM, Jing F, Barb AW. Preparation of holo- and malonyl-[acyl-carrier-protein] in a manner suitable for analog development. Protein Expr Purif 2015; 115:39-45. [PMID: 26008118 DOI: 10.1016/j.pep.2015.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 12/31/2022]
Abstract
The fatty acid biosynthetic pathway generates highly reduced carbon based molecules. For this reason fatty acid synthesis is a target of pathway engineering to produce novel specialty or commodity chemicals using renewable techniques to supplant molecules currently derived from petroleum. Malonyl-[acyl carrier protein] (malonyl-ACP) is a key metabolite in the fatty acid pathway and donates two carbon units to the growing fatty acid chain during each step of biosynthesis. Attempts to test engineered fatty acid biosynthesis enzymes in vitro will require malonyl-ACP or malonyl-ACP analogs. Malonyl-ACP is challenging to prepare due to the instability of the carboxylate leaving group and the multiple steps of post-translational modification required to activate ACP. Here we report the expression and purification of holo- and malonyl-ACP from Escherichia coli with high yields (>15 mg per L of expression). The malonyl-ACP is efficiently recognized by the E. coli keto-acyl synthase enzyme, FabH. A FabH assay using malonyl-ACP and a coumarin-based fluorescent reagent is described that provides a high throughput alternative to reported radioactive assays.
Collapse
Affiliation(s)
- Aaron M Marcella
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, United States
| | - Fuyuan Jing
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, United States
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
20
|
Dahlin JL, Nissink JWM, Strasser JM, Francis S, Higgins L, Zhou H, Zhang Z, Walters MA. PAINS in the assay: chemical mechanisms of assay interference and promiscuous enzymatic inhibition observed during a sulfhydryl-scavenging HTS. J Med Chem 2015; 58:2091-113. [PMID: 25634295 PMCID: PMC4360378 DOI: 10.1021/jm5019093] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significant resources in early drug discovery are spent unknowingly pursuing artifacts and promiscuous bioactive compounds, while understanding the chemical basis for these adverse behaviors often goes unexplored in pursuit of lead compounds. Nearly all the hits from our recent sulfhydryl-scavenging high-throughput screen (HTS) targeting the histone acetyltransferase Rtt109 were such compounds. Herein, we characterize the chemical basis for assay interference and promiscuous enzymatic inhibition for several prominent chemotypes identified by this HTS, including some pan-assay interference compounds (PAINS). Protein mass spectrometry and ALARM NMR confirmed these compounds react covalently with cysteines on multiple proteins. Unfortunately, compounds containing these chemotypes have been published as screening actives in reputable journals and even touted as chemical probes or preclinical candidates. Our detailed characterization and identification of such thiol-reactive chemotypes should accelerate triage of nuisance compounds, guide screening library design, and prevent follow-up on undesirable chemical matter.
Collapse
Affiliation(s)
- Jayme L Dahlin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine , Rochester, Minnesota 55905, United States
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Tarui M, Shindou H, Kumagai K, Morimoto R, Harayama T, Hashidate T, Kojima H, Okabe T, Nagano T, Nagase T, Shimizu T. Selective inhibitors of a PAF biosynthetic enzyme lysophosphatidylcholine acyltransferase 2. J Lipid Res 2014; 55:1386-96. [PMID: 24850807 PMCID: PMC4076079 DOI: 10.1194/jlr.m049205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Indexed: 01/09/2023] Open
Abstract
Platelet-activating factor (PAF) is a potent pro-inflammatory phospholipid mediator. In response to extracellular stimuli, PAF is rapidly biosynthesized by lyso-PAF acetyltransferase (lyso-PAFAT). Previously, we identified two types of lyso-PAFATs: lysophosphatidylcholine acyltransferase (LPCAT)1, mostly expressed in the lungs where it produces PAF and dipalmitoyl-phosphatidylcholine essential for respiration, and LPCAT2, which biosynthesizes PAF and phosphatidylcholine (PC) in the inflammatory cells. Under inflammatory conditions, LPCAT2, but not LPCAT1, is activated and upregulated to produce PAF. Thus, it is important to develop inhibitors specific for LPCAT2 in order to ameliorate PAF-related inflammatory diseases. Here, we report the first identification of LPCAT2-specific inhibitors, N-phenylmaleimide derivatives, selected from a 174,000-compound library using fluorescence-based high-throughput screening followed by the evaluation of the effects on LPCAT1 and LPCAT2 activities, cell viability, and cellular PAF production. Selected compounds competed with acetyl-CoA for the inhibition of LPCAT2 lyso-PAFAT activity and suppressed PAF biosynthesis in mouse peritoneal macrophages stimulated with a calcium ionophore. These compounds had low inhibitory effects on LPCAT1 activity, indicating that adverse effects on respiratory functions may be avoided. The identified compounds and their derivatives will contribute to the development of novel drugs for PAF-related diseases and facilitate the analysis of LPCAT2 functions in phospholipid metabolism in vivo.
Collapse
Affiliation(s)
- Megumi Tarui
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan Department of Respiratory Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Kazuo Kumagai
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ryo Morimoto
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeshi Harayama
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Tomomi Hashidate
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hirotatsu Kojima
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayoshi Okabe
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tetsuo Nagano
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takao Shimizu
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
22
|
Dahlin JL, Sinville R, Solberg J, Zhou H, Han J, Francis S, Strasser JM, John K, Hook DJ, Walters MA, Zhang Z. A cell-free fluorometric high-throughput screen for inhibitors of Rtt109-catalyzed histone acetylation. PLoS One 2013; 8:e78877. [PMID: 24260132 PMCID: PMC3832525 DOI: 10.1371/journal.pone.0078877] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 09/17/2013] [Indexed: 11/19/2022] Open
Abstract
The lysine acetyltransferase (KAT) Rtt109 forms a complex with Vps75 and catalyzes the acetylation of histone H3 lysine 56 (H3K56ac) in the Asf1-H3-H4 complex. Rtt109 and H3K56ac are vital for replication-coupled nucleosome assembly and genotoxic resistance in yeast and pathogenic fungal species such as Candida albicans. Remarkably, sequence homologs of Rtt109 are absent in humans. Therefore, inhibitors of Rtt109 are hypothesized as potential and minimally toxic antifungal agents. Herein, we report the development and optimization of a cell-free fluorometric high-throughput screen (HTS) for small-molecule inhibitors of Rtt109-catalyzed histone acetylation. The KAT component of the assay consists of the yeast Rtt109-Vps75 complex, while the histone substrate complex consists of full-length Drosophila histone H3-H4 bound to yeast Asf1. Duplicated assay runs of the LOPAC demonstrated day-to-day and plate-to-plate reproducibility. Approximately 225,000 compounds were assayed in a 384-well plate format with an average Z' factor of 0.71. Based on a 3σ cut-off criterion, 1,587 actives (0.7%) were identified in the primary screen. The assay method is capable of identifying previously reported KAT inhibitors such as garcinol. We also observed several prominent active classes of pan-assay interference compounds such as Mannich bases, catechols and p-hydroxyarylsulfonamides. The majority of the primary active compounds showed assay signal interference, though most assay artifacts can be efficiently removed by a series of straightforward counter-screens and orthogonal assays. Post-HTS triage demonstrated a comparatively small number of confirmed actives with IC50 values in the low micromolar range. This assay, which utilizes five label-free proteins involved in H3K56 acetylation in vivo, can in principle identify compounds that inhibit Rtt109-catalyzed H3K56 acetylation via different mechanisms. Compounds discovered via this assay or adaptations thereof could serve as chemical probes or leads for a new class of antifungals targeting an epigenetic enzyme.
Collapse
Affiliation(s)
- Jayme L. Dahlin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
- Medical Scientist Training Program, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Rondedrick Sinville
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jonathan Solberg
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Hui Zhou
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Junhong Han
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Subhashree Francis
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jessica M. Strasser
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kristen John
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Derek J. Hook
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Michael A. Walters
- Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zhiguo Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
23
|
A capillary electrophoretic assay for acetyl coenzyme A carboxylase. Anal Biochem 2013; 437:32-8. [PMID: 23435309 DOI: 10.1016/j.ab.2013.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 01/24/2013] [Accepted: 02/07/2013] [Indexed: 01/18/2023]
Abstract
A simple off-column capillary electrophoretic (CE) assay for measuring acetyl coenzyme A carboxylase holoenzyme (holo-ACC) activity and inhibition was developed. The two reactions catalyzed by the holo-ACC components, biotin carboxylase (BC) and carboxyltransferase (CT), were simultaneously monitored in this assay. Acetyl coenzyme A (CoA), malonyl-CoA, adenosine triphosphate (ATP), and adenosine diphosphate (ADP) were separated by capillary electrophoresis, and the depletion of ATP and acetyl-CoA as well as the production of ADP and malonyl-CoA were monitored. Inhibition of holo-ACC by the BC inhibitor, 2-amino-N,N-dibenzyloxazole-5-carboxamide, and the carboxyltransferase inhibitor, andrimid, was confirmed using this assay. A previously reported off-column CE assay for only the CT component of ACC was optimized, and an off-column CE assay for the BC component of ACC also was developed.
Collapse
|
24
|
Linsky TW, Fast W. Discovery of structurally-diverse inhibitor scaffolds by high-throughput screening of a fragment library with dimethylarginine dimethylaminohydrolase. Bioorg Med Chem 2012; 20:5550-8. [PMID: 22921743 DOI: 10.1016/j.bmc.2012.07.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 07/06/2012] [Accepted: 07/15/2012] [Indexed: 12/11/2022]
Abstract
Potent and selective inhibitors of the enzyme dimethylarginine dimethylaminohydrolase (DDAH) are useful as molecular probes to better understand cellular regulation of nitric oxide. Inhibitors are also potential therapeutic agents for treatment of pathological states associated with the inappropriate overproduction of nitric oxide, such as septic shock, selected types of cancer, and other conditions. Inhibitors with structures dissimilar to substrate may overcome limitations inherent to substrate analogs. Therefore, to identify structurally-diverse inhibitor scaffolds, high-throughput screening (HTS) of a 4000-member library of fragment-sized molecules was completed using the Pseudomonas aeruginosa DDAH and human DDAH-1 isoforms. Use of a substrate concentration equal to its K(M) value during the primary screen allowed for the detection of inhibitors with different modes of inhibition. A series of validation tests were designed and implemented in the identification of four inhibitors of human DDAH-1 that were unknown prior to the screen. Two inhibitors share a 4-halopyridine scaffold and act as quiescent affinity labels that selectively and covalently modify the active-site Cys residue. Two inhibitors are benzimidazole-like compounds that reversibly and competitively inhibit human DDAH-1 with Ligand Efficiency values ≥0.3 kcal/mol/heavy (non-hydrogen) atom, indicating their suitability for further development. Both inhibitor scaffolds have available sites to derivatize for further optimization. Therefore, use of this fragment-based HTS approach is demonstrated to successfully identify two novel scaffolds for development of DDAH-1 inhibitors.
Collapse
Affiliation(s)
- Thomas W Linsky
- Graduate Program in Biochemistry, University of Texas, Austin, TX 78712, USA
| | | |
Collapse
|
25
|
Mammalian fatty acid synthase activity from crude tissue lysates tracing ¹³C-labeled substrates using gas chromatography-mass spectrometry. Anal Biochem 2012; 428:158-66. [PMID: 22728958 DOI: 10.1016/j.ab.2012.06.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/11/2012] [Accepted: 06/13/2012] [Indexed: 01/08/2023]
Abstract
Fatty acid synthase (FASN or FAS, EC 2.3.1.85) is the sole mammalian enzyme to synthesize fatty acids de novo from acetyl- and malonyl-coenzyme A (CoA) esters. This article describes a new method that directly quantifies uniformly labeled (¹³C₁₆-labeled palmitate ([¹³C₁₆]palmitate) by tracing [¹³C₂]acetyl-CoA and [¹³C₃]malonyl-CoA using an in vitro FASN assay. This method used gas chromatography-mass spectrometry (GC-MS) to detect [¹³C₁₆]palmitate carboxylate anions (m/z 271) of pentafluorobenzyl (PFB) derivatives and was highly sensitive at femtomole quantities. Uniformly incorporated [¹³C₁₆]palmitate was the primary product of both recombinant and crude tissue lysate FASN. Quantification of FASN protein within crude tissue lysates ensured equal FASN amounts, preserved steady-state kinetics, and enabled calculation of FASN-specific activity. FASN activity determined by [¹³C₁₆]palmitate synthesis was consistent with values obtained from β-nicotinamide adenine dinucleotide 2'-phosphate (NADPH) oxidation assays. Analysis of FASN activity from tissue extracts was not hampered by contaminating enzymes or preexisting fatty acids. Crude mammary gland and liver lysates had significantly different activities at 82 and 65 nmol min⁻¹ mg⁻¹, respectively, suggesting that tissue-specific activity levels differ in a manner unrelated to FASN amount. GC-MS quantification of [¹³C₁₆]palmitate synthesis permits sensitive evaluation of FASN activity from tissues of varied physiological states and of purified FASN activity in the presence of modifying proteins, enzymes, or drugs.
Collapse
|
26
|
Goncalves V, Brannigan JA, Thinon E, Olaleye TO, Serwa R, Lanzarone S, Wilkinson AJ, Tate EW, Leatherbarrow RJ. A fluorescence-based assay for N-myristoyltransferase activity. Anal Biochem 2011; 421:342-4. [PMID: 22051857 DOI: 10.1016/j.ab.2011.10.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 09/30/2011] [Accepted: 10/05/2011] [Indexed: 11/26/2022]
Abstract
N-myristoylation is the irreversible attachment of a C(14) fatty acid, myristic acid, to the N-terminal glycine of a protein via formation of an amide bond. This modification is catalyzed by myristoyl-coenzyme A (CoA):protein N-myristoyltransferase (NMT), an enzyme ubiquitous in eukaryotes that is up-regulated in several cancers. Here we report a sensitive fluorescence-based assay to study the enzymatic activity of human NMT1 and NMT2 based on detection of CoA by 7-diethylamino-3-(4-maleimido-phenyl)-4-methylcoumarin. We also describe expression and characterization of NMT1 and NMT2 and assay validation with small molecule inhibitors. This assay should be broadly applicable to NMTs from a range of organisms.
Collapse
Affiliation(s)
- Victor Goncalves
- Department of Chemistry, Imperial College London, London SW7 2AZ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cao J, Zhou Y, Peng H, Huang X, Stahler S, Suri V, Qadri A, Gareski T, Jones J, Hahm S, Perreault M, McKew J, Shi M, Xu X, Tobin JF, Gimeno RE. Targeting Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) with small molecule inhibitors for the treatment of metabolic diseases. J Biol Chem 2011; 286:41838-41851. [PMID: 21990351 DOI: 10.1074/jbc.m111.245456] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is one of two known DGAT enzymes that catalyze the final step in triglyceride synthesis. Findings from genetically modified mice as well as pharmacological studies suggest that inhibition of DGAT1 is a promising strategy for the treatment of obesity and type 2 diabetes. Here we characterize a tool DGAT1 inhibitor compound, T863. We found that T863 is a potent inhibitor for both human and mouse DGAT1 in vitro, which acts on the acyl-CoA binding site of DGAT1 and inhibits DGAT1-mediated triacylglycerol formation in cells. In an acute lipid challenge model, oral administration of T863 significantly delayed fat absorption and resulted in lipid accumulation in the distal small intestine of mice, mimicking the effects of genetic ablation of DGAT1. In diet-induced obese mice, oral administration of T863 for 2 weeks caused weight loss, reduction in serum and liver triglycerides, and improved insulin sensitivity. In addition to the expected triglyceride-lowering activity, T863 also lowered serum cholesterol. Hepatic IRS2 protein was dramatically up-regulated in mice treated with T863, possibly contributing to improved insulin sensitivity. In differentiated 3T3-L1 adipocytes, T863 enhanced insulin-stimulated glucose uptake, suggesting a possible role for adipocytes to improve insulin sensitivity upon DGAT1 inhibition. These results reveal novel mechanistic insights into the insulin-sensitizing effects of DGAT1 inhibition in mouse models. Taken together, our study provides a comprehensive evaluation of a small molecule inhibitor for DGAT1 and suggests that pharmacological inhibition of DGAT1 holds promise in treating diverse metabolic disorders.
Collapse
Affiliation(s)
- Jingsong Cao
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140.
| | - Yingjiang Zhou
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Haibing Peng
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Xinyi Huang
- Screening Sciences, Pfizer, Collegeville, Pennsylvania 19426
| | - Shannon Stahler
- Screening Sciences, Pfizer, Collegeville, Pennsylvania 19426
| | - Vipin Suri
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Ariful Qadri
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Tiffany Gareski
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Juli Jones
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Seung Hahm
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Mylene Perreault
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - John McKew
- Exploratory Medicinal Chemistry, Pfizer, Cambridge, Massachusetts 02140
| | - Mengxiao Shi
- Discovery Pharmacokinetics, Pfizer, Andover, Massachusetts 01810
| | - Xin Xu
- Discovery Pharmacokinetics, Pfizer, Andover, Massachusetts 01810
| | - James F Tobin
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Ruth E Gimeno
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140.
| |
Collapse
|
28
|
Linsky T, Fast W. A continuous, fluorescent, high-throughput assay for human dimethylarginine dimethylaminohydrolase-1. JOURNAL OF BIOMOLECULAR SCREENING 2011; 16:1089-97. [PMID: 21921133 PMCID: PMC3248755 DOI: 10.1177/1087057111417712] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inhibitors of human dimethylarginine dimethylaminohydrolase-1 (DDAH-1) are of therapeutic interest for controlling pathological nitric oxide production. Only a limited number of biologically useful inhibitors have been identified, so structurally diverse lead compounds are desired. In contrast with previous assays that do not possess adequate sensitivity for optimal screening, herein is reported a high-throughput assay that uses an alternative thiol-releasing substrate, S-methyl-L-thiocitrulline, and a thiol-reactive fluorophore, 7-diethylamino-3-(4'-maleimidylphenyl)-4-methylcoumarin, to enable continuous detection of product formation by DDAH-1. The assay is applied to query two commercial libraries totaling 4446 compounds, and two representative hits are described, including a known DDAH-1 inhibitor. This is the most sensitive DDAH-1 assay reported to date and enables screening of compound libraries using [S] = K (M) conditions while displaying Z' factors from 0.6 to 0.8. Therefore, this strategy now makes possible high-throughput screening for human DDAH-1 inhibitors in pursuit of molecular probes and drugs to control excessive nitric oxide production.
Collapse
Affiliation(s)
- Thomas Linsky
- Graduate Program in Biochemistry, University of Texas, Austin, USA
| | | |
Collapse
|
29
|
Quinn AM, Simeonov A. Methods for Activity Analysis of the Proteins that Regulate Histone Methylation. CURRENT CHEMICAL GENOMICS 2011; 5:95-105. [PMID: 21966349 PMCID: PMC3180180 DOI: 10.2174/1875397301005010095] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 03/23/2011] [Accepted: 04/25/2011] [Indexed: 12/20/2022]
Abstract
The enzymes that regulate histone methylation states and the protein domains that recognize methylated histone residues have been implicated in a number of human diseases, including cancer, as a result of their ability to affect transcriptional changes by altering chromatin structure. These proteins are recognized as potential therapeutic targets for the treatment of diseases associated with epigenetic disruption; however, few inhibitors of their activity have been identified. The majority of histone demethylase and methyltransferase enzyme inhibitors have been discovered on the basis of their structural similarity to substrates or known inhibitors of enzymes with analogous mechanisms. The general lack of potency and specificity of these compounds indicates that novel chemotypes are needed to address the large number of recently discovered histone-modifying enzymes. High-throughput screening (HTS) allows rapid testing of chemically diverse small molecule libraries, provided assays amenable to HTS exist. Here we review the biochemical and cellular assays available for testing the proteins and enzymes that regulate histone methylation. Progress in the development of high-throughput, sensitive, and robust assays will enable discovery of small molecules for epigenetic therapy.
Collapse
Affiliation(s)
- Amy M Quinn
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-3370, USA
| | | |
Collapse
|
30
|
Alves J, Westling L, Peters EC, Harris JL, Trauger JW. Cloning, expression, and enzymatic activity of Acinetobacter baumannii and Klebsiella pneumoniae acetyl-coenzyme A carboxylases. Anal Biochem 2011; 417:103-11. [PMID: 21704013 DOI: 10.1016/j.ab.2011.05.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 05/02/2011] [Accepted: 05/25/2011] [Indexed: 10/18/2022]
Abstract
Pathogenic Gram-negative bacteria are a major public health concern because they are causative agents of life-threatening hospital-acquired infections. Due to the increasing rates of resistance to available antibiotics, there is an urgent need to develop new drugs. Acetyl-coenzyme A carboxylase (ACCase) is a promising target for the development of novel antibiotics. We describe here the expression, purification, and enzymatic activity of recombinant ACCases from two clinically relevant Gram-negative pathogens, Acinetobacter baumannii and Klebsiella pneumoniae. Recombinant ACCase subunits (AccAD, AccB, and AccC) were expressed and purified, and the holoenzymes were reconstituted. ACCase enzyme activity was monitored by direct detection of malonyl-coenzyme A (malonyl-CoA) formation by liquid chromatography tandem mass spectrometry (LC-MS/MS). Steady-state kinetics experiments showed similar k(cat) and K(M) values for both enzymes. In addition, similar IC(50) values were observed for inhibition of both enzymes by a previously reported ACCase inhibitor. To provide a higher throughput assay suitable for inhibitor screening, we developed and validated a luminescence-based ACCase assay that monitors ATP depletion. Finally, we established an enzyme activity assay for the isolated AccAD (carboxyltransferase) subunit, which is useful for determining whether novel ACCase inhibitors inhibit the biotin carboxylase or carboxyltransferase site of ACCase. The methods described here could be applied toward the identification and characterization of novel inhibitors.
Collapse
Affiliation(s)
- Juliano Alves
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | | | | | | | | |
Collapse
|
31
|
Antidiabetic and antisteatotic effects of the selective fatty acid synthase (FAS) inhibitor platensimycin in mouse models of diabetes. Proc Natl Acad Sci U S A 2011; 108:5378-83. [PMID: 21389266 DOI: 10.1073/pnas.1002588108] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Platensimycin (PTM) is a recently discovered broad-spectrum antibiotic produced by Streptomyces platensis. It acts by selectively inhibiting the elongation-condensing enzyme FabF of the fatty acid biosynthesis pathway in bacteria. We report here that PTM is also a potent and highly selective inhibitor of mammalian fatty acid synthase. In contrast to two agents, C75 and cerulenin, that are widely used as inhibitors of mammalian fatty acid synthase, platensimycin specifically inhibits fatty acid synthesis but not sterol synthesis in rat primary hepatocytes. PTM preferentially concentrates in liver when administered orally to mice and potently inhibits hepatic de novo lipogenesis, reduces fatty acid oxidation, and increases glucose oxidation. Chronic administration of platensimycin led to a net reduction in liver triglyceride levels and improved insulin sensitivity in db/+ mice fed a high-fructose diet. PTM also reduced ambient glucose levels in db/db mice. These results provide pharmacological proof of concept of inhibiting fatty acid synthase for the treatment of diabetes and related metabolic disorders in animal models.
Collapse
|
32
|
Cerne D, Zitnik IP, Sok M. Increased fatty acid synthase activity in non-small cell lung cancer tissue is a weaker predictor of shorter patient survival than increased lipoprotein lipase activity. Arch Med Res 2011; 41:405-9. [PMID: 21044743 DOI: 10.1016/j.arcmed.2010.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 07/21/2010] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Cumulative evidence suggests the involvement of fatty acid synthase (FAS) in tumor growth. We tested the hypothesis that increased FAS activity and gene expression in non-small cell lung cancer (NSCLC) tissue have a prognostic significance that is independent of that of increased lipoprotein lipase (LPL) activity in the same tissue. METHODS Forty two consecutive patients with resected NSCLC were enrolled in the study. Paired samples of lung cancer tissue and adjacent non-cancer lung tissue were collected from resected specimens for estimation of FAS activity and expression of its gene. LPL activity had previously been measured in the same tissues. During a 4-year follow-up, 21 patients died due to tumor progression. One patient died due to a non-cancer reason and was not included in the analysis. RESULTS High FAS activity in cancerous tissue relative to that in the adjacent non-cancer lung tissue was associated with weight loss in the 3 months immediately before tumor excision and patient death during the follow-up. Higher FAS activity in the cancer tissue was associated with higher LPL activity in the same tissue, which also predicted shorter patient survival, but LPL was the stronger predictor. FAS gene expression was higher in the adjacent non-cancer tissue than in the cancer tissue but had no predictive value. CONCLUSION Our study further underlines the involvement of cancer tissue FAS activity in tumor growth but also indicates its weaker importance compared to LPL activity.
Collapse
Affiliation(s)
- Darko Cerne
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| | | | | |
Collapse
|
33
|
Tanaka K, Koresawa M, Iida M, Fukasawa K, Stec E, Cassaday J, Chase P, Rickert K, Hodder P, Takagi T, Komatani H. Multiplexed random peptide library and phospho-specific antibodies facilitate human polo-like kinase 1 inhibitor screen. Assay Drug Dev Technol 2010; 8:47-62. [PMID: 20085455 PMCID: PMC3532019 DOI: 10.1089/adt.2009.0212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
One of the challenges to develop time-resolved fluorescence resonance energy transfer (TR-FRET) assay for serine/threonine (Ser/Thr) protein kinase is to select an optimal peptide substrate and a specific phosphor Ser/Thr antibody. This report describes a multiplexed random screen-based development of TR-FRET assay for ultra-high-throughput screening (uHTS) of small molecule inhibitors for a potent cancer drug target polo-like kinase 1 (Plk1). A screen of a diverse peptide library in a 384-well plate format identified several highly potent substrates that share the consensus motif for phosphorylation by Plk1. Their potencies were comparable to FKD peptide, a designed peptide substrate derived from well-described Plk1 substrate Cdc25C. A specific anti-phosphor Ser/Thr antibody p(S/T)F antibody that detects the phosphorylation of FKD peptide was screened out of 87 antibodies with time-resolved fluorometry technology in a 96-well plate format. Using FKD peptide and p(S/T)F antibody, we successfully developed a robust TR-FRET assay in 384-well plate format, and further miniaturized this assay to 1,536-well plate format to perform uHTS. We screened about 1.2 million compounds for Plk1 inhibitors using a Plk1 deletion mutant that only has the kinase domain and subsequently screened the same compound library using a full-length active-mutant Plk1. These uHTSs identified a number of hit compounds, and some of them had selectivity to either the deletion mutant or the full-length protein. Our results prove that a combination of random screen for substrate peptide and phospho-specific antibodies is very powerful strategy to develop TR-FRET assays for protein kinases.
Collapse
Affiliation(s)
- Kenji Tanaka
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., Tsukuba, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Planey SL, Zacharias DA. Identification of targets and inhibitors of protein palmitoylation. Expert Opin Drug Discov 2010; 5:155-64. [DOI: 10.1517/17460440903548218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
35
|
Bays NW, Hill AD, Kariv I. A simplified scintillation proximity assay for fatty acid synthase activity: development and comparison with other FAS activity assays. ACTA ACUST UNITED AC 2009; 14:636-42. [PMID: 19531664 DOI: 10.1177/1087057109335746] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Fatty acid synthase (FAS), an essential enzyme for de novo lipogenesis, has been implicated in a number of disease states, including obesity, dyslipidemia, and cancer. To identify small-molecule inhibitors of FAS, the authors developed a bead-based scintillation proximity assay (SPA) to detect the fatty acid products of FAS enzymatic activity. This homogeneous SPA assay discriminates between a radiolabeled hydrophilic substrate of FAS (acetyl-coenzyme A) and the labeled lipophilic products of FAS (fatty acids), generating signal only when labeled fatty acids are present. The assay requires a single addition of unmodified polystyrene imaging SPA beads and can be miniaturized to 384- or 1536-well density with appropriate assay statistics for high-throughput screening. High-potency FAS inhibitors were used to compare the sensitivity of the SPA bead assay with previously described assays that measure FAS reaction intermediates (CoA-SH and NADP+). The advantages and disadvantages of these different FAS assays in small-molecule inhibitor discovery are discussed.
Collapse
Affiliation(s)
- Nathan W Bays
- Department of In Vitro Sciences, Merck Research Laboratories, Boston, Massachusetts, USA.
| | | | | |
Collapse
|