1101
|
Halouska S, Fenton RJ, Barletta RG, Powers R. Predicting the in vivo mechanism of action for drug leads using NMR metabolomics. ACS Chem Biol 2012; 7:166-71. [PMID: 22007661 DOI: 10.1021/cb200348m] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
New strategies are needed to circumvent increasing outbreaks of resistant strains of pathogens and to expand the dwindling supply of effective antimicrobials. A common impediment to drug development is the lack of an easy approach to determine the in vivo mechanism of action and efficacy of novel drug leads. Toward this end, we describe an unbiased approach to predict in vivo mechanisms of action from NMR metabolomics data. Mycobacterium smegmatis, a non-pathogenic model organism for Mycobacterium tuberculosis, was treated with 12 known drugs and 3 chemical leads identified from a cell-based assay. NMR analysis of drug-induced changes to the M. smegmatis metabolome resulted in distinct clustering patterns correlating with in vivo drug activity. The clustering of novel chemical leads relative to known drugs provides a mean to identify a protein target or predict in vivo activity.
Collapse
Affiliation(s)
- Steven Halouska
- Department
of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304,
United States
| | - Robert J. Fenton
- School of
Veterinary Medicine
and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0905, United States
| | - Raúl G. Barletta
- School of
Veterinary Medicine
and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0905, United States
| | - Robert Powers
- Department
of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304,
United States
| |
Collapse
|
1102
|
Chhabra S, Dolezal O, Collins BM, Newman J, Simpson JS, Macreadie IG, Fernley R, Peat TS, Swarbrick JD. Structure of S. aureus HPPK and the discovery of a new substrate site inhibitor. PLoS One 2012; 7:e29444. [PMID: 22276115 PMCID: PMC3261883 DOI: 10.1371/journal.pone.0029444] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 11/28/2011] [Indexed: 12/17/2022] Open
Abstract
The first structural and biophysical data on the folate biosynthesis pathway enzyme and drug target, 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (SaHPPK), from the pathogen Staphylococcus aureus is presented. HPPK is the second essential enzyme in the pathway catalysing the pyrophosphoryl transfer from cofactor (ATP) to the substrate (6-hydroxymethyl-7,8-dihydropterin, HMDP). In-silico screening identified 8-mercaptoguanine which was shown to bind with an equilibrium dissociation constant, Kd, of ∼13 µM as measured by isothermal titration calorimetry (ITC) and surface plasmon resonance (SPR). An IC50 of ∼41 µM was determined by means of a luminescent kinase assay. In contrast to the biological substrate, the inhibitor has no requirement for magnesium or the ATP cofactor for competitive binding to the substrate site. The 1.65 Å resolution crystal structure of the inhibited complex showed that it binds in the pterin site and shares many of the key intermolecular interactions of the substrate. Chemical shift and 15N heteronuclear NMR measurements reveal that the fast motion of the pterin-binding loop (L2) is partially dampened in the SaHPPK/HMDP/α,β-methylene adenosine 5′-triphosphate (AMPCPP) ternary complex, but the ATP loop (L3) remains mobile on the µs-ms timescale. In contrast, for the SaHPPK/8-mercaptoguanine/AMPCPP ternary complex, the loop L2 becomes rigid on the fast timescale and the L3 loop also becomes more ordered – an observation that correlates with the large entropic penalty associated with inhibitor binding as revealed by ITC. NMR data, including 15N-1H residual dipolar coupling measurements, indicate that the sulfur atom in the inhibitor is important for stabilizing and restricting important motions of the L2 and L3 catalytic loops in the inhibited ternary complex. This work describes a comprehensive analysis of a new HPPK inhibitor, and may provide a foundation for the development of novel antimicrobials targeting the folate biosynthetic pathway.
Collapse
Affiliation(s)
- Sandeep Chhabra
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- CSIRO Division of Materials, Science and Engineering, Parkville, Australia
| | - Olan Dolezal
- CSIRO Division of Materials, Science and Engineering, Parkville, Australia
| | - Brett M. Collins
- Institute for Molecular Bioscience, The University of Queensland, Australia
| | - Janet Newman
- CSIRO Division of Materials, Science and Engineering, Parkville, Australia
| | - Jamie S. Simpson
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Ian G. Macreadie
- School of Applied Sciences, RMIT University, Bundoora, Australia
| | - Ross Fernley
- CSIRO Division of Materials, Science and Engineering, Parkville, Australia
| | - Thomas S. Peat
- CSIRO Division of Materials, Science and Engineering, Parkville, Australia
| | - James D. Swarbrick
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- * E-mail:
| |
Collapse
|
1103
|
Brown MF, Reilly U, Abramite JA, Arcari JT, Oliver R, Barham RA, Che Y, Chen JM, Collantes EM, Chung SW, Desbonnet C, Doty J, Doroski M, Engtrakul JJ, Harris TM, Huband M, Knafels JD, Leach KL, Liu S, Marfat A, Marra A, McElroy E, Melnick M, Menard CA, Montgomery JI, Mullins L, Noe MC, O'Donnell J, Penzien J, Plummer MS, Price LM, Shanmugasundaram V, Thoma C, Uccello DP, Warmus JS, Wishka DG. Potent inhibitors of LpxC for the treatment of Gram-negative infections. J Med Chem 2012; 55:914-23. [PMID: 22175825 DOI: 10.1021/jm2014748] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this paper, we present the synthesis and SAR as well as selectivity, pharmacokinetic, and infection model data for representative analogues of a novel series of potent antibacterial LpxC inhibitors represented by hydroxamic acid.
Collapse
Affiliation(s)
- Matthew F Brown
- Worldwide Medicinal Chemistry, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1104
|
Abstract
Mycobacterium tuberculosis is a difficult pathogen to combat and the first-line drugs currently in use are 40-60 years old. The need for new TB drugs is urgent, but the time to identify, develop and ultimately advance new drug regimens onto the market has been excruciatingly slow. On the other hand, the drugs currently in clinical development, and the recent gains in knowledge of the pathogen and the disease itself give us hope for finding new drug targets and new drug leads. In this article we highlight the unique biology of the pathogen and several possible ways to identify new TB chemical leads. The Global Alliance for TB Drug Development (TB Alliance) is a not-for-profit organization whose mission is to accelerate the discovery and development of new TB drugs. The organization carries out research and development in collaboration with many academic laboratories and pharmaceutical companies around the world. In this perspective we will focus on the early discovery phases of drug development and try to provide snapshots of both the current status and future prospects.
Collapse
|
1105
|
Jaiganesh R, Sampath Kumar NS. Marine bacterial sources of bioactive compounds. ADVANCES IN FOOD AND NUTRITION RESEARCH 2012; 65:389-408. [PMID: 22361201 DOI: 10.1016/b978-0-12-416003-3.00025-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Thousands of novel compounds have been isolated from various marine bacteria and tested for pharmacological properties, many of which are commercially available. Many more are being tested as potential bioactive compound at the preclinical and clinical stages. The growing interest in marine-derived antiviral compounds, along with the development of new technology in marine cultures and extraction, will significantly expedite the current exploration of the marine environment for compounds with significant pharmacological applications, which will continue to be a promising strategy and new trend for modern medicine. Marine actinomycetes and cyanobacteria are a prolific but underexploited source for the discovery of novel secondary metabolites.
Collapse
Affiliation(s)
- R Jaiganesh
- Department of Biotechnology, School of Bioengineering, SRM University, Tamil Nadu, India.
| | | |
Collapse
|
1106
|
Vudumula U, Adhikari MD, Ojha B, Goswami S, Das G, Ramesh A. Tuning the bactericidal repertoire and potency of quinoline-based amphiphiles for enhanced killing of pathogenic bacteria. RSC Adv 2012. [DOI: 10.1039/c2ra20140b] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
1107
|
Abstract
Antibiotics remain one of our most important pharmacological tools for the control of infectious disease. However, unlike most other drugs, the use of antibiotics selects for resistant organisms and erodes their clinical utility. Resistance can emerge within populations of bacteria by mutation and be retained by subsequent selection or by the acquisition of resistance elements laterally from other organisms. The source of these resistance genes is only now being understood. The evidence supports a large bacterial resistome-the collection of all resistance genes and their precursors in both pathogenic and nonpathogenic bacteria. These genes have arisen by various means including self-protection in the case of antibiotic producers, transport of small molecules for various reasons including nutrition and detoxification of noxious chemicals, and to accomplish other goals, such as metabolism, and demonstrate serendipitous selectivity for antibiotics. Regardless of their origins, resistance genes can rapidly move through bacterial populations and emerge in pathogenic bacteria. Understanding the processes that contribute to the evolution and selection of resistance is essential to mange current stocks of antibiotics and develop new ones.
Collapse
Affiliation(s)
- Gerard D Wright
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
1108
|
Seyedsayamdost MR, Traxler MF, Clardy J, Kolter R. Old meets new: using interspecies interactions to detect secondary metabolite production in actinomycetes. Methods Enzymol 2012; 517:89-109. [PMID: 23084935 PMCID: PMC4004031 DOI: 10.1016/b978-0-12-404634-4.00005-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Actinomycetes, a group of filamentous, Gram-positive bacteria, have long been a remarkable source of useful therapeutics. Recent genome sequencing and transcriptomic studies have shown that these bacteria, responsible for half of the clinically used antibiotics, also harbor a large reservoir of gene clusters, which have the potential to produce novel secreted small molecules. Yet, many of these clusters are not expressed under common culture conditions. One reason why these clusters have not been linked to a secreted small molecule lies in the way that actinomycetes have typically been studied: as pure cultures in nutrient-rich media that do not mimic the complex environments in which these bacteria evolved. New methods based on multispecies culture conditions provide an alternative approach to investigating the products of these gene clusters. We have recently implemented binary interspecies interaction assays to mine for new secondary metabolites and to study the underlying biology of interactinomycete interactions. Here, we describe the detailed biological and chemical methods comprising these studies.
Collapse
Affiliation(s)
- Mohammad R Seyedsayamdost
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew F Traxler
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Roberto Kolter
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
1109
|
Amori L, Katkevica S, Bruno A, Campanini B, Felici P, Mozzarelli A, Costantino G. Design and synthesis of trans-2-substituted-cyclopropane-1-carboxylic acids as the first non-natural small molecule inhibitors of O-acetylserine sulfhydrylase. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20100c] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
1110
|
Pieroni M, Sabatini S, Massari S, Kaatz GW, Cecchetti V, Tabarrini O. Searching for innovative quinolone-like scaffolds: synthesis and biological evaluation of 2,1-benzothiazine 2,2-dioxide derivatives. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20101a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
1111
|
O' Connor CJ, Beckmann HSG, Spring DR. Diversity-oriented synthesis: producing chemical tools for dissecting biology. Chem Soc Rev 2012; 41:4444-56. [DOI: 10.1039/c2cs35023h] [Citation(s) in RCA: 348] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
1112
|
Abstract
Phage therapy is the clinical or veterinary application of bacterial viruses (bacteriophages) as antibacterial "drugs." More generally, phages can be used as biocontrol agents against plant as well as foodborne pathogens. In this chapter, we consider the therapeutic use of phage cocktails, which is the combining of two or more phage types to produce more pharmacologically diverse formulations. The primary motivation for the use of cocktails is their broader spectra of activity in comparison to individual phage isolates: they can impact either more bacterial types or achieve effectiveness under a greater diversity of conditions. The combining of phages can also facilitate better targeting of multiple strains making up individual bacterial species or covering multiple species that might be responsible for similar disease states, in general providing, relative to individual phage isolates, a greater potential for presumptive or empirical treatment. Contrasting the use of phage banks, or even phage isolation against specific etiologies that have been obtained directly from patients under treatment, here we consider the utility as well as potential shortcomings associated with the use of phage cocktails as therapeutic antibacterial agents.
Collapse
Affiliation(s)
- Benjamin K Chan
- Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | | |
Collapse
|
1113
|
Antistaphylococcal activity of TD-1792, a multivalent glycopeptide-cephalosporin antibiotic. Antimicrob Agents Chemother 2011; 56:1584-7. [PMID: 22203585 DOI: 10.1128/aac.05532-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
TD-1792 is a new multivalent glycopeptide-cephalosporin antibiotic with potent activity against Gram-positive bacteria. The in vitro activity of TD-1792 was tested against 527 Staphylococcus aureus isolates, including multidrug-resistant isolates. TD-1792 was highly active against methicillin-susceptible S. aureus (MIC(90), 0.015 μg/ml), methicillin-resistant S. aureus, and heterogeneous vancomycin-intermediate S. aureus (MIC(90), 0.03 μg/ml). Time-kill studies demonstrated the potent bactericidal activity of TD-1792 at concentrations of ≤ 0.12 μg/ml. A postantibiotic effect of >2 h was observed after exposure to TD-1792.
Collapse
|
1114
|
Polyak SW, Abell AD, Wilce MCJ, Zhang L, Booker GW. Structure, function and selective inhibition of bacterial acetyl-coa carboxylase. Appl Microbiol Biotechnol 2011; 93:983-92. [PMID: 22183085 DOI: 10.1007/s00253-011-3796-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 11/21/2011] [Accepted: 11/24/2011] [Indexed: 11/24/2022]
Abstract
Acetyl-CoA carboxylase (ACC) catalyses the first committed step in fatty acid biosynthesis: a metabolic pathway required for several important biological processes including the synthesis and maintenance of cellular membranes. ACC employs a covalently attached biotin moiety to bind a carboxyl anion and then transfer it to acetyl-CoA, yielding malonyl-CoA. These activities occur at two different subsites: the biotin carboxylase (BC) and carboxyltransferase (CT). Structural biology, together with small molecule inhibitor studies, has provided new insights into the molecular mechanisms that govern ACC catalysis, specifically the BC and CT subunits. Here, we review these recent findings and highlight key differences between the bacterial and eukaryotic isozymes with a view to establish those features that provide an opportunity for selective inhibition. Especially important are examples of highly selective small molecule inhibitors capable of differentiating between ACCs from different phyla. The implications for early stage antibiotic discovery projects, stemming from these studies, are discussed.
Collapse
Affiliation(s)
- S W Polyak
- School of Molecular and Biomedical Science, University of Adelaide, North Tce, Adelaide, South Australia 5005, Australia.
| | | | | | | | | |
Collapse
|
1115
|
Cui Y, Zhao Y, Tian Y, Zhang W, Lü X, Jiang X. The molecular mechanism of action of bactericidal gold nanoparticles on Escherichia coli. Biomaterials 2011; 33:2327-33. [PMID: 22182745 DOI: 10.1016/j.biomaterials.2011.11.057] [Citation(s) in RCA: 464] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 11/22/2011] [Indexed: 11/17/2022]
Abstract
This work examines the molecular mechanism of action of a class of bactericidal gold nanoparticles (NPs) which show potent antibacterial activities against multidrug-resistant Gram-negative bacteria by transcriptomic and proteomic approaches. Gold NPs exert their antibacterial activities mainly by two ways: one is to collapse membrane potential, inhibiting ATPase activities to decrease the ATP level; the other is to inhibit the subunit of ribosome from binding tRNA. Gold NPs enhance chemotaxis in the early-phase reaction. The action of gold NPs did not include reactive oxygen species (ROS)-related mechanism, the cause for cellular death induced by most bactericidal antibiotics and nanomaterials. Our investigation would allow the development of antibacterial agents that target the energy-metabolism and transcription of bacteria without triggering the ROS reaction, which may be at the same time harmful for the host when killing bacteria.
Collapse
Affiliation(s)
- Yan Cui
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Si Pailou, Nanjing 210096, China
| | | | | | | | | | | |
Collapse
|
1116
|
Birkenstock T, Liebeke M, Winstel V, Krismer B, Gekeler C, Niemiec MJ, Bisswanger H, Lalk M, Peschel A. Exometabolome analysis identifies pyruvate dehydrogenase as a target for the antibiotic triphenylbismuthdichloride in multiresistant bacterial pathogens. J Biol Chem 2011; 287:2887-95. [PMID: 22144679 DOI: 10.1074/jbc.m111.288894] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The desperate need for new therapeutics against notoriously antibiotic-resistant bacteria has led to a quest for novel antibacterial target structures and compounds. Moreover, defining targets and modes of action of new antimicrobial compounds remains a major challenge with standard technologies. Here we characterize the antibacterial properties of triphenylbismuthdichloride (TPBC), which has recently been successfully used against device-associated infections. We demonstrate that TPBC has potent antimicrobial activity against many bacterial pathogens. Using an exometabolome profiling approach, a unique TPBC-mediated change in the metabolites of Staphylococcus aureus was identified, indicating that TPBC blocks bacterial pyruvate catabolism. Enzymatic studies showed that TPBC is a highly efficient, uncompetitive inhibitor of the bacterial pyruvate dehydrogenase complex. Our study demonstrates that metabolomics approaches can offer new avenues for studying the modes of action of antimicrobial compounds, and it indicates that inhibition of the bacterial pyruvate dehydrogenase complex may represent a promising strategy for combating multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Timo Birkenstock
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology Division, University of Tübingen, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
1117
|
Kirst HA. Recent derivatives from smaller classes of fermentation-derived antibacterials. Expert Opin Ther Pat 2011; 22:15-35. [DOI: 10.1517/13543776.2012.642370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
1118
|
Laffan AM, McKenzie R, Forti J, Conklin D, Marcinko R, Shrestha R, Bellantoni M, Greenough WB. Lactoferrin for the prevention of post-antibiotic diarrhoea. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2011; 29:547-551. [PMID: 22283027 PMCID: PMC3259716 DOI: 10.3329/jhpn.v29i6.9889] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Antibiotic-associated diarrhoea (AAD) is a common cause of morbidity and mortality. Older individuals in long-term care facilities are particularly vulnerable due to multisystem illnesses and the prevailing conditions for nosocomial infections. Lactoferrin, an antimicrobial protein in human breastmilk, was tested to determine whether it would prevent or reduce AAD, including Clostridium difficile in tube-fed long-term care patients. Thirty patients were enrolled in a randomized double-blind study, testing eight weeks of human recombinant lactoferrin compared to placebo for the prevention of antibiotic-associated diarrhoea in long-term care patients. Fewer patients in the lactoferrin group experienced diarrhoea compared to controls (p = 0.023). Based on the findings, it is concluded that human lactoferrin may reduce post-antibiotic diarrhoea.
Collapse
Affiliation(s)
- Alison M. Laffan
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Robin McKenzie
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Jennifer Forti
- Division of Geriatric Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Dawn Conklin
- Division of Geriatric Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Richard Marcinko
- Division of Geriatric Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Ruchee Shrestha
- Division of Geriatric Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Michele Bellantoni
- Division of Geriatric Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - William B. Greenough
- Division of Geriatric Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| |
Collapse
|
1119
|
A Small Molecule Discrimination Map of the Antibiotic Resistance Kinome. ACTA ACUST UNITED AC 2011; 18:1591-601. [DOI: 10.1016/j.chembiol.2011.10.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/21/2011] [Accepted: 10/24/2011] [Indexed: 12/21/2022]
|
1120
|
Kim HU, Sohn SB, Lee SY. Metabolic network modeling and simulation for drug targeting and discovery. Biotechnol J 2011; 7:330-42. [DOI: 10.1002/biot.201100159] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 09/09/2011] [Accepted: 10/08/2011] [Indexed: 11/08/2022]
|
1121
|
Wang F, Qin L, Pace CJ, Wong P, Malonis R, Gao J. Solubilized Gramicidin A as Potential Systemic Antibiotics. Chembiochem 2011; 13:51-5. [DOI: 10.1002/cbic.201100671] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Indexed: 11/07/2022]
|
1122
|
Lu X, Zhou R, Sharma I, Li X, Kumar G, Swaminathan S, Tonge PJ, Tan DS. Stable analogues of OSB-AMP: potent inhibitors of MenE, the o-succinylbenzoate-CoA synthetase from bacterial menaquinone biosynthesis. Chembiochem 2011; 13:129-36. [PMID: 22109989 DOI: 10.1002/cbic.201100585] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Indexed: 12/15/2022]
Abstract
MenE, the o-succinylbenzoate (OSB)-CoA synthetase from bacterial menaquinone biosynthesis, is a promising new antibacterial target. Sulfonyladenosine analogues of the cognate reaction intermediate, OSB-AMP, have been developed as inhibitors of the MenE enzymes from Mycobacterium tuberculosis (mtMenE), Staphylococcus aureus (saMenE) and Escherichia coli (ecMenE). Both a free carboxylate and a ketone moiety on the OSB side chain are required for potent inhibitory activity. OSB-AMS (4) is a competitive inhibitor of mtMenE with respect to ATP (K(i) =5.4±0.1 nM) and a noncompetitive inhibitor with respect to OSB (K(i) =11.2±0.9 nM). These data are consistent with a Bi Uni Uni Bi Ping-Pong kinetic mechanism for these enzymes. In addition, OSB-AMS inhibits saMenE with K(i)(app) =22±8 nM and ecMenE with K(i)(OSB) =128±5 nM. Putative active-site residues, Arg222, which may interact with the OSB aromatic carboxylate, and Ser302, which may bind the OSB ketone oxygen, have been identified through computational docking of OSB-AMP with the unliganded crystal structure of saMenE. A pH-dependent interconversion of the free keto acid and lactol forms of the inhibitors is also described, along with implications for inhibitor design.
Collapse
Affiliation(s)
- Xuequan Lu
- Molecular Pharmacology and Chemistry Program and Tri-Institutional Research Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
1123
|
Drake EJ, Gulick AM. Structural characterization and high-throughput screening of inhibitors of PvdQ, an NTN hydrolase involved in pyoverdine synthesis. ACS Chem Biol 2011; 6:1277-86. [PMID: 21892836 DOI: 10.1021/cb2002973] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The human pathogen Pseudomonas aeruginosa produces a variety of virulence factors including pyoverdine, a nonribosomally produced peptide siderophore. The maturation pathway of the pyoverdine peptide is complex and provides a unique target for inhibition. Within the pyoverdine biosynthetic cluster is a periplasmic hydrolase, PvdQ, that is required for pyoverdine production. However, the precise role of PvdQ in the maturation pathway has not been biochemically characterized. We demonstrate herein that the initial module of the nonribosomal peptide synthetase PvdL adds a myristate moiety to the pyoverdine precursor. We extracted this acylated precursor, called PVDIq, from a pvdQ mutant strain and show that the PvdQ enzyme removes the fatty acid catalyzing one of the final steps in pyoverdine maturation. Incubation of PVDIq with crystals of PvdQ allowed us to capture the acylated enzyme and confirm through structural studies the chemical composition of the incorporated acyl chain. Finally, because inhibition of siderophore synthesis has been identified as a potential antibiotic strategy, we developed a high-throughput screening assay and tested a small chemical library for compounds that inhibit PvdQ activity. Two compounds that block PvdQ have been identified, and their binding within the fatty acid binding pocket was structurally characterized.
Collapse
Affiliation(s)
- Eric J. Drake
- Hauptman-Woodward Medical Research Institute and Department of Structural Biology, State University of New York at Buffalo, 700 Ellicott Street, Buffalo, New York 14203-1102, United States
| | - Andrew M. Gulick
- Hauptman-Woodward Medical Research Institute and Department of Structural Biology, State University of New York at Buffalo, 700 Ellicott Street, Buffalo, New York 14203-1102, United States
| |
Collapse
|
1124
|
Burian J, Ramón-García S, Sweet G, Gómez-Velasco A, Av-Gay Y, Thompson CJ. The mycobacterial transcriptional regulator whiB7 gene links redox homeostasis and intrinsic antibiotic resistance. J Biol Chem 2011; 287:299-310. [PMID: 22069311 DOI: 10.1074/jbc.m111.302588] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Intrinsic drug resistance in Mycobacterium tuberculosis limits therapeutic options for treating tuberculosis. The mycobacterial transcriptional regulator whiB7 contributes to intrinsic resistance by activating its own expression and many drug resistance genes in response to antibiotics. To investigate whiB7 activation, we constructed a GFP reporter to monitor its expression, and we used it to investigate the whiB7 promoter and to screen our custom library of almost 600 bioactive compounds, including the majority of clinical antibiotics. Results showed whiB7 was transcribed from a promoter that was conserved across mycobacteria and other actinomycetes, including an AT-rich sequence that was likely targeted by WhiB7. Expression was induced by compounds having diverse structures and targets, independent of the ability of whiB7 to mediate resistance, and was dependent on media composition. Pretreatment with whiB7 activators resulted in clinically relevant increases in intrinsic drug resistance. Antibiotic-induced transcription was synergistically increased by the reductant dithiothreitol, an effect mirrored by a whiB7-dependent shift to a highly reduced cytoplasm reflected by the ratio of reduced/oxidized mycothiol. These data provided evidence that intrinsic resistance resulting from whiB7 activation is linked to fundamental changes in cell metabolism.
Collapse
Affiliation(s)
- Ján Burian
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Centre for Tuberculosis Research, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Santiago Ramón-García
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Centre for Tuberculosis Research, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Gaye Sweet
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Centre for Tuberculosis Research, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Anaximandro Gómez-Velasco
- Centre for Tuberculosis Research, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Yossef Av-Gay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Centre for Tuberculosis Research, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Charles J Thompson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Centre for Tuberculosis Research, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
1125
|
Kadam RU, Bergmann M, Hurley M, Garg D, Cacciarini M, Swiderska MA, Nativi C, Sattler M, Smyth AR, Williams P, Cámara M, Stocker A, Darbre T, Reymond JL. A glycopeptide dendrimer inhibitor of the galactose-specific lectin LecA and of Pseudomonas aeruginosa biofilms. Angew Chem Int Ed Engl 2011; 50:10631-5. [PMID: 21919164 PMCID: PMC3262149 DOI: 10.1002/anie.201104342] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 08/17/2011] [Indexed: 11/26/2022]
Affiliation(s)
- Rameshwar U Kadam
- Department of Chemistry and Biochemistry, University of BerneFreiestrasse 3, 3012 Berne (Switzerland)
| | - Myriam Bergmann
- Department of Chemistry and Biochemistry, University of BerneFreiestrasse 3, 3012 Berne (Switzerland)
| | - Matthew Hurley
- School of Molecular Medical Sciences, University of NottinghamNottingham NG7 2UH (UK)
- School of Clinical Sciences, University of NottinghamNottingham NG7 2UH (UK)
| | - Divita Garg
- Institute of Structural Biology, Helmholtz Zentrum München and Center for Integrated Protein Science Munich, Departement Chemie, Technische Universität MünchenLichtenbergstrasse 4, 85747 Garching (Germany)
| | - Martina Cacciarini
- Dipartimento di Chimica, Polo Scientifico e Tecnologico, Universita' degli Studi di FirenzeVia della Lastruccia 3, 13, 50019 Sesto Fiorentino—Firenze (Italy)
| | - Magdalena A Swiderska
- Department of Chemistry and Biochemistry, University of BerneFreiestrasse 3, 3012 Berne (Switzerland)
| | - Cristina Nativi
- Dipartimento di Chimica, Polo Scientifico e Tecnologico, Universita' degli Studi di FirenzeVia della Lastruccia 3, 13, 50019 Sesto Fiorentino—Firenze (Italy)
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München and Center for Integrated Protein Science Munich, Departement Chemie, Technische Universität MünchenLichtenbergstrasse 4, 85747 Garching (Germany)
| | - Alan R Smyth
- School of Clinical Sciences, University of NottinghamNottingham NG7 2UH (UK)
| | - Paul Williams
- School of Molecular Medical Sciences, University of NottinghamNottingham NG7 2UH (UK)
| | - Miguel Cámara
- School of Molecular Medical Sciences, University of NottinghamNottingham NG7 2UH (UK)
| | - Achim Stocker
- Department of Chemistry and Biochemistry, University of BerneFreiestrasse 3, 3012 Berne (Switzerland)
| | - Tamis Darbre
- Department of Chemistry and Biochemistry, University of BerneFreiestrasse 3, 3012 Berne (Switzerland)
| | - Jean-Louis Reymond
- Department of Chemistry and Biochemistry, University of BerneFreiestrasse 3, 3012 Berne (Switzerland)
| |
Collapse
|
1126
|
Bendigoles D–F, bioactive sterols from the marine sponge-derived Actinomadura sp. SBMs009. Bioorg Med Chem 2011; 19:6570-5. [DOI: 10.1016/j.bmc.2011.05.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 04/21/2011] [Accepted: 05/23/2011] [Indexed: 11/19/2022]
|
1127
|
Liu WT, Kersten RD, Yang YL, Moore BS, Dorrestein PC. Imaging mass spectrometry and genome mining via short sequence tagging identified the anti-infective agent arylomycin in Streptomyces roseosporus. J Am Chem Soc 2011; 133:18010-3. [PMID: 21999343 DOI: 10.1021/ja2040877] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Here, we described the discovery of anti-infective agent arylomycin and its biosynthetic gene cluster in an industrial daptomycin producing strain Streptomyces roseosporus. This was accomplished via the use of MALDI imaging mass spectrometry (IMS) along with peptidogenomic approach in which we have expanded to short sequence tagging (SST) described herein. Using IMS, we observed that prior to the production of daptomycin, a cluster of ions (1-3) was produced by S. roseosporus and correlated well with the decreased staphylococcal cell growth. With a further adopted SST peptidogenomics approach, which relies on the generation of sequence tags from tandem mass spectrometric data and query against genomes to identify the biosynthetic genes, we were able to identify these three molecules (1-3) to arylomycins, a class of broad-spectrum antibiotics that target type I signal peptidase. The gene cluster was then identified. This highlights the strength of IMS and MS guided genome mining approaches in effectively bridging the gap between phenotypes, chemotypes, and genotypes.
Collapse
Affiliation(s)
- Wei-Ting Liu
- Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, Unites States
| | | | | | | | | |
Collapse
|
1128
|
Protein engineering towards natural product synthesis and diversification. J Ind Microbiol Biotechnol 2011; 39:227-41. [PMID: 22006344 DOI: 10.1007/s10295-011-1044-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/29/2011] [Indexed: 10/16/2022]
Abstract
A dazzling array of enzymes is used by nature in making structurally complex natural products. These enzymes constitute a molecular toolbox that may be used in the construction and fine-tuning of pharmaceutically active molecules. Aided by technological advancements in protein engineering, it is now possible to tailor the activities and specificities of these enzymes as biocatalysts in the production of both natural products and their unnatural derivatives. These efforts are crucial in drug discovery and development, where there is a continuous quest for more potent agents. Both rational and random evolution techniques have been utilized in engineering these enzymes. This review will highlight some examples from several large families of natural products.
Collapse
|
1129
|
Wencewicz TA, Yang B, Rudloff JR, Oliver AG, Miller MJ. N-O chemistry for antibiotics: discovery of N-alkyl-N-(pyridin-2-yl)hydroxylamine scaffolds as selective antibacterial agents using nitroso Diels-Alder and ene chemistry. J Med Chem 2011; 54:6843-58. [PMID: 21859126 PMCID: PMC3188665 DOI: 10.1021/jm200794r] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The discovery, syntheses, and structure-activity relationships (SAR) of a new family of heterocyclic antibacterial compounds based on N-alkyl-N-(pyridin-2-yl)hydroxylamine scaffolds are described. A structurally diverse library of ∼100 heterocyclic molecules generated from Lewis acid-mediated nucleophilic ring-opening reactions with nitroso Diels-Alder cycloadducts and nitroso ene reactions with substituted alkenes was evaluated in whole cell antibacterial assays. Compounds containing the N-alkyl-N-(pyridin-2-yl)hydroxylamine structure demonstrated selective and potent antibacterial activity against the Gram-positive bacterium Micrococcus luteus ATCC 10240 (MIC(90) = 2.0 μM or 0.41 μg/mL) and moderate activity against other Gram-positive strains including antibiotic resistant strains of Staphylococcus aureus (MRSA) and Enterococcus faecalis (VRE). A new synthetic route to the active core was developed using palladium-catalyzed Buchwald-Hartwig amination reactions of N-alkyl-O-(4-methoxybenzyl)hydroxylamines with 2-halo-pyridines that facilitated SAR studies and revealed the simplest active structural fragment. This work shows the value of using a combination of diversity-oriented synthesis (DOS) and parallel synthesis for identifying new antibacterial scaffolds.
Collapse
Affiliation(s)
- Timothy A. Wencewicz
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Baiyuan Yang
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | - James R. Rudloff
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Allen G. Oliver
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Marvin J. Miller
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
1130
|
Discovery of parallel pathways of kanamycin biosynthesis allows antibiotic manipulation. Nat Chem Biol 2011; 7:843-52. [PMID: 21983602 DOI: 10.1038/nchembio.671] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 07/28/2011] [Indexed: 11/08/2022]
Abstract
Kanamycin is one of the most widely used antibiotics, yet its biosynthetic pathway remains unclear. Current proposals suggest that the kanamycin biosynthetic products are linearly related via single enzymatic transformations. To explore this system, we have reconstructed the entire biosynthetic pathway through the heterologous expression of combinations of putative biosynthetic genes from Streptomyces kanamyceticus in the non-aminoglycoside-producing Streptomyces venezuelae. Unexpectedly, we discovered that the biosynthetic pathway contains an early branch point, governed by the substrate promiscuity of a glycosyltransferase, that leads to the formation of two parallel pathways in which early intermediates are further modified. Glycosyltransferase exchange can alter flux through these two parallel pathways, and the addition of other biosynthetic enzymes can be used to synthesize known and new highly active antibiotics. These results complete our understanding of kanamycin biosynthesis and demonstrate the potential of pathway engineering for direct in vivo production of clinically useful antibiotics and more robust aminoglycosides.
Collapse
|
1131
|
Thirunavukkarasu N, Suryanarayanan TS, Girivasan KP, Venkatachalam A, Geetha V, Ravishankar JP, Doble M. Fungal symbionts of marine sponges from Rameswaram, southern India: species composition and bioactive metabolites. FUNGAL DIVERS 2011. [DOI: 10.1007/s13225-011-0137-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1132
|
Juhas M, Eberl L, Glass JI. Essence of life: essential genes of minimal genomes. Trends Cell Biol 2011; 21:562-8. [DOI: 10.1016/j.tcb.2011.07.005] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/25/2011] [Accepted: 07/27/2011] [Indexed: 11/29/2022]
|
1133
|
Putty S, Rai A, Jamindar D, Pagano P, Quinn CL, Mima T, Schweizer HP, Gutheil WG. Characterization of d-boroAla as a novel broad-spectrum antibacterial agent targeting d-Ala-d-Ala ligase. Chem Biol Drug Des 2011; 78:757-63. [PMID: 21827632 DOI: 10.1111/j.1747-0285.2011.01210.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
d-boroAla was previously characterized as an inhibitor of bacterial alanine racemase and d-Ala-d-Ala ligase enzymes (Biochemistry, 28, 1989, 3541). In this study, d-boroAla was identified and characterized as an antibacterial agent. d-boroAla has activity against both Gram-positive and Gram-negative organisms, with minimal inhibitory concentrations down to 8 μg / mL. A structure-function study on the alkyl side chain (NH(2) -CHR-B(OR')(2) ) revealed that d-boroAla is the most effective agent in a series including boroGly, d-boroHomoAla, and d-boroVal. l-boroAla was much less active, and N-acetylation completely abolished activity. An LC-MS / MS assay was used to demonstrate that d-boroAla exerts its antibacterial activity by inhibition of d-Ala-d-Ala ligase. d-boroAla is bactericidal at 1× minimal inhibitory concentration against Staphylococcus aureus and Bacillus subtilis, which each encode one copy of d-Ala-d-Ala ligase, and at 4× minimal inhibitory concentration against Escherichia coli and Salmonella enterica serovar Typhimurium, which each encode two copies of d-Ala-d-Ala ligase. d-boroAla demonstrated a frequency of resistance of 8 × 10(-8) at 4× minimal inhibitory concentration in S. aureus. These results demonstrate that d-boroAla has promising antibacterial activity and could serve as the lead agent in a new class of d-Ala-d-Ala ligase targeted antibacterial agents. This study also demonstrates d-boroAla as a possible probe for d-Ala-d-Ala ligase function.
Collapse
Affiliation(s)
- Sandeep Putty
- Division of Pharmaceutical Sciences, University of Missouri, Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | | | | | | | | | | | | | | |
Collapse
|
1134
|
Bhunia A, Bhattacharjya S. Mapping residue-specific contacts of polymyxin B with lipopolysaccharide by saturation transfer difference NMR: insights into outer-membrane disruption and endotoxin neutralization. Biopolymers 2011; 96:273-87. [PMID: 20683937 DOI: 10.1002/bip.21530] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
High-resolution interactions studies of molecules with lipopolysaccharide (LPS) or endotoxin are important for health, diseases and environment. LPS is the major constituent of the outer layer of the outer membrane of the gram-negative bacteria. LPS provides an efficient barrier against permeation of variety of compounds including antibacterial agents and antimicrobial peptides. In the intensive care units, LPS is known for the fatal septic shock syndromes. Because of LPS toxicity, high affinity LPS sensors are sought-after for the assessment of the quality of water and pharmaceutical products. Therefore, elucidation of binding epitopes of LPS interacting molecules would be vital for the development of antimicrobial, antiendotoxic molecules. Polymyxin B (PMB), an antibacterial cyclic lipo-peptide, is well known for its LPS sequestering and neutralizing activities. Here, we have used saturation transfer difference (STD) NMR methods for characterizing interactions of PMB with LPS from E. coli 0111:B4 and P. aeruginosa. The dissociation constants of the LPS-PMB complexes were obtained from concentration dependent STD studies. Further a detailed epitope mapping of PMB has been carried out in E. coli 0111:B4 LPS micelles. Experiments including one-dimensional 1H STD, two-dimensional 1H-1H STD-TOCSY and naturalabundance 13C-1H STD-HSQC, are performed to determine the site(s) of interactions of PMB with endotoxin at atomic resolution. Our studies reveal that the hydrophobic sidechains of PMB including a part of the N-terminus lipidic tail demonstrate close contacts with LPS. In contrast, cyclic backbone structure of PMB has the lowest STD effects suggesting a rather loose association with endotoxin.
Collapse
Affiliation(s)
- Anirban Bhunia
- Biomolecular NMR and Drug Discovery Laboratory, School of Biological Sciences, Division of Structural and Computational Biology, Nanyang Technological University, Singapore 637551
| | | |
Collapse
|
1135
|
Contreras-Martel C, Amoroso A, Woon ECY, Zervosen A, Inglis S, Martins A, Verlaine O, Rydzik AM, Job V, Luxen A, Joris B, Schofield CJ, Dessen A. Structure-guided design of cell wall biosynthesis inhibitors that overcome β-lactam resistance in Staphylococcus aureus (MRSA). ACS Chem Biol 2011; 6:943-51. [PMID: 21732689 DOI: 10.1021/cb2001846] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
β-Lactam antibiotics have long been a treatment of choice for bacterial infections since they bind irreversibly to Penicillin-Binding Proteins (PBPs), enzymes that are vital for cell wall biosynthesis. Many pathogens express drug-insensitive PBPs rendering β-lactams ineffective, revealing a need for new types of PBP inhibitors active against resistant strains. We have identified alkyl boronic acids that are active against pathogens including methicillin-resistant S. aureus (MRSA). The crystal structures of PBP1b complexed to 11 different alkyl boronates demonstrate that in vivo efficacy correlates with the mode of inhibitor side chain binding. Staphylococcal membrane analyses reveal that the most potent alkyl boronate targets PBP1, an autolysis system regulator, and PBP2a, a low β-lactam affinity enzyme. This work demonstrates the potential of boronate-based PBP inhibitors for circumventing β-lactam resistance and opens avenues for the development of novel antibiotics that target Gram-positive pathogens.
Collapse
Affiliation(s)
| | - Ana Amoroso
- Centre d'Ingénierie des Protéines, Institut de Chimie, B6a, Université de Liège, Sart Tilman, B4000 Liège, Belgium
| | - Esther C. Y. Woon
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | - Astrid Zervosen
- Centre de Recherches du Cyclotron, B30, Université de Liège, Sart Tilman, B4000 Liège, Belgium
| | - Steven Inglis
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | | | - Olivier Verlaine
- Centre d'Ingénierie des Protéines, Institut de Chimie, B6a, Université de Liège, Sart Tilman, B4000 Liège, Belgium
| | - Anna M. Rydzik
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | | | - André Luxen
- Centre de Recherches du Cyclotron, B30, Université de Liège, Sart Tilman, B4000 Liège, Belgium
| | - Bernard Joris
- Centre d'Ingénierie des Protéines, Institut de Chimie, B6a, Université de Liège, Sart Tilman, B4000 Liège, Belgium
| | | | | |
Collapse
|
1136
|
Kadam RU, Bergmann M, Hurley M, Garg D, Cacciarini M, Swiderska MA, Nativi C, Sattler M, Smyth AR, Williams P, Cámara M, Stocker A, Darbre T, Reymond JL. A Glycopeptide Dendrimer Inhibitor of the Galactose-Specific Lectin LecA and of Pseudomonas aeruginosa Biofilms. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201104342] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1137
|
Affiliation(s)
- Diego Romero
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
| | - Matthew F. Traxler
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
| | | | - Roberto Kolter
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
1138
|
Serrano CM, Looper RE. Synthesis of Cytimidine through a One-Pot Copper-Mediated Amidation Cascade. Org Lett 2011; 13:5000-3. [DOI: 10.1021/ol2018196] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Catherine M. Serrano
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112, United States
| | - Ryan E. Looper
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112, United States
| |
Collapse
|
1139
|
Sundaramurthi JC, Ramanandan P, Brindha S, Subhasree CR, Prasad A, Kumaraswami V, Hanna LE. DDTRP: Database of Drug Targets for Resistant Pathogens. Bioinformation 2011; 7:98-101. [PMID: 21938213 PMCID: PMC3174044 DOI: 10.6026/97320630007098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 07/21/2011] [Indexed: 11/30/2022] Open
Abstract
Emergence of drug resistance is a major threat to public health. Many pathogens have developed resistance to most of the existing antibiotics, and multidrug-resistant and extensively drug resistant strains are extremely difficult to treat. This has resulted in an urgent need for novel drugs. We describe a database called 'Database of Drug Targets for Resistant Pathogens' (DDTRP). The database contains information on drugs with reported resistance, their respective targets, metabolic pathways involving these targets, and a list of potential alternate targets for seven pathogens. The database can be accessed freely at http://bmi.icmr.org.in/DDTRP.
Collapse
Affiliation(s)
| | - Prabhakaran Ramanandan
- ICMR-Biomedical Informatics Centre, Tuberculosis Research Centre (ICMR), Chetpet, Chennai-600031, Tamil Nadu, India
| | - Sridharan Brindha
- ICMR-Biomedical Informatics Centre, Tuberculosis Research Centre (ICMR), Chetpet, Chennai-600031, Tamil Nadu, India
| | | | - Abhimanyu Prasad
- ICMR-Biomedical Informatics Centre, Tuberculosis Research Centre (ICMR), Chetpet, Chennai-600031, Tamil Nadu, India
| | - Vasanthapuram Kumaraswami
- ICMR-Biomedical Informatics Centre, Tuberculosis Research Centre (ICMR), Chetpet, Chennai-600031, Tamil Nadu, India
| | - Luke Elizabeth Hanna
- ICMR-Biomedical Informatics Centre, Tuberculosis Research Centre (ICMR), Chetpet, Chennai-600031, Tamil Nadu, India
| |
Collapse
|
1140
|
Fischbach MA. Combination therapies for combating antimicrobial resistance. Curr Opin Microbiol 2011; 14:519-23. [PMID: 21900036 DOI: 10.1016/j.mib.2011.08.003] [Citation(s) in RCA: 173] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 08/17/2011] [Accepted: 08/17/2011] [Indexed: 11/29/2022]
Abstract
New drug development strategies are needed to combat antimicrobial resistance. The object of this perspective is to highlight one such strategy: treating infections with sets of drugs rather than individual drugs. We will highlight three categories of combination therapy: those that inhibit targets in different pathways; those that inhibit distinct nodes in the same pathway; and those that inhibit the very same target in different ways. We will then consider examples of naturally occurring combination therapies produced by micro-organisms, and conclude by discussing key opportunities and challenges for making more widespread use of drug combinations.
Collapse
Affiliation(s)
- Michael A Fischbach
- Department of Bioengineering and Therapeutic Sciences and the California Institute for Quantitative Biosciences, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
1141
|
Hedstrom L, Liechti G, Goldberg JB, Gollapalli DR. The antibiotic potential of prokaryotic IMP dehydrogenase inhibitors. Curr Med Chem 2011; 18:1909-18. [PMID: 21517780 DOI: 10.2174/092986711795590129] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 04/04/2011] [Indexed: 12/30/2022]
Abstract
Inosine 5'-monophosphate dehydrogenase (IMPDH) catalyzes the first committed step of guanosine 5'-monophosphate (GMP) biosynthesis, and thus regulates the guanine nucleotide pool, which in turn governs proliferation. Human IMPDHs are validated targets for immunosuppressive, antiviral and anticancer drugs, but as yet microbial IMPDHs have not been exploited in antimicrobial chemotherapy. Selective inhibitors of IMPDH from Cryptosporidium parvum have recently been discovered that display anti-parasitic activity in cell culture models of infection. X-ray crystal structure and mutagenesis experiments identified the structural features that determine inhibitor susceptibility. These features are found in IMPDHs from a wide variety of pathogenic bacteria, including select agents and multiply drug resistant strains. A second generation inhibitor displays antibacterial activity against Helicobacter pylori, demonstrating the antibiotic potential of IMPDH inhibitors.
Collapse
Affiliation(s)
- L Hedstrom
- Brandeis University, Departments of Biology, Waltham, MA 02454-9110, USA.
| | | | | | | |
Collapse
|
1142
|
Durrant JD, Cao R, Gorfe AA, Zhu W, Li J, Sankovsky A, Oldfield E, McCammon JA. Non-bisphosphonate inhibitors of isoprenoid biosynthesis identified via computer-aided drug design. Chem Biol Drug Des 2011; 78:323-32. [PMID: 21696546 PMCID: PMC3155669 DOI: 10.1111/j.1747-0285.2011.01164.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/03/2011] [Accepted: 06/17/2011] [Indexed: 11/28/2022]
Abstract
The relaxed complex scheme, a virtual-screening methodology that accounts for protein receptor flexibility, was used to identify a low-micromolar, non-bisphosphonate inhibitor of farnesyl diphosphate synthase. Serendipitously, we also found that several predicted farnesyl diphosphate synthase inhibitors were low-micromolar inhibitors of undecaprenyl diphosphate synthase. These results are of interest because farnesyl diphosphate synthase inhibitors are being pursued as both anti-infective and anticancer agents, and undecaprenyl diphosphate synthase inhibitors are antibacterial drug leads.
Collapse
Affiliation(s)
- Jacob D Durrant
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, Mail Code 0365, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1143
|
Jabes D. The antibiotic R&D pipeline: an update. Curr Opin Microbiol 2011; 14:564-9. [PMID: 21873107 DOI: 10.1016/j.mib.2011.08.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 08/06/2011] [Accepted: 08/08/2011] [Indexed: 11/19/2022]
Abstract
There is an urgent need for new antibacterials to target emerging multidrug-resistant bacteria. The need for such agents is rising while the efforts in antibacterial research have declined dramatically in the past few decades with the result of only four compounds belonging to new chemical classes being approved for clinical use. The main reasons that led to this critical situation are shortly described. A renewed interest in the research of new effective antimicrobials is nonetheless delivering compounds deriving mainly from modification of existing drugs, yet new chemical classes are appearing. Because many of these activities have started relatively recently, we should expect a long period before new antibiotics are added to the medical armamentarium.
Collapse
Affiliation(s)
- Daniela Jabes
- NAICONS Scrl. Via Fantoli 16/15, 20138 Milano, Italy.
| |
Collapse
|
1144
|
Cole ST, Riccardi G. New tuberculosis drugs on the horizon. Curr Opin Microbiol 2011; 14:570-6. [PMID: 21821466 DOI: 10.1016/j.mib.2011.07.022] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 07/11/2011] [Accepted: 07/17/2011] [Indexed: 10/17/2022]
Abstract
Tuberculosis (TB) remains a major global health concern whose control has been exacerbated by HIV and the emergence of multidrug-resistant (MDR-TB) and extensively drug-resistant (XDR-TB) strains of Mycobacterium tuberculosis. The demand for new and faster acting TB drugs is thus greater than ever. In the past decade intensive efforts have been made to discover new leads for TB drug development using both target-based and cell-based approaches. Here, we describe the most promising anti-tubercular drug candidates that are in clinical development and introduce some nitro-aromatic compounds that inhibit a new target, DprE1, an essential enzyme involved in a crucial step in mycobacterial cell wall biosynthesis.
Collapse
Affiliation(s)
- Stewart T Cole
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | |
Collapse
|
1145
|
Hast MA, Nichols CB, Armstrong SM, Kelly SM, Hellinga HW, Alspaugh JA, Beese LS. Structures of Cryptococcus neoformans protein farnesyltransferase reveal strategies for developing inhibitors that target fungal pathogens. J Biol Chem 2011; 286:35149-62. [PMID: 21816822 DOI: 10.1074/jbc.m111.250506] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cryptococcus neoformans is a fungal pathogen that causes life-threatening infections in immunocompromised individuals, including AIDS patients and transplant recipients. Few antifungals can treat C. neoformans infections, and drug resistance is increasing. Protein farnesyltransferase (FTase) catalyzes post-translational lipidation of key signal transduction proteins and is essential in C. neoformans. We present a multidisciplinary study validating C. neoformans FTase (CnFTase) as a drug target, showing that several anticancer FTase inhibitors with disparate scaffolds can inhibit C. neoformans and suggesting structure-based strategies for further optimization of these leads. Structural studies are an essential element for species-specific inhibitor development strategies by revealing similarities and differences between pathogen and host orthologs that can be exploited. We, therefore, present eight crystal structures of CnFTase that define the enzymatic reaction cycle, basis of ligand selection, and structurally divergent regions of the active site. Crystal structures of clinically important anticancer FTase inhibitors in complex with CnFTase reveal opportunities for optimization of selectivity for the fungal enzyme by modifying functional groups that interact with structurally diverse regions. A substrate-induced conformational change in CnFTase is observed as part of the reaction cycle, a feature that is mechanistically distinct from human FTase. Our combined structural and functional studies provide a framework for developing FTase inhibitors to treat invasive fungal infections.
Collapse
Affiliation(s)
- Michael A Hast
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | |
Collapse
|
1146
|
Amini S, Tavazoie S. Antibiotics and the post-genome revolution. Curr Opin Microbiol 2011; 14:513-8. [PMID: 21816663 DOI: 10.1016/j.mib.2011.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 12/28/2022]
Abstract
The emergence of pathogenic bacteria resistant to multiple antimicrobial agents is turning into a major crisis in human and veterinary medicine. This necessitates a serious re-evaluation of our approaches toward antibacterial drug discovery and use. Concurrent advances in genomics including whole-genome sequencing, genotyping, and gene expression profiling have the potential to transform our basic understanding of antimicrobial pathways and lead to the discovery of novel targets and therapeutics.
Collapse
Affiliation(s)
- Sasan Amini
- Department of Molecular Biology & Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | | |
Collapse
|
1147
|
Brugarolas P, Duguid EM, Zhang W, Poor CB, He C. Structural and biochemical characterization of N5-carboxyaminoimidazole ribonucleotide synthetase and N5-carboxyaminoimidazole ribonucleotide mutase from Staphylococcus aureus. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2011; 67:707-15. [PMID: 21795812 PMCID: PMC3144853 DOI: 10.1107/s0907444911023821] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 06/17/2011] [Indexed: 11/10/2022]
Abstract
With the rapid rise of methicillin-resistant Staphylococcus aureus infections, new strategies against S. aureus are urgently needed. De novo purine biosynthesis is a promising yet unexploited target, insofar as abundant evidence has shown that bacteria with compromised purine biosynthesis are attenuated. Fundamental differences exist within the process by which humans and bacteria convert 5-aminoimidazole ribonucleotide (AIR) to 4-carboxy-5-aminoimidazole ribonucleotide (CAIR). In bacteria, this transformation occurs through a two-step conversion catalyzed by PurK and PurE; in humans, it is mediated by a one-step conversion catalyzed by class II PurE. Thus, these bacterial enzymes are potential targets for selective antibiotic development. Here, the first comprehensive structural and biochemical characterization of PurK and PurE from S. aureus is presented. Structural analysis of S. aureus PurK reveals a nonconserved phenylalanine near the AIR-binding site that occupies the putative position of the imidazole ring of AIR. Mutation of this phenylalanine to isoleucine or tryptophan reduced the enzyme efficiency by around tenfold. The K(m) for bicarbonate was determined for the first time for a PurK enzyme and was found to be ∼18.8 mM. The structure of PurE is described in comparison to that of human class II PurE. It is confirmed biochemically that His38 is essential for function. These studies aim to provide foundations for future structure-based drug-discovery efforts against S. aureus purine biosynthesis.
Collapse
Affiliation(s)
- Pedro Brugarolas
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, GCIS E321, Chicago, IL 60637, USA
| | - Erica M. Duguid
- Department of Biochemistry and Molecular Biology, The University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| | - Wen Zhang
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, GCIS E321, Chicago, IL 60637, USA
| | - Catherine B. Poor
- Department of Biochemistry and Molecular Biology, The University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| | - Chuan He
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, GCIS E321, Chicago, IL 60637, USA
| |
Collapse
|
1148
|
Baquero F, Coque TM, de la Cruz F. Ecology and evolution as targets: the need for novel eco-evo drugs and strategies to fight antibiotic resistance. Antimicrob Agents Chemother 2011; 55:3649-60. [PMID: 21576439 PMCID: PMC3147629 DOI: 10.1128/aac.00013-11] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In recent years, the explosive spread of antibiotic resistance determinants among pathogenic, commensal, and environmental bacteria has reached a global dimension. Classical measures trying to contain or slow locally the progress of antibiotic resistance in patients on the basis of better antibiotic prescribing policies have clearly become insufficient at the global level. Urgent measures are needed to directly confront the processes influencing antibiotic resistance pollution in the microbiosphere. Recent interdisciplinary research indicates that new eco-evo drugs and strategies, which take ecology and evolution into account, have a promising role in resistance prevention, decontamination, and the eventual restoration of antibiotic susceptibility. This minireview summarizes what is known and what should be further investigated to find drugs and strategies aiming to counteract the "four P's," penetration, promiscuity, plasticity, and persistence of rapidly spreading bacterial clones, mobile genetic elements, or resistance genes. The term "drug" is used in this eco-evo perspective as a tool to fight resistance that is able to prevent, cure, or decrease potential damage caused by antibiotic resistance, not necessarily only at the individual level (the patient) but also at the ecological and evolutionary levels. This view offers a wealth of research opportunities for science and technology and also represents a large adaptive challenge for regulatory agencies and public health officers. Eco-evo drugs and interventions constitute a new avenue for research that might influence not only antibiotic resistance but the maintenance of a healthy interaction between humans and microbial systems in a rapidly changing biosphere.
Collapse
Affiliation(s)
- Fernando Baquero
- Department of Microbiology, Institute Ramón and Cajal for Health Research (IRYCIS), CIBER Research Network in Epidemiology and Public Health (CIBERESP), Ramón y Cajal University Hospital, Madrid, Spain.
| | | | | |
Collapse
|
1149
|
Anti-infectives: Can cellular screening deliver? Curr Opin Chem Biol 2011; 15:529-33. [DOI: 10.1016/j.cbpa.2011.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/18/2011] [Accepted: 06/07/2011] [Indexed: 12/29/2022]
|
1150
|
Chai SC, Wang WL, Ding DR, Ye QZ. Growth inhibition of Escherichia coli and methicillin-resistant Staphylococcus aureus by targeting cellular methionine aminopeptidase. Eur J Med Chem 2011; 46:3537-40. [PMID: 21575996 PMCID: PMC3114176 DOI: 10.1016/j.ejmech.2011.04.056] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 04/21/2011] [Accepted: 04/25/2011] [Indexed: 11/23/2022]
Abstract
Methionine aminopeptidase (MetAP) catalyzes the N-terminal methionine excision from the majority of newly synthesized proteins, which is an essential cotranslational process required for cell survival. As such, MetAP has become an appealing target for the development of antimicrobial therapeutics with novel mechanisms of action. By screening a library of small organic molecules, we previously discovered a class of compounds that selectively inhibit the Fe(II)-form of MetAP. Herein, we demonstrate that some of these compounds and their newly synthesized derivatives halt the growth of Escherichia coli and Staphylococcus aureus cells with significant potency. The most potent compound inhibited methicillin-resistant S. aureus (MRSA) growth with an IC(50) value of 1 μM and MIC of 0.7 μg/ml. Two cell-based assays were used to verify that MetAP is the intracellular target in E. coli cells. These findings can serve as foundation for the development of novel therapeutics against an ever increasing threat by drug resistant staphylococcal infections.
Collapse
Affiliation(s)
| | - Wen-Long Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - De-Rong Ding
- 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
|