1
|
Deschner F, Mostert D, Daniel JM, Voltz A, Schneider DC, Khangholi N, Bartel J, Pessanha de Carvalho L, Brauer M, Gorelik TE, Kleeberg C, Risch T, Haeckl FPJ, Herraiz Benítez L, Andreas A, Kany AM, Jézéquel G, Hofer W, Müsken M, Held J, Bischoff M, Seemann R, Brötz-Oesterhelt H, Schneider T, Sieber S, Müller R, Herrmann J. Natural products chlorotonils exert a complex antibacterial mechanism and address multiple targets. Cell Chem Biol 2025; 32:586-602.e15. [PMID: 40203831 DOI: 10.1016/j.chembiol.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Antimicrobial resistance is a threat to human health rendering current first-line antibiotics ineffective. New agents overcoming resistance mechanisms are urgently needed to guarantee successful treatment of human disease in the future. Chlorotonils, a natural product class with yet unknown mode of action, were shown to have broad-spectrum activity against multi-resistant Gram-positive bacteria and the malaria parasite Plasmodium falciparum, with promising activity and safety in murine infection models. Here, we report that chlorotonils can target the cell membrane, cell wall, and protein biosynthesis. They can be characterized by a rapid onset of action via interference with ion homeostasis leading to membrane depolarization, however, without inducing severe barrier failure or cellular lysis. Further characterization confirmed binding of chlorotonils to bacterial membrane lipids eventually leading to uncontrolled potassium transport. Additionally, we identified functional inhibition of the peptidoglycan biosynthesis protein YbjG and methionine aminopeptidase MetAP as secondary targets of chlorotonils.
Collapse
Affiliation(s)
- Felix Deschner
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Dietrich Mostert
- Center for Functional Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Jan-Martin Daniel
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, 53127 Bonn, Germany
| | - Alexander Voltz
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Dana Carina Schneider
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany
| | - Navid Khangholi
- Experimental Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Jürgen Bartel
- Department of Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489 Greifswald, Germany
| | | | - Madita Brauer
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, 17489 Greifswald, Germany
| | - Tatiana E Gorelik
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Institute of Inorganic and Analytical Chemistry, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany; Ernst Ruska-Centre for Microscopy and Spectroscopy with Electrons, Forschungszentrum Jülich GmbH, Jülich 52428, Germany
| | - Christian Kleeberg
- Institute for Inorganic and Analytical Chemistry, Technical University of Braunschweig, 38106 Braunschweig, Germany
| | - Timo Risch
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - F P Jake Haeckl
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Laura Herraiz Benítez
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Anastasia Andreas
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Andreas Martin Kany
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Gwenaëlle Jézéquel
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Walter Hofer
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Jana Held
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute of Tropical Medicine, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Markus Bischoff
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Institute for Medical Microbiology and Hygiene, Saarland University, 66421 Homburg, Germany
| | - Ralf Seemann
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany; Cluster or Excellence "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Tanja Schneider
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, 53127 Bonn, Germany
| | - Stephan Sieber
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Center for Functional Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Rolf Müller
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Jennifer Herrmann
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany.
| |
Collapse
|
2
|
Hodyna D, Kovalishyn V, Kachaeva M, Shulha Y, Klipkov A, Shaitanova E, Kobzar O, Shablykin O, Metelytsia L. In Silico, in Vitro and in Vivo Study of Substituted Imidazolidinone Sulfonamides as Antibacterial Agents. Chem Biodivers 2023; 20:e202301267. [PMID: 37943002 DOI: 10.1002/cbdv.202301267] [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: 08/21/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/10/2023]
Abstract
New substituted imidazolidinone sulfonamides have been developed using a rational drug design strategy. Predictive QSAR models for the search of new antibacterials were created using the OCHEM platform. Regression models were applied to verify a virtual chemical library of new imidazolidinone derivatives designed to have antibacterial activity. A number of substituted imidazolidinone sulfonamides as effective antibacterial agents were identified by QSAR prediction, synthesized and characterized by spectral and elemental, and tested in vitro. Six studied compounds have shown the highest in vitro antibacterial activity against Gram-negative E. coli and Gram-positive S. aureus multidrug-resistant strains. The in vivo acute toxicity of these imidazolidinone sulfonamides based on the LC50 value ranged from 16.01 to 44.35 mg/L (slightly toxic compounds class). The results of molecular docking suggest that the antibacterial mechanism of the compounds can be associated with the inhibition of post-translational modification processes of bacterial peptides and proteins.
Collapse
Affiliation(s)
- Diana Hodyna
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Vasyl Kovalishyn
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Maryna Kachaeva
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Yurii Shulha
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Anton Klipkov
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Elena Shaitanova
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Oleksandr Kobzar
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Oleh Shablykin
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| | - Larysa Metelytsia
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the National Academy of Sciences of Ukraine, 02094, Academician Kukhar Str, 1, Kyiv, Ukraine
| |
Collapse
|
3
|
Bala S, Yellamanda KV, Kadari A, Ravinuthala VSU, Kattula B, Singh OV, Gundla R, Addlagatta A. Selective inhibition of Helicobacter pylori methionine aminopeptidase by azaindole hydroxamic acid derivatives: Design, synthesis, in vitro biochemical and structural studies. Bioorg Chem 2021; 115:105185. [PMID: 34329997 DOI: 10.1016/j.bioorg.2021.105185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022]
Abstract
Methionine aminopeptidases (MetAPs) are an important class of enzymes that work co-translationally for the removal of initiator methionine. Chemical inhibition or gene knockdown is lethal to the microbes suggesting that they can be used as antibiotic targets. However, sequence and structural similarity between the microbial and host MetAPs has been a challenge in the identification of selective inhibitors. In this study, we have analyzed several thousands of MetAP sequences and established a pattern of variation in the S1 pocket of the enzyme. Based on this knowledge, we have designed a library of 17 azaindole based hydroxamic acid derivatives which selectively inhibited the MetAP from H. pylori compared to the human counterpart. Structural studies provided the molecular basis for the selectivity.
Collapse
Affiliation(s)
- Sandeepchowdary Bala
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Kalisha Vali Yellamanda
- Department of Chemistry, School of Science, GITAM Deemed to be University, Hyderabad 502 102, Telangana, India
| | - Anilkumar Kadari
- Department of Chemistry, School of Science, GITAM Deemed to be University, Hyderabad 502 102, Telangana, India
| | - Venkata S U Ravinuthala
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India
| | - Bhavita Kattula
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Om V Singh
- Department of Chemistry, School of Science, GITAM Deemed to be University, Hyderabad 502 102, Telangana, India
| | - Rambabu Gundla
- Department of Chemistry, School of Science, GITAM Deemed to be University, Hyderabad 502 102, Telangana, India.
| | - Anthony Addlagatta
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| |
Collapse
|
4
|
Chen AY, Adamek RN, Dick BL, Credille CV, Morrison CN, Cohen SM. Targeting Metalloenzymes for Therapeutic Intervention. Chem Rev 2019; 119:1323-1455. [PMID: 30192523 PMCID: PMC6405328 DOI: 10.1021/acs.chemrev.8b00201] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metalloenzymes are central to a wide range of essential biological activities, including nucleic acid modification, protein degradation, and many others. The role of metalloenzymes in these processes also makes them central for the progression of many diseases and, as such, makes metalloenzymes attractive targets for therapeutic intervention. Increasing awareness of the role metalloenzymes play in disease and their importance as a class of targets has amplified interest in the development of new strategies to develop inhibitors and ultimately useful drugs. In this Review, we provide a broad overview of several drug discovery efforts focused on metalloenzymes and attempt to map out the current landscape of high-value metalloenzyme targets.
Collapse
Affiliation(s)
- Allie Y Chen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Rebecca N Adamek
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Benjamin L Dick
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Cy V Credille
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Christine N Morrison
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Seth M Cohen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
5
|
Helgren TR, Seven ES, Chen C, Edwards TE, Staker BL, Abendroth J, Myler PJ, Horn JR, Hagen TJ. The identification of inhibitory compounds of Rickettsia prowazekii methionine aminopeptidase for antibacterial applications. Bioorg Med Chem Lett 2018; 28:1376-1380. [PMID: 29551481 PMCID: PMC5908248 DOI: 10.1016/j.bmcl.2018.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 11/25/2022]
Abstract
Methionine aminopeptidase (MetAP) is a dinuclear metalloprotease responsible for the cleavage of methionine initiator residues from nascent proteins. MetAP activity is necessary for bacterial proliferation and is therefore a projected novel antibacterial target. A compound library consisting of 294 members containing metal-binding functional groups was screened against Rickettsia prowazekii MetAP to determine potential inhibitory motifs. The compounds were first screened against the target at a concentration of 10 µM and potential hits were determined to be those exhibiting greater than 50% inhibition of enzymatic activity. These hit compounds were then rescreened against the target in 8-point dose-response curves and 11 compounds were found to inhibit enzymatic activity with IC50 values of less than 10 µM. Finally, compounds (1-5) were docked against RpMetAP with AutoDock to determine potential binding mechanisms and the results were compared with crystal structures deposited within the PDB.
Collapse
Affiliation(s)
- Travis R Helgren
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Elif S Seven
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Congling Chen
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Thomas E Edwards
- Beryllium Discovery Corp., 7869 NE Day Road West, Bainbridge Island, WA 98110, USA; Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Bart L Staker
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA; Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA
| | - Jan Abendroth
- Beryllium Discovery Corp., 7869 NE Day Road West, Bainbridge Island, WA 98110, USA; Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Peter J Myler
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA; Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA
| | - James R Horn
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Timothy J Hagen
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA.
| |
Collapse
|
6
|
Žalubovskis R, Winum JY. Inhibitors of Selected Bacterial Metalloenzymes. Curr Med Chem 2018; 26:2690-2714. [PMID: 29611472 DOI: 10.2174/0929867325666180403154018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/14/2018] [Accepted: 03/19/2018] [Indexed: 12/26/2022]
Abstract
The utilization of bacterial metalloenzymes, especially ones not having mammalian (human) counterparts, has drawn attention to develop novel antibacterial agents to overcome drug resistance and especially multidrug resistance. In this review, we focus on the recent achievements on the development of inhibitors of bacterial enzymes peptide deformylase (PDF), metallo-β-lactamase (MBL), methionine aminopeptidase (MetAP) and UDP-3-O-acyl- N-acetylglucosamine deacetylase (LpxC). The state of the art of the design and investigation of inhibitors of bacterial metalloenzymes is presented, and challenges are outlined and discussed.
Collapse
Affiliation(s)
- Raivis Žalubovskis
- Latvian Institute of Organic Synthesis, Riga, Latvia.,Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Latvia
| | - Jean-Yves Winum
- Institut des Biomolecules Max Mousseron, Universite de Montpellier, France
| |
Collapse
|
7
|
Helgren TR, Chen C, Wangtrakuldee P, Edwards TE, Staker BL, Abendroth J, Sankaran B, Housley NA, Myler PJ, Audia JP, Horn JR, Hagen TJ. Rickettsia prowazekii methionine aminopeptidase as a promising target for the development of antibacterial agents. Bioorg Med Chem 2017; 25:813-824. [PMID: 28089350 PMCID: PMC5319851 DOI: 10.1016/j.bmc.2016.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/06/2016] [Accepted: 11/08/2016] [Indexed: 01/07/2023]
Abstract
Methionine aminopeptidase (MetAP) is a class of ubiquitous enzymes essential for the survival of numerous bacterial species. These enzymes are responsible for the cleavage of N-terminal formyl-methionine initiators from nascent proteins to initiate post-translational modifications that are often essential to proper protein function. Thus, inhibition of MetAP activity has been implicated as a novel antibacterial target. We tested this idea in the present study by targeting the MetAP enzyme in the obligate intracellular pathogen Rickettsia prowazekii. We first identified potent RpMetAP inhibitory species by employing an in vitro enzymatic activity assay. The molecular docking program AutoDock was then utilized to compare published crystal structures of inhibited MetAP species to docked poses of RpMetAP. Based on these in silico and in vitro screens, a subset of 17 compounds was tested for inhibition of R. prowazekii growth in a pulmonary vascular endothelial cell (EC) culture infection model system. All compounds were tested over concentration ranges that were determined to be non-toxic to the ECs and 8 of the 17 compounds displayed substantial inhibition of R. prowazekii growth. These data highlight the therapeutic potential for inhibiting RpMetAP as a novel antimicrobial strategy and set the stage for future studies in pre-clinical animal models of infection.
Collapse
Affiliation(s)
- Travis R Helgren
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Congling Chen
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Phumvadee Wangtrakuldee
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Thomas E Edwards
- Beryllium Discovery Corp., 7869 NE Day Road West, Bainbridge Island, WA 98110, USA; Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Bart L Staker
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA; Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Jan Abendroth
- Beryllium Discovery Corp., 7869 NE Day Road West, Bainbridge Island, WA 98110, USA; Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Berkeley Center for Structural Biology, Ernest Orlando Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Nicole A Housley
- Department of Microbiology and Immunology and The Center for Lung Biology, University of South Alabama College of Medicine, Laboratory of Infectious Diseases, 307 North University Blvd, Mobile, AL 36688, USA
| | - Peter J Myler
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA; Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA; Department of Global Health and Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Jonathon P Audia
- Department of Microbiology and Immunology and The Center for Lung Biology, University of South Alabama College of Medicine, Laboratory of Infectious Diseases, 307 North University Blvd, Mobile, AL 36688, USA
| | - James R Horn
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA
| | - Timothy J Hagen
- Department of Chemistry and Biochemistry, Northern Illinois University, 1425 W. Lincoln Hwy, DeKalb, IL 60115, USA.
| |
Collapse
|
8
|
A Synopsis of the Properties and Applications of Heteroaromatic Rings in Medicinal Chemistry. ADVANCES IN HETEROCYCLIC CHEMISTRY 2017. [DOI: 10.1016/bs.aihch.2016.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
9
|
Novel broad-spectrum inhibitors of bacterial methionine aminopeptidase. Bioorg Med Chem Lett 2015; 25:3301-6. [PMID: 26099541 DOI: 10.1016/j.bmcl.2015.05.061] [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: 04/13/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 01/09/2023]
Abstract
With increasing emergence of multi-drug resistant infections, there is a dire need for new classes of compounds that act through unique mechanisms. In this work, we describe the discovery and optimization of a novel series of inhibitors of bacterial methionine aminopeptidase (MAP). Through a high-throughput screening campaign, one azepinone amide hit was found that resembled the native peptide substrate and possessed moderate biochemical potency against three bacterial isozymes. X-ray crystallography was used in combination with substrate-based design to direct the rational optimization of analogs with sub-micromolar potency. The novel compounds presented here represent potent broad-spectrum biochemical inhibitors of bacterial MAP and have the potential to lead to the development of new medicines to combat serious multi-drug resistant infections.
Collapse
|
10
|
Arya T, Reddi R, Kishor C, Ganji RJ, Bhukya S, Gumpena R, McGowan S, Drag M, Addlagatta A. Identification of the molecular basis of inhibitor selectivity between the human and streptococcal type I methionine aminopeptidases. J Med Chem 2015; 58:2350-7. [PMID: 25699713 DOI: 10.1021/jm501790e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The methionine aminopeptidase (MetAP) family is responsible for the cleavage of the initiator methionine from newly synthesized proteins. Currently, there are no small molecule inhibitors that show selectivity toward the bacterial MetAPs compared to the human enzyme. In our current study, we have screened 20 α-aminophosphonate derivatives and identified a molecule (compound 15) that selectively inhibits the S. pneumonia MetAP in low micromolar range but not the human enzyme. Further bioinformatics, biochemical, and structural analyses suggested that phenylalanine (F309) in the human enzyme and methionine (M205) in the S. pneumonia MetAP at the analogous position render them with different susceptibilities against the identified inhibitor. X-ray crystal structures of various inhibitors in complex with wild type and F309M enzyme further established the molecular basis for the inhibitor selectivity.
Collapse
Affiliation(s)
- Tarun Arya
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Mitić N, Miraula M, Selleck C, Hadler KS, Uribe E, Pedroso MM, Schenk G. Catalytic mechanisms of metallohydrolases containing two metal ions. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 97:49-81. [PMID: 25458355 DOI: 10.1016/bs.apcsb.2014.07.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
At least one-third of enzymes contain metal ions as cofactors necessary for a diverse range of catalytic activities. In the case of polymetallic enzymes (i.e., two or more metal ions involved in catalysis), the presence of two (or more) closely spaced metal ions gives an additional advantage in terms of (i) charge delocalisation, (ii) smaller activation barriers, (iii) the ability to bind larger substrates, (iv) enhanced electrostatic activation of substrates, and (v) decreased transition-state energies. Among this group of proteins, enzymes that catalyze the hydrolysis of ester and amide bonds form a very prominent family, the metallohydrolases. These enzymes are involved in a multitude of biological functions, and an increasing number of them gain attention for translational research in medicine and biotechnology. Their functional versatility and catalytic proficiency are largely due to the presence of metal ions in their active sites. In this chapter, we thus discuss and compare the reaction mechanisms of several closely related enzymes with a view to highlighting the functional diversity bestowed upon them by their metal ion cofactors.
Collapse
Affiliation(s)
- Nataša Mitić
- Department of Chemistry, National University of Ireland, Maynooth, Maynooth, Co. Kildare, Ireland.
| | - Manfredi Miraula
- Department of Chemistry, National University of Ireland, Maynooth, Maynooth, Co. Kildare, Ireland; School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Christopher Selleck
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Kieran S Hadler
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Elena Uribe
- Department of Biochemistry and Molecular Biology, University of Concepción, Concepción, Chile
| | - Marcelo M Pedroso
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Gerhard Schenk
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
12
|
Uda NR, Upert G, Angelici G, Nicolet S, Schmidt T, Schwede T, Creus M. Zinc-selective inhibition of the promiscuous bacterial amide-hydrolase DapE: implications of metal heterogeneity for evolution and antibioticdrug design. Metallomics 2014; 6:88-95. [DOI: 10.1039/c3mt00125c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
13
|
Daumann LJ, Schenk G, Ollis DL, Gahan LR. Spectroscopic and mechanistic studies of dinuclear metallohydrolases and their biomimetic complexes. Dalton Trans 2013; 43:910-28. [PMID: 24135968 DOI: 10.1039/c3dt52287c] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An enhanced understanding of the metal ion binding and active site structural features of phosphoesterases such as the glycerophosphodiesterase from Enterobacter aerogenes (GpdQ), and the organophosphate degrading agent from Agrobacterium radiobacter (OpdA) have important consequences for potential applications. Coupled with investigations of the metalloenzymes, programs of study to synthesise and characterise model complexes based on these metalloenzymes can add to our understanding of structure and function of the enzymes themselves. This review summarises some of our work and illustrates the significance and contributions of model studies to knowledge in the area.
Collapse
Affiliation(s)
- Lena J Daumann
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | |
Collapse
|
14
|
Wangtrakuldee P, Byrd MS, Campos CG, Henderson MW, Zhang Z, Clare M, Masoudi A, Myler PJ, Horn JR, Cotter PA, Hagen TJ. Discovery of Inhibitors of Burkholderia pseudomallei Methionine Aminopeptidase with Antibacterial Activity. ACS Med Chem Lett 2013; 4. [PMID: 24376907 DOI: 10.1021/ml400034m] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Evaluation of a series of MetAP inhibitors in an in vitro enzyme activity assay led to the first identification of potent molecules that show significant growth inhibition against Burkholderia pseudomallei. Nitroxoline analogs show excellent inhibition potency in the BpMetAP1 enzyme activity assay with the lowest IC50 of 30 nM, and inhibit the growth of B. pseudomallei and B. thailandensis at concentrations ≥ 31 μM.
Collapse
Affiliation(s)
- Phumvadee Wangtrakuldee
- Department
of Chemistry and
Biochemistry, Northern Illinois University, 1425 West Lincoln Highway, DeKalb, Illinois 60115-2828, United
States
| | - Matthew S. Byrd
- Department of Microbiology
and
Immunology, University of North Carolina, 116 Manning Drive, Chapel Hill, North Carolina 27599, United States
| | - Cristine G. Campos
- Department of Microbiology
and
Immunology, University of North Carolina, 116 Manning Drive, Chapel Hill, North Carolina 27599, United States
| | - Michael W. Henderson
- Department of Microbiology
and
Immunology, University of North Carolina, 116 Manning Drive, Chapel Hill, North Carolina 27599, United States
| | - Zheng Zhang
- Department
of Chemistry and
Biochemistry, Northern Illinois University, 1425 West Lincoln Highway, DeKalb, Illinois 60115-2828, United
States
| | - Michael Clare
- Clare Associates LLC, 5154 West Brown Street, Skokie, Illinois 60077,
United States
| | - Ali Masoudi
- Seattle Biomedical Research Institute, 307 Westlake Avenue N, Seattle,
Washington 98109-5219, United States, and Department of Global Health
and Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, Washington 98195,
United States
| | - Peter J. Myler
- Seattle Biomedical Research Institute, 307 Westlake Avenue N, Seattle,
Washington 98109-5219, United States, and Department of Global Health
and Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, Washington 98195,
United States
| | - James R. Horn
- Department
of Chemistry and
Biochemistry, Northern Illinois University, 1425 West Lincoln Highway, DeKalb, Illinois 60115-2828, United
States
| | - Peggy A. Cotter
- Department of Microbiology
and
Immunology, University of North Carolina, 116 Manning Drive, Chapel Hill, North Carolina 27599, United States
| | - Timothy J. Hagen
- Department
of Chemistry and
Biochemistry, Northern Illinois University, 1425 West Lincoln Highway, DeKalb, Illinois 60115-2828, United
States
| |
Collapse
|
15
|
Kishor C, Arya T, Reddi R, Chen X, Saddanapu V, Marapaka AK, Gumpena R, Ma D, Liu JO, Addlagatta A. Identification, Biochemical and Structural Evaluation of Species-Specific Inhibitors against Type I Methionine Aminopeptidases. J Med Chem 2013; 56:5295-305. [DOI: 10.1021/jm400395p] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Chandan Kishor
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| | - Tarun Arya
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| | - Ravikumar Reddi
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| | - Xiaochun Chen
- Department of Pediatrics, University of Maryland School of Medicine, 655 West
Baltimore, Street, Baltimore, Maryland 21201, United States
| | - Venkateshwarlu Saddanapu
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| | - Anil Kumar Marapaka
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| | - Rajesh Gumpena
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| | - Dawei Ma
- State Key Laboratory of Bioorganic
and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 354
Fenglin Road, Shanghai 200032, China
| | - Jun O. Liu
- Departments of Pharmacology
and Molecular Sciences, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland
21205, United States
| | - Anthony Addlagatta
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka,
Hyderabad AP-500 007, India
| |
Collapse
|
16
|
Remy L, Carrière M, Derré-Bobillot A, Martini C, Sanguinetti M, Borezée-Durant E. The Staphylococcus aureus Opp1 ABC transporter imports nickel and cobalt in zinc-depleted conditions and contributes to virulence. Mol Microbiol 2012; 87:730-43. [PMID: 23279021 DOI: 10.1111/mmi.12126] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2012] [Indexed: 11/27/2022]
Abstract
Metals are common enzymatic cofactors, and their acquisition must be assured under the various conditions encountered in the host. Although some strategies for acquisition of common metals such as iron and manganese have been elucidated, little is known about the conditions and mechanisms used to capture trace metals. Nickel is a transition metal required as a cofactor for several bacterial enzymes, including urease. Staphylococcus aureus does express a nickel ABC transporter, Nik, which functions in metal-replete medium and is necessary for nickel urease activity and urinary tract colonization. In this work, we identified a novel cobalt and nickel transporter, which we named Cnt (previously annotated Opp1), in the major opportunistic pathogen S. aureus. Metal transport activity was revealed by growing cells in a chemically defined medium devoid of metals. Zinc specifically inhibits Cnt-mediated nickel and cobalt uptake, on both functional and transcriptional levels. Mortality due to S. aureus cnt mutant in systemic infection and colonization of the bladder and kidneys in ascending urinary tract infection model were reduced compared to the parent strain. This study identifies a novel S. aureus trace metal transporter and its restricted conditions of activity, and establishes its role in infection.
Collapse
Affiliation(s)
- Laetitia Remy
- UMR1319 Micalis, INRA, F-78350, Jouy en Josas, France
| | | | | | | | | | | |
Collapse
|
17
|
Kishor C, Gumpena R, Reddi R, Addlagatta A. Structural studies of Enterococcus faecalis methionine aminopeptidase and design of microbe specific 2,2′-bipyridine based inhibitors. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20096a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
18
|
Siwek A, Stączek P, Stefańska J. Synthesis and structure-activity relationship studies of 4-arylthiosemicarbazides as topoisomerase IV inhibitors with Gram-positive antibacterial activity. Search for molecular basis of antibacterial activity of thiosemicarbazides. Eur J Med Chem 2011; 46:5717-26. [PMID: 21978836 DOI: 10.1016/j.ejmech.2011.09.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/16/2011] [Accepted: 09/20/2011] [Indexed: 11/25/2022]
Abstract
1-(indol-2-carbonyl)-4-(4-nitrophenyl)-thiosemicarbazide was synthesized and antibacterial and type IIA topoisomerases (DNA gyrase and topoisomerase IV) activity was evaluated. It was found that it shows activity against Gram-positive bacteria with MICs of 50 μg/mL and inhibitory action against topoisomerase IV with an IC(50) of 14 μM. Although modification of its structure resulted in molecules with a lower biological profile, our observations strongly implicate that thiosemicarbazide derivatives participate in at least two different mechanisms of antibacterial activity; one is connected with the inhibition of topoisomerase IV, while the nature of the other cannot be elucidated from the limited data collected thus far. The differences in bioactivity further investigated by the molecular modeling approach and docking studies suggest that inhibitory activity of 4-arylthiosemicarbazides is connected with electronic structure rather than the geometry of the molecule.
Collapse
Affiliation(s)
- Agata Siwek
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University, Chodźki 4a, 20-093 Lublin, Poland.
| | | | | |
Collapse
|
19
|
Mucha A, Drag M, Dalton JP, Kafarski P. Metallo-aminopeptidase inhibitors. Biochimie 2010; 92:1509-29. [PMID: 20457213 PMCID: PMC7117057 DOI: 10.1016/j.biochi.2010.04.026] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 04/29/2010] [Indexed: 01/05/2023]
Abstract
Aminopeptidases are enzymes that selectively hydrolyze an amino acid residue from the N-terminus of proteins and peptides. They are important for the proper functioning of prokaryotic and eukaryotic cells, but very often are central players in the devastating human diseases like cancer, malaria and diabetes. The largest aminopeptidase group include enzymes containing metal ion(s) in their active centers, which often determines the type of inhibitors that are the most suitable for them. Effective ligands mostly bind in a non-covalent mode by forming complexes with the metal ion(s). Here, we present several approaches for the design of inhibitors for metallo-aminopeptidases. The optimized structures should be considered as potential leads in the drug discovery process against endogenous and infectious diseases.
Collapse
Affiliation(s)
- Artur Mucha
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| | | | | | | |
Collapse
|
20
|
Chai SC, Ye QZ. A cell-based assay that targets methionine aminopeptidase in a physiologically relevant environment. Bioorg Med Chem Lett 2010; 20:2129-32. [PMID: 20207144 PMCID: PMC2844763 DOI: 10.1016/j.bmcl.2010.02.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 02/10/2010] [Accepted: 02/10/2010] [Indexed: 11/28/2022]
Abstract
Methionine aminopeptidase (MetAP) is a promising target for the development of novel antibiotics. However, many potent inhibitors of the purified enzyme failed to show significant antibacterial activity. It is uncertain which divalent metal MetAP uses as its native cofactor in bacterial cells. Herein, we describe a cell-based assay that monitors the hydrolysis of a fluorogenic substrate by overexpressed MetAP in permeabilized Escherichia coli cells and its validation with a set of MetAP inhibitors. This cell-based assay is applicable to those cellular targets with poorly defined native cofactor, increasing the chances of identifying inhibitors that can inhibit the cellular target.
Collapse
Affiliation(s)
- Sergio C Chai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
21
|
Lu JP, Chai SC, Ye QZ. Catalysis and inhibition of Mycobacterium tuberculosis methionine aminopeptidase. J Med Chem 2010; 53:1329-37. [PMID: 20038112 PMCID: PMC2820511 DOI: 10.1021/jm901624n] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Methionine aminopeptidase (MetAP) carries out an important cotranslational N-terminal methionine excision of nascent proteins and represents a potential target to develop antibacterial and antitubercular drugs. We cloned one of the two MetAPs in Mycobacterium tuberculosis (MtMetAP1c from the mapB gene) and purified it to homogeneity as an apoenzyme. Its activity required a divalent metal ion, and Co(II), Ni(II), Mn(II), and Fe(II) were among activators of the enzyme. Co(II) and Fe(II) had the tightest binding, while Ni(II) was the most efficient cofactor for the catalysis. MtMetAP1c was also functional in E. coli cells because a plasmid-expressed MtMetAP1c complemented the essential function of MetAP in E. coli and supported the cell growth. A set of potent MtMetAP1c inhibitors were identified, and they showed high selectivity toward the Fe(II)-form, the Mn(II)-form, or the Co(II) and Ni(II) forms of the enzyme, respectively. These metalloform selective inhibitors were used to assign the metalloform of the cellular MtMetAP1c. The fact that only the Fe(II)-form selective inhibitors inhibited the cellular MtMetAP1c activity and inhibited the MtMetAP1c-complemented cell growth suggests that Fe(II) is the native metal used by MtMetAP1c in an E. coli cellular environment. Finally, X-ray structures of MtMetAP1c in complex with three metalloform-selective inhibitors were analyzed, which showed different binding modes and different interactions with metal ions and active site residues.
Collapse
Affiliation(s)
| | | | - Qi-Zhuang Ye
- To whom correspondence should be addressed: Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202. Tel.: 317-278-0304; Fax: 317-278-4686;
| |
Collapse
|
22
|
|
23
|
ALVARADO JOHNJEFF, NEMKAL ANJANA, SAUDER JMICHAEL, RUSSELL MARIJANE, AKIYOSHI DONNAE, SHI WUXIAN, ALMO STEVENC, WEISS LOUISM. Structure of a microsporidian methionine aminopeptidase type 2 complexed with fumagillin and TNP-470. Mol Biochem Parasitol 2009; 168:158-67. [PMID: 19660503 PMCID: PMC2759695 DOI: 10.1016/j.molbiopara.2009.07.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 07/21/2009] [Accepted: 07/26/2009] [Indexed: 01/14/2023]
Abstract
Microsporidia are protists that have been reported to cause infections in both vertebrates and invertebrates. They have emerged as human pathogens particularly in patients that are immunosuppressed and cases of gastrointestinal infection, encephalitis, keratitis, sinusitis, myositis and disseminated infection are well described in the literature. While benzimidazoles are active against many species of microsporidia, these drugs do not have significant activity against Enterocytozoon bieneusi. Fumagillin and its analogues have been demonstrated to have activity invitro and in animal models of microsporidiosis and human infections due to E. bieneusi. Fumagillin and its analogues inhibit methionine aminopeptidase type 2. Encephalitozoon cuniculi MetAP2 (EcMetAP2) was cloned and expressed as an active enzyme using a baculovirus system. The crystal structure of EcMetAP2 was determined with and without the bound inhibitors fumagillin and TNP-470. This structure classifies EcMetAP2 as a member of the MetAP2c family. The EcMetAP2 structure was used to generate a homology model of the E. bieneusi MetAP2. Comparison of microsporidian MetAP2 structures with human MetAP2 provides insights into the design of inhibitors that might exhibit specificity for microsporidian MetAP2.
Collapse
Affiliation(s)
- JOHN JEFF ALVARADO
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - ANJANA NEMKAL
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | - DONNA E. AKIYOSHI
- Division of Infectious Diseases, Tufts University School of Veterinary Medicine, Grafton, MA, USA
| | - WUXIAN SHI
- Center for Synchrotron Biosciences, Case Western Reserve University, Cleveland, OH, USA
| | - STEVEN C. ALMO
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - LOUIS M. WEISS
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
24
|
Olaleye OA, Bishai WR, Liu JO. Targeting the role of N-terminal methionine processing enzymes in Mycobacterium tuberculosis. Tuberculosis (Edinb) 2009; 89 Suppl 1:S55-9. [DOI: 10.1016/s1472-9792(09)70013-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
25
|
Chen X, Xie S, Bhat S, Kumar N, Shapiro TA, Liu JO. Fumagillin and fumarranol interact with P. falciparum methionine aminopeptidase 2 and inhibit malaria parasite growth in vitro and in vivo. ACTA ACUST UNITED AC 2009; 16:193-202. [PMID: 19246010 DOI: 10.1016/j.chembiol.2009.01.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 12/21/2008] [Accepted: 01/05/2009] [Indexed: 11/24/2022]
Abstract
The fumagillin family of natural products is known to inhibit angiogenesis through irreversible inhibition of human type 2 methionine aminopeptidase (MetAP2). Recently, fumagillin and TNP-470 were reported to possess antimalarial activity in vitro, and it was hypothesized that this inhibition was mediated by interaction with the putative malarial ortholog of human MetAP2. In this report, we have overexpressed and purified to near-homogeneity PfMetAP2 from bacteria, yeast, and insect cells. Although none of the recombinant forms of PfMetAP2 exhibited enzymatic activity in existing assays, PfMetAP2 proteins expressed in both yeast and insect cells were able to bind to fumagillin in a pull-down assay. The interaction between fumagillin and analogs with PfMetAP2 was further demonstrated using a newly established mammalian three-hybrid assay incorporating a conjugate between dexamethasone and fumagillin. Unlike human (Hs)MetAP2, it was found that PfMetAP2 is bound to fumagillin noncovalently. Importantly, a new analog of fumagillin, fumarranol, was demonstrated to interact with PfMetAP2 and inhibit the growth of both chloroquine-sensitive and drug-resistant Plasmodium falciparum strains in vitro. Antiparasite activity of fumagillin and fumarranol was also demonstrated in vivo using a mouse malaria model. These findings suggest that PfMetAP2 is a viable target, and fumarranol is a promising lead compound for the development of novel antimalarial agents.
Collapse
Affiliation(s)
- Xiaochun Chen
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
26
|
Mitra S, Sheppard G, Wang J, Bennett B, Holz RC. Analyzing the binding of Co(II)-specific inhibitors to the methionyl aminopeptidases from Escherichia coli and Pyrococcus furiosus. J Biol Inorg Chem 2009; 14:573-85. [PMID: 19198897 PMCID: PMC2678238 DOI: 10.1007/s00775-009-0471-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 12/31/2008] [Indexed: 10/21/2022]
Abstract
Methionine aminopeptidases (MetAPs) represent a unique class of protease that is capable of the hydrolytic removal of an N-terminal methionine residue from nascent polypeptide chains. MetAPs are physiologically important enzymes; hence, there is considerable interest in developing inhibitors that can be used as antiangiogenic and antimicrobial agents. A detailed kinetic and spectroscopic study has been performed to probe the binding of a triazole-based inhibitor and a bestatin-based inhibitor to both Mn(II)- and Co(II)-loaded type-I (Escherichia coli) and type-II (Pyrococcus furiosus) MetAPs. Both inhibitors were found to be moderate competitive inhibitors. The triazole-type inhibitor was found to interact with both active-site metal ions, while the bestatin-type inhibitor was capable of switching its mode of binding depending on the metal in the active site and the type of MetAP enzyme.
Collapse
Affiliation(s)
- Sanghamitra Mitra
- Department of Chemistry, Loyola University-Chicago, Chicago, IL 60626, USA
| | | | | | | | | |
Collapse
|
27
|
Wang WL, Chai SC, Ye QZ. Synthesis and structure-function analysis of Fe(II)-form-selective antibacterial inhibitors of Escherichia coli methionine aminopeptidase. Bioorg Med Chem Lett 2009; 19:1080-3. [PMID: 19167218 PMCID: PMC2643329 DOI: 10.1016/j.bmcl.2009.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 01/05/2009] [Accepted: 01/06/2009] [Indexed: 11/18/2022]
Abstract
Methionine aminopeptidase (MetAP) is a promising target for the development of novel antibacterial, antifungal and anticancer therapy. Based on our previous results, catechol derivatives coupled with a thiazole or thiophene moiety showed high potency and selectivity toward the Fe(II)-form of Escherichia coli MetAP, and some of them clearly showed antibacterial activity, indicating that Fe(II) is likely the physiologically relevant metal for MetAP in E. coli and other bacterial cells. To further understand the structure-function relationship of these Fe(II)-form selective MetAP inhibitors, a series of catechol derivatives was designed and synthesized by replacement of the thiazole or thiophene moiety with different five-membered and six-membered heterocycles. Inhibitory activities of these newly synthesized MetAP inhibitors indicate that many five- and six-membered rings can be accommodated by MetAP and potency on the Fe(II)-form can be improved by introducing substitutions on the heterocyles to explore additional interactions with the enzyme. The furan-containing catechols 11-13 showed the highest potency at 1muM on the Fe(II)-form MetAP, and they were also among the best inhibitors for growth inhibition against E. coli AS19 strain. These findings provide useful information for the design and discovery of more effective MetAP inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Wen-Long Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Sergio C. Chai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Qi-Zhuang Ye
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| |
Collapse
|
28
|
Mitra S, Bennett B, Holz RC. Mutation of H63 and its catalytic affect on the methionine aminopeptidase from Escherichia coli. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1794:137-43. [PMID: 18952013 PMCID: PMC2674292 DOI: 10.1016/j.bbapap.2008.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 09/11/2008] [Accepted: 09/13/2008] [Indexed: 05/27/2023]
Abstract
In order to gain insight into the mechanistic role of a flexible exterior loop near the active site, made up of Y62, H63, G64, and Y65, that has been proposed to play an important role in substrate binding and recognition in the methionyl aminopeptidase from Escherichia coli (EcMetAP-I), the H63A enzyme was prepared. Mutation of H63 to alanine does not affect the ability of the enzyme to bind divalent metal ions. The specific activity of H63A EcMetAP-I was determined using four different substrates of varying lengths, namely, l-Met-p-NA, MAS, MGMM and MSSHRWDW. For the smallest/shortest substrate (l-Met-p-NA) the specific activity decreased nearly seven fold but as the peptide length increased, the specific activity also increased and became comparable to WT EcMetAP-I. This decrease in specific activity is primarily due to a decrease in the observed k(cat) values, which decreases nearly sixty-fold for l-Met-p-NA while only a four-fold decrease is observed for the tri- and tetra-peptide substrates. Interestingly, no change in k(cat) was observed when the octa-peptide MSSHRWDW was used as a substrate. These data suggest that H63 affects the hydrolysis of small peptide substrates whereas large peptides can overcome the observed loss in binding energy, as predicted from K(m) values, by additional hydrophilic and hydrophobic interactions.
Collapse
Affiliation(s)
- Sanghamitra Mitra
- Department of Chemistry, Boston University, Boston, MA 02215-2521, USA
| | | | | |
Collapse
|
29
|
Mitra S, Job KM, Meng L, Bennett B, Holz RC. Analyzing the catalytic role of Asp97 in the methionine aminopeptidase from Escherichia coli. FEBS J 2008; 275:6248-59. [PMID: 19019076 PMCID: PMC2699115 DOI: 10.1111/j.1742-4658.2008.06749.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An active site aspartate residue, Asp97, in the methionine aminopeptidase (MetAPs) from Escherichia coli (EcMetAP-I) was mutated to alanine, glutamate, and asparagine. Asp97 is the lone carboxylate residue bound to the crystallographically determined second metal-binding site in EcMetAP-I. These mutant EcMetAP-I enzymes have been kinetically and spectroscopically characterized. Inductively coupled plasma-atomic emission spectroscopy analysis revealed that 1.0 +/- 0.1 equivalents of cobalt were associated with each of the Asp97-mutated EcMetAP-Is. The effect on activity after altering Asp97 to alanine, glutamate or asparagine is, in general, due to a approximately 9000-fold decrease in k(ca) towards Met-Gly-Met-Met as compared to the wild-type enzyme. The Co(II) d-d spectra for wild-type, D97E and D97A EcMetAP-I exhibited very little difference in form, in each case, between the monocobalt(II) and dicobalt(II) EcMetAP-I, and only a doubling of intensity was observed upon addition of a second Co(II) ion. In contrast, the electronic absorption spectra of [Co_(D97N EcMetAP-I)] and [CoCo(D97N EcMetAP-I)] were distinct, as were the EPR spectra. On the basis of the observed molar absorptivities, the Co(II) ions binding to the D97E, D97A and D97N EcMetAP-I active sites are pentacoordinate. Combination of these data suggests that mutating the only nonbridging ligand in the second divalent metal-binding site in MetAPs to an alanine, which effectively removes the ability of the enzyme to form a dinuclear site, provides a MetAP enzyme that retains catalytic activity, albeit at extremely low levels. Although mononuclear MetAPs are active, the physiologically relevant form of the enzyme is probably dinuclear, given that the majority of the data reported to date are consistent with weak cooperative binding.
Collapse
Affiliation(s)
- Sanghamitra Mitra
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | | | | | | | | |
Collapse
|
30
|
Chai SC, Wang WL, Ye QZ. FE(II) is the native cofactor for Escherichia coli methionine aminopeptidase. J Biol Chem 2008; 283:26879-85. [PMID: 18669631 DOI: 10.1074/jbc.m804345200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Divalent metal ions play a critical role in the removal of N-terminal methionine from nascent proteins by methionine aminopeptidase (MetAP). Being an essential enzyme for bacteria, MetAP is an appealing target for the development of novel antibacterial drugs. Although purified enzyme can be activated by several divalent metal ions, the exact metal ion used by MetAP in cells is unknown. Many MetAP inhibitors are highly potent on purified enzyme, but they fail to show significant inhibition of bacterial growth. One possibility for the failure is a disparity of the metal used in activation of purified MetAP and the metal actually used by MetAP inside bacterial cells. Therefore, the challenge is to elucidate the physiologically relevant metal for MetAP and discover MetAP inhibitors that can effectively inhibit cellular MetAP. We have recently discovered MetAP inhibitors with selectivity toward different metalloforms of Escherichia coli MetAP, and with these unique inhibitors, we characterized their inhibition of MetAP enzyme activity in a cellular environment. We observed that only inhibitors that are selective for the Fe(II)-form of MetAP were potent in this assay. Further, we found that only these Fe(II)-form selective inhibitors showed significant inhibition of growth of five E. coli strains and two Bacillus strains. We confirmed their cellular target as MetAP by analysis of N-terminal processed and unprocessed recombinant glutathione S-transferase proteins. Therefore, we conclude that Fe(II) is the likely metal used by MetAP in E. coli and other bacterial cells.
Collapse
Affiliation(s)
- Sergio C Chai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
31
|
Babor M, Gerzon S, Raveh B, Sobolev V, Edelman M. Prediction of transition metal-binding sites from apo protein structures. Proteins 2008; 70:208-17. [PMID: 17657805 DOI: 10.1002/prot.21587] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Metal ions are crucial for protein function. They participate in enzyme catalysis, play regulatory roles, and help maintain protein structure. Current tools for predicting metal-protein interactions are based on proteins crystallized with their metal ions present (holo forms). However, a majority of resolved structures are free of metal ions (apo forms). Moreover, metal binding is a dynamic process, often involving conformational rearrangement of the binding pocket. Thus, effective predictions need to be based on the structure of the apo state. Here, we report an approach that identifies transition metal-binding sites in apo forms with a resulting selectivity >95%. Applying the approach to apo forms in the Protein Data Bank and structural genomics initiative identifies a large number of previously unknown, putative metal-binding sites, and their amino acid residues, in some cases providing a first clue to the function of the protein.
Collapse
Affiliation(s)
- Mariana Babor
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | |
Collapse
|
32
|
Vedantham P, Zhang M, Gor PJ, Huang M, Georg GI, Lushington GH, Mitscher LA, Ye QZ, Hanson PR. Studies towards the synthesis of methionine aminopeptidase inhibitors: diversification utilizing a ROMP-derived coupling reagent. ACTA ACUST UNITED AC 2007; 10:195-203. [PMID: 18163594 DOI: 10.1021/cc7000869] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Efforts to synthesize potential methionine aminopeptidase inhibitors is described. Preliminary SAR and docking studies served as a guide to design the compound libraries. "Chromatography-free" synthesis of various heterocyclic amides was realized by using a high-load, soluble coupling reagent derived via ring-opening metathesis polymerization (ROMP). Subsequent microwave-assisted Suzuki reactions with ortho-substituted arylboronic acids, followed by chromatographic purification afforded a 55-member library in high yields and purities. While the biological testing was not satisfactory, concurrent X-ray crystallography studies revealed key structural features essential for inhibition of methionine aminopeptidase, which directed fruitful results reported in the accompanying manuscript. In addition, in silico Lipinksi profiles and ADME properties of the library are also reported.
Collapse
Affiliation(s)
- Punitha Vedantham
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ma ZQ, Xie SX, Huang QQ, Nan FJ, Hurley TD, Ye QZ. Structural analysis of inhibition of E. coli methionine aminopeptidase: implication of loop adaptability in selective inhibition of bacterial enzymes. BMC STRUCTURAL BIOLOGY 2007; 7:84. [PMID: 18093325 PMCID: PMC2238726 DOI: 10.1186/1472-6807-7-84] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 12/19/2007] [Indexed: 11/17/2022]
Abstract
Background Methionine aminopeptidase is a potential target of future antibacterial and anticancer drugs. Structural analysis of complexes of the enzyme with its inhibitors provides valuable information for structure-based drug design efforts. Results Five new X-ray structures of such enzyme-inhibitor complexes were obtained. Analysis of these and other three similar structures reveals the adaptability of a surface-exposed loop bearing Y62, H63, G64 and Y65 (the YHGY loop) that is an integral part of the substrate and inhibitor binding pocket. This adaptability is important for accommodating inhibitors with variations in size. When compared with the human isozymes, this loop either becomes buried in the human type I enzyme due to an N-terminal extension that covers its position or is replaced by a unique insert in the human type II enzyme. Conclusion The adaptability of the YHGY loop in E. coli methionine aminopeptidase, and likely in other bacterial methionine aminopeptidases, enables the enzyme active pocket to accommodate inhibitors of differing size. The differences in this adaptable loop between the bacterial and human methionine aminopeptidases is a structural feature that can be exploited to design inhibitors of bacterial methionine aminopeptidases as therapeutic agents with minimal inhibition of the corresponding human enzymes.
Collapse
Affiliation(s)
- Ze-Qiang Ma
- High Throughput Screening Laboratory, University of Kansas, Lawrence, Kansas 66047, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Huang M, Xie SX, Ma ZQ, Huang QQ, Nan FJ, Ye QZ. Inhibition of monometalated methionine aminopeptidase: inhibitor discovery and crystallographic analysis. J Med Chem 2007; 50:5735-42. [PMID: 17948983 PMCID: PMC2522305 DOI: 10.1021/jm700930k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two divalent metal ions are commonly seen in the active-site cavity of methionine aminopeptidase, and at least one of the metal ions is directly involved in catalysis. Although ample structural and functional information is available for dimetalated enzyme, methionine aminopeptidase likely functions as a monometalated enzyme under physiological conditions. Information on structure, as well as catalysis and inhibition, of the monometalated enzyme is lacking. By improving conditions of high-throughput screening, we identified a unique inhibitor with specificity toward the monometalated enzyme. Kinetic characterization indicates a mutual exclusivity in binding between the inhibitor and the second metal ion at the active site. This is confirmed by X-ray structure, and this inhibitor coordinates with the first metal ion and occupies the space normally occupied by the second metal ion. Kinetic and structural analyses of the inhibition by this and other inhibitors provide insight in designing effective inhibitors of methionine aminopeptidase.
Collapse
Affiliation(s)
| | | | | | | | | | - Qi-Zhuang Ye
- * To whom correspondence should be addressed. Phone: 317 278-0304. Fax: 317 274-4686. E-Mail:
| |
Collapse
|
35
|
Hu XV, Chen X, Han KC, Mildvan AS, Liu JO. Kinetic and Mutational Studies of the Number of Interacting Divalent Cations Required by Bacterial and Human Methionine Aminopeptidases. Biochemistry 2007; 46:12833-43. [DOI: 10.1021/bi701127x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaoyi V. Hu
- Departments of Pharmacology and Molecular Sciences and Biological Chemistry and Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205
| | - Xiaochun Chen
- Departments of Pharmacology and Molecular Sciences and Biological Chemistry and Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205
| | - Kee Chung Han
- Departments of Pharmacology and Molecular Sciences and Biological Chemistry and Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205
| | - Albert S. Mildvan
- Departments of Pharmacology and Molecular Sciences and Biological Chemistry and Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205
| | - Jun O. Liu
- Departments of Pharmacology and Molecular Sciences and Biological Chemistry and Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205
| |
Collapse
|
36
|
Marino JP, Fisher PW, Hofmann GA, Kirkpatrick RB, Janson CA, Johnson RK, Ma C, Mattern M, Meek TD, Ryan MD, Schulz C, Smith WW, Tew DG, Tomazek TA, Veber DF, Xiong WC, Yamamoto Y, Yamashita K, Yang G, Thompson SK. Highly Potent Inhibitors of Methionine Aminopeptidase-2 Based on a 1,2,4-Triazole Pharmacophore. J Med Chem 2007; 50:3777-85. [PMID: 17636946 DOI: 10.1021/jm061182w] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
High-throughput screening for inhibitors of the human metalloprotease, methionine aminopeptidase-2 (MetAP2), identified a potent class of 3-anilino-5-benzylthio-1,2,4-triazole compounds. Efficient array and interative synthesis of triazoles led to rapid SAR development around the aniline, benzylthio, and triazole moeities. Evaluation of these analogs in a human MetAP2 enzyme assay led to the identification of several inhibitors with potencies in the 50-100 picomolar range. The deleterious effects on inhibitor potency by methylation of the anilino-triazole nitrogens, as well as the X-ray crystal structure of triazole 102 bound in the active site of MetAP2, confirm the key interactions between the triazole nitrogens, the active site cobalt atoms, and the His-231 side-chain. The structure has also provided a rationale for interpreting SAR within the triazole series. Key aniline (2-isopropylphenyl) and sulfur substituents (furanylmethyl) identified in the SAR studies led to the identification of potent inhibitors (103 and 104) of endothelial cell proliferation. Triazoles 103 and 104 also exhibited dose-dependent activity in an aortic ring tissue model of angiogenesis highlighting the potential utility of MetAP2 inhibitors as anticancer agents.
Collapse
Affiliation(s)
- Joseph P Marino
- Department of Medicinal Chemistry, Enzymology, Oncology, and Structural Biology, GlaxoSmithkline, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wang GT, Mantei RA, Kawai M, Tedrow JS, Barnes DM, Wang J, Zhang Q, Lou P, Garcia LA, Bouska J, Yates M, Park C, Judge RA, Lesniewski R, Sheppard GS, Bell RL. Lead optimization of methionine aminopeptidase-2 (MetAP2) inhibitors containing sulfonamides of 5,6-disubstituted anthranilic acids. Bioorg Med Chem Lett 2007; 17:2817-22. [PMID: 17350258 DOI: 10.1016/j.bmcl.2007.02.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 02/16/2007] [Accepted: 02/22/2007] [Indexed: 11/30/2022]
Abstract
A series of aryl sulfonamides of 5,6-disubstituted anthranilic acids were identified as potent inhibitors of methionine aminopeptidase-2 (MetAP2). Small alkyl groups and 3-furyl were tolerated at the 5-position of anthranilic acid, while -OCH(3), CH(3), and Cl were found optimal for the 6-position. Placement of 2-aminoethoxy group at the 6-position enabled interaction with the second Mn(2+) but did not result in enhancement in potency. Introduction of a tertiary amino moiety at the ortho-position of the sulfonyl phenyl ring gave reduced protein binding and improved cellular activity, but led to lower oral bioavailability.
Collapse
Affiliation(s)
- Gary T Wang
- Cancer Research, Global Pharmaceutical Research & Development, Abbott Laboratories, Abbott Park, IL 60064, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Evdokimov AG, Pokross M, Walter RL, Mekel M, Barnett BL, Amburgey J, Seibel WL, Soper SJ, Djung JF, Fairweather N, Diven C, Rastogi V, Grinius L, Klanke C, Siehnel R, Twinem T, Andrews R, Curnow A. Serendipitous discovery of novel bacterial methionine aminopeptidase inhibitors. Proteins 2007; 66:538-46. [PMID: 17120228 DOI: 10.1002/prot.21207] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this article we describe the application of structural biology methods to the discovery of novel potent inhibitors of methionine aminopeptidases. These enzymes are employed by the cells to cleave the N-terminal methionine from nascent peptides and proteins. As this is one of the critical steps in protein maturation, it is very likely that inhibitors of these enzymes may prove useful as novel antibacterial agents. Involvement of crystallography at the very early stages of the inhibitor design process resulted in serendipitous discovery of a new inhibitor class, the pyrazole-diamines. Atomic-resolution structures of several inhibitors bound to the enzyme illuminate a new mode of inhibitor binding.
Collapse
Affiliation(s)
- Artem G Evdokimov
- Structural Biology Core Facility, The Procter & Gamble Pharmaceuticals, Mason, Ohio 45040, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lee HS, Kim YJ, Bae SS, Jeon JH, Lim JK, Jeong BC, Kang SG, Lee JH. Cloning, expression, and characterization of a methionyl aminopeptidase from a hyperthermophilic archaeon Thermococcus sp. NA1. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2006; 8:425-32. [PMID: 16761197 DOI: 10.1007/s10126-005-6124-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Accepted: 01/13/2006] [Indexed: 05/10/2023]
Abstract
Genomic analysis of a hyperthermophilic archaeon Thermococcus sp. NA1 revealed the presence of an 885-bp open reading frame encoding a protein of 295 amino acids with a calculated molecular mass of 32,981 Da. Analysis of the deduced amino acid sequence showed that amino acid residues important for catalytic activity and the metal binding ligands conserved in all of methionyl aminopeptidases (MetAP) were also conserved and belonged to type IIa MetAP. The protein, designated TNA1_MetAP (Thermococcus sp. NA1 MetAP), was cloned and expressed in Escherichia coli. The recombinant enzyme was a Mn(2+)-, Ni(2+)-, Fe(2+)-, or Co(2+)-dependent metallopeptidase. Optimal MetAP activity against L: -methionine p-nitroanilide (Met-pNA) (K (m) = 0.68 mM) occurred at pH 7.0 and 80 to 90 degrees C. The MetAP was very unstable compared to Pyrococcus furiosus MetAP, which was completely inactivated by heating at 80 degrees C for 5 min. It seemed likely that the cysteine residue (Cys53) played a critical role in regulating the thermostability of TNA1_MetAP.
Collapse
Affiliation(s)
- H S Lee
- Korean Ocean Research & Development Institute, Ansan, P.O. Box 29, Seoul, 425-600, Korea.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Huang LF, Su B, Jao SC, Liu KT, Li WS. Aminopeptidase p mediated detoxification of organophosphonate analogues of sarin: mechanistic and stereochemical study at the phosphorus atom of the substrate. Chembiochem 2006; 7:506-14. [PMID: 16470765 DOI: 10.1002/cbic.200500412] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The activity of the aminopeptidase P from Escherichia coli in hydrolyzing a series of organophosphonate sarin analogues (1-6) was evaluated. The enzymatic rates of hydrolysis for methylphosphonate 1 with a methoxy group attached to the phosphorus center were 7- to 15-fold higher than those for the corresponding analogues 2-6. Double mutant R153W/R370L was able to hydrolyze the S(p) enantiomer of racemic 1 at a considerable rate. This mutant allowed the preparation of the R(p) isomer of the sarin analogue 1. All the mutants, R370L, R153A, W88L, R153L/R370L, and R153W/R370L, preferred the formation of (S(p))-8 to that of the corresponding (R(p))-8 enantiomer and displayed a better enantiomeric excess of products, by 1.4- to 2-fold as compared to the wild-type enzyme. Enzymatic hydrolysis of O,O-diisopropyl-p-nitrophenyl phosphate (9) in H(2) (18)O led to the formation of the (18)O-labeled O,O-diisopropyl phosphate product and confirmed that the catalytic reaction starts with cleavage of the P--O bond. From chemical and kinetic studies, the utilization of an optically pure S(p) enantiomer of O-methyl-p-nitrophenyl methylphosphonothioate (S(p))-MNMPT, 7) has demonstrated that the enzymatic reaction proceeds through a displacement mechanism and generates a chiral product in situ with an inversion of stereochemical configuration at the phosphorus atom. The results also lead to the conclusion that alteration of the active site through site-directed mutagenesis can result in a preference for (S(p))-MNMPT (7) rather than the R(p) isomer.
Collapse
Affiliation(s)
- Li-Fang Huang
- Institute of Chemistry, Academia Sinica, 128 Academia Road Sec. 2, Taipei 115, Taiwan
| | | | | | | | | |
Collapse
|
41
|
Ye QZ, Xie SX, Ma ZQ, Huang M, Hanzlik RP. Structural basis of catalysis by monometalated methionine aminopeptidase. Proc Natl Acad Sci U S A 2006; 103:9470-5. [PMID: 16769889 PMCID: PMC1480431 DOI: 10.1073/pnas.0602433103] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Methionine aminopeptidase (MetAP) removes the amino-terminal methionine residue from newly synthesized proteins, and it is a target for the development of antibacterial and anticancer agents. Available x-ray structures of MetAP, as well as other metalloaminopeptidases, show an active site containing two adjacent divalent metal ions bridged by a water molecule or hydroxide ion. The predominance of dimetalated structures leads naturally to proposed mechanisms of catalysis involving both metal ions. However, kinetic studies indicate that in many cases, only a single metal ion is required for full activity. By limiting the amount of metal ion present during crystal growth, we have now obtained a crystal structure for a complex of Escherichia coli MetAP with norleucine phosphonate, a transition-state analog, and only a single Mn(II) ion bound at the active site in the position designated M1, and three related structures of the same complex that show the transition from the mono-Mn(II) form to the di-Mn(II) form. An unliganded structure was also solved. In view of the full kinetic competence of the monometalated MetAP, the much weaker binding constant for occupancy of the M2 site compared with the M1 site, and the newly determined structures, we propose a revised mechanism of peptide bond hydrolysis by E. coli MetAP. We also suggest that the crystallization of dimetalated forms of metallohydrolases may, in some cases, be a misleading experimental artifact, and caution must be taken when structures are generated to aid in elucidation of reaction mechanisms or to support structure-aided drug design efforts.
Collapse
Affiliation(s)
- Qi-Zhuang Ye
- High Throughput Screening Laboratory and Department of Medicinal Chemistry, University of Kansas, 1501 Wakarusa Drive, Lawrence, KS 66045, USA.
| | | | | | | | | |
Collapse
|
42
|
Jao SC, Huang LF, Hwang SM, Li WS. Tyrosine 387 and arginine 404 are critical in the hydrolytic mechanism of Escherichia coli aminopeptidase P. Biochemistry 2006; 45:1547-53. [PMID: 16460001 DOI: 10.1021/bi051786m] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Analysis of the pH-rate profile for catalysis of bradykinin cleavage by aminopeptidase P (AMPP), a manganese-containing hydrolase from Escherichia coli, was carried out to show that optimal catalytic function is obtained at neutral pH. On the basis of information derived from the crystal structure, peptidase sequence alignments, and the hydrolysis of organophosphate triesters, active site residues Arg153, Arg370, Trp88, Tyr387, and Arg404 were identified as potential catalytic residues. Site-directed mutagenesis was used to substitute these residues with Leu, Ala, Trp, Lys, or Phe. The kcat values for the Arg153, Arg370, and Trp88 mutants were nearly the same as that for the wild-type enzyme. The kcat values of the R404K, R404A, and Y387A mutants were lower by factors of 285, 400, and 16, respectively. Inductively coupled plasma mass spectrometry and circular dichroism spectroscopy showed that Arg404 is not required for metal chelation or stabilization of protein secondary structure. The hydrogen bond network observed between the side chains of conserved residues Asp260, Arg404, and Tyr387 indicated that Arg404 participates in proton relay. This was further evidenced by the return of activity in the R404A mutant by the addition of guanidine. Also, reduced catalytic efficiency in the R404K mutant, which conserves the positive charge at the bridge site, shows that only the arginine group of Arg404 (not the ammonium group of Lys404) can participate in the hydrogen bond network. The hydrogen bond interaction between the Arg404 and the Tyr387 ring hydroxyl group is suggested by the reduced catalytic efficiency of the Y387F mutant.
Collapse
Affiliation(s)
- Shu-Chuan Jao
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | | | | | | |
Collapse
|
43
|
Schiffmann R, Neugebauer A, Klein CD. Metal-Mediated Inhibition of Escherichia coli Methionine Aminopeptidase: Structure−Activity Relationships and Development of a Novel Scoring Function for Metal−Ligand Interactions. J Med Chem 2005; 49:511-22. [PMID: 16420038 DOI: 10.1021/jm050476z] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report the discovery of thiabendazole as a potent inhibitor (K(i) = 0.4 microM) of Escherichia coli methionine aminopeptidase (ecMetAP) and the synthesis and pharmacological evaluation of thiabendazole congeners with activity in the upper nanomolar range. Elucidation of the X-ray structure of ecMetAP in complex with thiabendazole and an unrelated inhibitor that was independently described by another group showed that that both compounds bind to an additional Co(II) ion at the entrance of the active site. This unexpected finding explains the inactivity of the compounds under in vivo conditions. It also allows us to discuss the structure-activity relationships of this series of compounds in a meaningful way, based upon docking runs with an auxiliary metal ion. We describe a new scoring function for the evaluation of metal-mediated inhibitor binding that, unlike the previously used scoring function implemented in the docking program, allows us to distinguish between active and inactive compounds. Finally, conclusions for the structure-based design of in vivo-active inhibitors of ecMetAP are drawn.
Collapse
Affiliation(s)
- Rolf Schiffmann
- Saarland University, Pharmaceutical and Medicinal Chemistry, D-66041 Saarbrücken, Germany
| | | | | |
Collapse
|
44
|
Huang M, Xie SX, Ma ZQ, Hanzlik RP, Ye QZ. Metal mediated inhibition of methionine aminopeptidase by quinolinyl sulfonamides. Biochem Biophys Res Commun 2005; 339:506-13. [PMID: 16300729 DOI: 10.1016/j.bbrc.2005.11.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Accepted: 11/06/2005] [Indexed: 11/28/2022]
Abstract
Quinolinyl sulfonamides, such as N-(quinolin-8-yl)methanesulfonamide (10) and N-(5-chloroquinolin-8-yl)methanesulfonamide (11), were identified as potent methionine aminopeptidase (MetAP) inhibitors by high throughput screening of a diverse chemical library of small organic compounds. They showed different inhibitory potencies on Co(II)-, Ni(II)-, Fe(II)-, Mn(II)-, and Zn(II)-forms of Escherichia coli MetAP, and their inhibition is dependent on metal concentration. X-ray structures of E. coli MetAP complexed with 10 revealed that the inhibitor forms a metal complex with the residue H79 at the enzyme active site; the complex is further stabilized by an extended H-bond and metal interaction network. Analysis of the inhibition of MetAP by these inhibitors indicates that this is a typical mechanism of inhibition for many non-peptidic MetAP inhibitors and emphasizes the importance of defining in vitro conditions for identifying and evaluating MetAP inhibitors that will be capable of giving information relevant to the in vivo situation.
Collapse
Affiliation(s)
- Min Huang
- High Throughput Screening Laboratory, University of Kansas, Lawrence, KS 66045, USA
| | | | | | | | | |
Collapse
|
45
|
Kallander LS, Lu Q, Chen W, Tomaszek T, Yang G, Tew D, Meek TD, Hofmann GA, Schulz-Pritchard CK, Smith WW, Janson CA, Ryan MD, Zhang GF, Johanson KO, Kirkpatrick RB, Ho TF, Fisher PW, Mattern MR, Johnson RK, Hansbury MJ, Winkler JD, Ward KW, Veber DF, Thompson SK. 4-Aryl-1,2,3-triazole: A Novel Template for a Reversible Methionine Aminopeptidase 2 Inhibitor, Optimized To Inhibit Angiogenesis in Vivo. J Med Chem 2005; 48:5644-7. [PMID: 16134930 DOI: 10.1021/jm050408c] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Inhibitors of human methionine aminopeptidase type 2 (hMetAP2) are of interest as potential treatments for cancer. A new class of small molecule reversible inhibitors of hMetAP2 was discovered and optimized, the 4-aryl-1,2,3-triazoles. Compound 24, a potent inhibitor of cobalt-activated hMetAP2, also inhibits human and mouse endothelial cell growth. Using a mouse matrigel model, this reversible hMetAP2 inhibitor was also shown to inhibit angiogenesis in vivo.
Collapse
Affiliation(s)
- Lara S Kallander
- GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania 19406, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cui YM, Huang QQ, Xu J, Chen LL, Li JY, Ye QZ, Li J, Nan FJ. Identification of potent type I MetAP inhibitors by simple bioisosteric replacement. Part 1: Synthesis and preliminary SAR studies of thiazole-4-carboxylic acid thiazol-2-ylamide derivatives. Bioorg Med Chem Lett 2005; 15:3732-6. [PMID: 15993057 DOI: 10.1016/j.bmcl.2005.05.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Revised: 05/14/2005] [Accepted: 05/18/2005] [Indexed: 10/25/2022]
Abstract
A series of thiazole-4-carboxylic acid thiazol-2-ylamide (TCAT, 4) derivatives were designed and synthesized according to simple bioisosteric replacement from previously reported pyridine-2-carboxylic acid thiazol-2-ylamide (PCAT) MetAP inhibitors. The preliminary SAR studies demonstrated that these TCAT series of compounds showed different activity and selectivity compared with those of the corresponding PCAT compounds. These findings provide useful information for the design and discovery of more potent inhibitors of type I MetAPs.
Collapse
Affiliation(s)
- Yong-Mei Cui
- Chinese National Center for Drug Screening, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Addlagatta A, Quillin ML, Omotoso O, Liu JO, Matthews BW. Identification of an SH3-Binding Motif in a New Class of Methionine Aminopeptidases from Mycobacterium tuberculosis Suggests a Mode of Interaction with the Ribosome,. Biochemistry 2005; 44:7166-74. [PMID: 15882055 DOI: 10.1021/bi0501176] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The crystal structure of the methionine aminopeptidase (MetAP) from Mycobacterium tuberculosis (MtMetAP1c) has been determined in the apo- and methionine-bound forms. This is the first structure of a type I MetAP with a significant extension at the amino terminus. The catalytic domain is similar to that of Escherichia coli MetAP (EcMetAP), and the additional 40-residue segment wraps around the surface with an extended but well-defined structure. There are several members of the actinomyces family of bacteria that contain MetAPs with such N-terminal extensions, and we classify these as MetAP type Ic (MetAP1c). Some members of this family of bacteria also contain a second MetAP (type Ia) similar in size to EcMetAP. The main difference between the apo- and the methionine-bound forms of MtMetAP1c is in the conformation of the metal-binding residues. The position of the methionine bound in the active site is very similar to that found in many of the known members of this family. Side chains of several residues in the S1 and S1' subsites shift as much as 1.5 A compared to EcMetAP. Residues 14-17 have the sequence Pro-Thr-Arg-Pro and adopt the conformation of a polyproline II helix. Model-building suggests that this PxxP segment can bind to an SH3 protein motif. Other type Ib and type Ic MetAPs with N-terminal extensions contain similarly located PxxP motifs. Also, several ribosomal proteins are known to include SH3 domains, one of which is located close to the tunnel from which the nascent polypeptide chain exits the ribosome. Therefore, it is proposed that the binding of MetAPs to the ribosome is mediated by a complex between a PxxP motif on the protein and an SH3 domain on the ribosome. It is also possible that zinc-finger domains, which are located at the extreme N-terminus of type I MetAPs, may participate in interactions with the ribosome.
Collapse
Affiliation(s)
- Anthony Addlagatta
- Institute of Molecular Biology, Howard Hughes Medical Institute and Department of Physics, 1229 University of Oregon, Eugene, Oregon 97403-1229, USA
| | | | | | | | | |
Collapse
|
48
|
Copik AJ, Nocek BP, Swierczek SI, Ruebush S, Jang SB, Meng L, D'souza VM, Peters JW, Bennett B, Holz RC. EPR and X-ray crystallographic characterization of the product-bound form of the MnII-loaded methionyl aminopeptidase from Pyrococcus furiosus. Biochemistry 2005; 44:121-9. [PMID: 15628852 DOI: 10.1021/bi048123+] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Methionine aminopeptidases (MetAPs) are ubiquitous metallohydrolases that remove the N-terminal methionine from nascent polypeptide chains. Although various crystal structures of MetAP in the presence of inhibitors have been solved, the structural aspects of the product-bound step has received little attention. Both perpendicular- and parallel-mode electron paramagnetic resonance (EPR) spectra were recorded for the Mn(II)-loaded forms of the type-I (Escherichia coli) and type-II (Pyrococcus furiosus) MetAPs in the presence of the reaction product l-methionine (L-Met). In general, similar EPR features were observed for both [MnMn(EcMetAP-I)]-L-Met and [MnMn(PfMetAP-II)]-L-Met. The observed perpendicular-mode EPR spectra consisted of a six-line hyperfine pattern at g = 2.03 (A = 8.8 mT) with less intense signals with eleven-line splitting at g = 2.4 and 1.7 (A = 4.4 mT). The former feature results from mononuclear, magnetically isolated Mn(II) ions and this signal are 3-fold more intense in the [MnMn(PfMetAP-II)]-L-Met EPR spectrum than in the [MnMn(EcMetAP-I)]-L-Met spectrum. Inspection of the EPR spectra of both [MnMn(EcMetAP-I)]-L-Met and [MnMn(PfMetAP-II)]-L-Met at 40 K in the parallel mode reveals that the [Mn(EcMetAP-I)]-L-Met spectrum exhibits a well-resolved hyperfine split pattern at g = 7.6 with a hyperfine splitting constant of A = 4.4 mT. These data suggest the presence of a magnetically coupled dinuclear Mn(II) center. On the other hand, a similar feature was not observed for the [MnMn(PfMetAP-II)]-L-Met complex. Therefore, the EPR data suggest that L-Met binds to [MnMn(EcMetAP-I)] differently than [MnMn(PfMetAP-II)]. To confirm these data, the X-ray crystal structure of [MnMn(PfMetAP-II)]-L-Met was solved to 2.3 A resolution. Both Mn1 and Mn2 reside in a distorted trigonal bipyramidal geometry, but the bridging water molecule, observed in the [CoCo(PfMetAP-II)] structure, is absent. Therefore, L-Met binding displaces this water molecule, but the carboxylate oxygen atom of L-Met does not bridge between the two Mn(II) ions. Instead, a single carboxylate oxygen atom of L-Met interacts with only Mn1, while the N-terminal amine nitrogen atom binds to M2. This L-Met binding mode is different from that observed for L-Met binding [CoCo(EcMetAP-I)]. Therefore, the catalytic mechanisms of type-I MetAPs may differ somewhat from type-II enzymes when a dinuclear metalloactive site is present.
Collapse
Affiliation(s)
- Alicja J Copik
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ye QZ, Xie SX, Huang M, Huang WJ, Lu JP, Ma ZQ. Metalloform-selective inhibitors of escherichia coli methionine aminopeptidase and X-ray structure of a Mn(II)-form enzyme complexed with an inhibitor. J Am Chem Soc 2004; 126:13940-1. [PMID: 15506752 DOI: 10.1021/ja045864p] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Methionine aminopeptidase (MetAP) enzymes require a divalent metal ion such as Mn(II), Fe(II), Co(II), Ni(II), or Zn(II) for its removal of the N-terminal methionine from newly synthesized proteins, but it is not certain which of these ions is most important in vivo. Metalloform-selective MetAP inhibitors could be valuable for defining which metals are physiologically relevant for MetAP activation and could serve as leads for development of new therapeutic agents. We have screened a library of 43 736 small drug-like molecules against Escherichia coli MetAP and identified two groups of potent and highly metalloform-selective inhibitors of the Co(II)-form, and of the Mn(II)-form, of this enzyme. Compound 1 is 790-fold more selective for the Co(II)-form, while compound 4 is over 640-fold more potent toward the Mn(II)-form. The X-ray structure of a di-Mn(II) form of E. coli MetAP complexed with the Mn(II)-form-selective compound 4 was obtained, and it shows that the inhibitor interacts with both Mn(II) ions through the two oxygen atoms of its free carboxylate group. The preferential coordination of the hard (oxygen) donors to Mn(II) may contribute to its superb selectivity toward the Mn(II)-form.
Collapse
Affiliation(s)
- Qi-Zhuang Ye
- High Throughput Screening Laboratory and Protein Structure Laboratory, University of Kansas, Lawrence, Kansas 66045, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Chan PF, Holmes DJ, Payne DJ. Finding the gems using genomic discovery: antibacterial drug discovery strategies – the successes and the challenges. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/j.ddstr.2004.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|