1001
|
Neres J, Engelhart CA, Drake EJ, Wilson DJ, Fu P, Boshoff HI, Barry CE, Gulick AM, Aldrich CC. Non-nucleoside inhibitors of BasE, an adenylating enzyme in the siderophore biosynthetic pathway of the opportunistic pathogen Acinetobacter baumannii. J Med Chem 2013; 56:2385-405. [PMID: 23437866 DOI: 10.1021/jm301709s] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Siderophores are small-molecule iron chelators produced by bacteria and other microorganisms for survival under iron limiting conditions such as found in a mammalian host. Siderophore biosynthesis is essential for the virulence of many important Gram-negative pathogens including Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Escherichia coli. We performed high-throughput screening against BasE, which is involved in siderophore biosynthesis in A. baumannii, and identified 6-phenyl-1-(pyridin-4-ylmethyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid 15. Herein we report the synthesis, biochemical, and microbiological evaluation of a systematic series of analogues of the HTS hit 15. Analogue 67 is the most potent analogue with a KD of 2 nM against BasE. Structural characterization of the inhibitors with BasE reveals that they bind in a unique orientation in the active site, occupying all three substrate binding sites, and thus can be considered as multisubstrate inhibitors. These results provide a foundation for future studies aimed at increasing both enzyme potency and antibacterial activity.
Collapse
Affiliation(s)
- João Neres
- Center for Drug Design, Academic Health Center, University of Minnesota , Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1002
|
Abstract
INTRODUCTION Natural products have long been instrumental for discovering antibiotics, but many pharmaceutical companies abandoned this field and new antibiotics declined. In contrast, microbial resistance to current antibiotics has approached critical levels. AREAS COVERED This article gives historical perspectives by providing background about present-day economic realities and medical needs for antibiotic research, whose pipeline is mostly focused toward older known agents and newer semi-synthetic derivatives. Future research trends and projected technological developments open many innovative opportunities to discover novel antibacterials and find ways to control pathogenic bacteria without conventional antibiotics that provoke resistance. EXPERT OPINION The successful registration of daptomycin, retapamulin and fidaxomicin indicate the re-emergence of natural products has already begun. Semi-synthetic derivatives from other under-explored classes are progressing. More effort is being put into approaches such as total synthesis, discovery of new structural scaffolds for synthesis, alterations of biosynthetic pathways, combinatorial biosynthesis, new screening targets and new resources from which to isolate natural products. A return to successful screening of actinomycetes depends on solving the rate-limiting dereplication obstacle. Long-term solutions need to come from greater exploration of the massive numbers of uncultured microbes. An ultimate solution to the antibiotic-promoted microbial resistance cycle may lie in finding ways to control bacteria by non-lethal means.
Collapse
|
1003
|
Marceau AH, Bernstein DA, Walsh BW, Shapiro W, Simmons LA, Keck JL. Protein interactions in genome maintenance as novel antibacterial targets. PLoS One 2013; 8:e58765. [PMID: 23536821 PMCID: PMC3594151 DOI: 10.1371/journal.pone.0058765] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 02/06/2013] [Indexed: 11/19/2022] Open
Abstract
Antibacterial compounds typically act by directly inhibiting essential bacterial enzyme activities. Although this general mechanism of action has fueled traditional antibiotic discovery efforts for decades, new antibiotic development has not kept pace with the emergence of drug resistant bacterial strains. These limitations have severely restricted the therapeutic tools available for treating bacterial infections. Here we test an alternative antibacterial lead-compound identification strategy in which essential protein-protein interactions are targeted rather than enzymatic activities. Bacterial single-stranded DNA-binding proteins (SSBs) form conserved protein interaction “hubs” that are essential for recruiting many DNA replication, recombination, and repair proteins to SSB/DNA nucleoprotein substrates. Three small molecules that block SSB/protein interactions are shown to have antibacterial activity against diverse bacterial species. Consistent with a model in which the compounds target multiple SSB/protein interactions, treatment of Bacillus subtilis cultures with the compounds leads to rapid inhibition of DNA replication and recombination, and ultimately to cell death. The compounds also have unanticipated effects on protein synthesis that could be due to a previously unknown role for SSB/protein interactions in translation or to off-target effects. Our results highlight the potential of targeting protein-protein interactions, particularly those that mediate genome maintenance, as a powerful approach for identifying new antibacterial compounds.
Collapse
Affiliation(s)
- Aimee H. Marceau
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Douglas A. Bernstein
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Brian W. Walsh
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Walker Shapiro
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Lyle A. Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - James L. Keck
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
1004
|
Antibacterial activity of CTBT (7-chlorotetrazolo[5,1-c]benzo[1,2,4]triazine) generating reactive oxygen species. Microbiol Res 2013. [DOI: 10.1016/j.micres.2012.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
1005
|
D'Elia RV, Harrison K, Oyston PC, Lukaszewski RA, Clark GC. Targeting the "cytokine storm" for therapeutic benefit. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:319-27. [PMID: 23283640 PMCID: PMC3592351 DOI: 10.1128/cvi.00636-12] [Citation(s) in RCA: 239] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation is the body's first line of defense against infection or injury, responding to challenges by activating innate and adaptive responses. Microbes have evolved a diverse range of strategies to avoid triggering inflammatory responses. However, some pathogens, such as the influenza virus and the Gram-negative bacterium Francisella tularensis, do trigger life-threatening "cytokine storms" in the host which can result in significant pathology and ultimately death. For these diseases, it has been proposed that downregulating inflammatory immune responses may improve outcome. We review some of the current candidates for treatment of cytokine storms which may prove useful in the clinic in the future and compare them to more traditional therapeutic candidates that target the pathogen rather than the host response.
Collapse
|
1006
|
Trzoss M, Bensen DC, Li X, Chen Z, Lam T, Zhang J, Creighton CJ, Cunningham ML, Kwan B, Stidham M, Nelson K, Brown-Driver V, Castellano A, Shaw KJ, Lightstone FC, Wong SE, Nguyen TB, Finn J, Tari LW. Pyrrolopyrimidine inhibitors of DNA gyrase B (GyrB) and topoisomerase IV (ParE), Part II: Development of inhibitors with broad spectrum, Gram-negative antibacterial activity. Bioorg Med Chem Lett 2013; 23:1537-43. [DOI: 10.1016/j.bmcl.2012.11.073] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 10/27/2022]
|
1007
|
Staphylococcus aureus SasA is responsible for binding to the salivary agglutinin gp340, derived from human saliva. Infect Immun 2013; 81:1870-9. [PMID: 23439307 DOI: 10.1128/iai.00011-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen that can colonize the nasal cavity, skin, intestine, and oral cavity as a commensal bacterium. gp340, also known as DMBT1 (deleted in malignant brain tumors 1), is associated with epithelial differentiation and innate immunity. In the oral cavity, gp340 induces salivary aggregation with several oral bacteria and promotes bacterial adhesion to tissues such as the teeth and mucosa. S. aureus is often isolated from the oral cavity, but the mechanism underlying its persistence in the oral cavity remains unclear. In this study, we investigated the interaction between S. aureus and gp340 and found that S. aureus interacts with saliva- and gp340-coated resin. We then identified the S. aureus factor(s) responsible for binding to gp340. The cell surface protein SasA, which is rich in basic amino acids (BR domain) at the N terminus, was responsible for binding to gp340. Inactivation of the sasA gene resulted in a significant decrease in S. aureus binding to gp340-coated resin. Also, recombinant SasA protein (rSasA) showed binding affinity to gp340, which was inhibited by the addition of N-acetylneuraminic acid. Surface plasmon resonance analysis showed that rSasA significantly bound to the NeuAcα(2-3)Galβ(1-4)GlcNAc structure. These results indicate that SasA is responsible for binding to gp340 via the N-acetylneuraminic acid moiety.
Collapse
|
1008
|
Wencewicz TA, Long TE, Möllmann U, Miller MJ. Trihydroxamate siderophore-fluoroquinolone conjugates are selective sideromycin antibiotics that target Staphylococcus aureus. Bioconjug Chem 2013; 24:473-86. [PMID: 23350642 DOI: 10.1021/bc300610f] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Siderophores are multidentate iron(III) chelators used by bacteria for iron assimilation. Sideromycins, also called siderophore-antibiotic conjugates, are a unique subset of siderophores that enter bacterial cells via siderophore uptake pathways and deliver the toxic antibiotic in a "Trojan horse" fashion. Sideromycins represent a novel antibiotic delivery technology with untapped potential for developing sophisticated microbe-selective antibacterial agents that limit the emergence of bacterial resistance. The chemical synthesis of a series of mono-, bis-, and trihydroxamate sideromycins are described here along with their biological evaluation in antibacterial susceptibility assays. The linear hydroxamate siderophores used for the sideromycins in this study were derived from the ferrioxamine family and inspired by the naturally occurring salmycin sideromycins. The antibacterial agents used were a β-lactam carbacepholosporin, Lorabid, and a fluoroquinolone, ciprofloxacin, chosen for the different locations of their biological targets, the periplasm (extracellular) and the cytoplasm (intracellular). The linear hydroxamate-based sideromycins were selectively toxic toward Gram-positive bacteria, especially Staphylococcus aureus SG511 (MIC = 1.0 μM for the trihydroxamate-fluoroquinolone sideromycin). Siderophore-sideromycin competition assays demonstrated that only the fluoroquinolone sideromycins required membrane transport to reach their cytoplasmic biological target and that a trihydroxamate siderophore backbone was required for protein-mediated active transport of the sideromycins into S. aureus cells via siderophore uptake pathways. This work represents a comprehensive study of linear hydroxamate sideromycins and teaches how to build effective hydroxamate-based sideromycins as Gram-positive selective antibiotic agents.
Collapse
Affiliation(s)
- Timothy A Wencewicz
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | |
Collapse
|
1009
|
Wargo MJ. Choline catabolism to glycine betaine contributes to Pseudomonas aeruginosa survival during murine lung infection. PLoS One 2013; 8:e56850. [PMID: 23457628 PMCID: PMC3572970 DOI: 10.1371/journal.pone.0056850] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/15/2013] [Indexed: 12/31/2022] Open
Abstract
Pseudomonas aeruginosa can acquire and metabolize a variety of molecules including choline, an abundant host-derived molecule. In P. aeruginosa, choline is oxidized to glycine betaine which can be used as an osmoprotectant, a sole source of carbon and nitrogen, and as an inducer of the virulence factor, hemolytic phospholipase C (PlcH) via the transcriptional regulator GbdR. The primary objective was to determine the contribution of choline conversion to glycine betaine to P. aeruginosa survival during mouse lung infection. A secondary objective was to gain insight into the relative contributions of the different roles of glycine betaine to P. aeruginosa survival during infection. Using a model of acute murine pneumonia, we determined that deletion of the choline oxidase system (encoded by betBA) decreased P. aeruginosa survival in the mouse lung. Deletion of the glycine betaine demethylase genes (gbcA-B), required for glycine betaine catabolism, did not impact P. aeruginosa survival in the lung. Thus, the defect of the betBA mutant was not due to a requirement for glycine betaine catabolism or dependence on a downstream metabolite. Deletion of betBA decreased the abundance of plcH transcript during infection, which suggested a role for PlcH in the betBA survival defect. To test the contribution of plcH to the betBA mutant phenotype a betBAplcHR double deletion mutant was generated. The betBA and betBAplcHR double mutant had a small but significant survival defect compared to the plcHR single mutant, suggesting that regulation of plcH expression is not the only role for glycine betaine during infection. The conclusion was that choline acquisition and its oxidation to glycine betaine contribute to P. aeruginosa survival in the mouse lung. While defective plcH induction can explain a portion of the betBA mutant phenotype, the exact mechanisms driving the betBA mutant survival defect remain unknown.
Collapse
Affiliation(s)
- Matthew J Wargo
- Department of Microbiology and Molecular Genetics and The Vermont Lung Center, University of Vermont College of Medicine, Burlington, Vermont, United States of America.
| |
Collapse
|
1010
|
Peetsch A, Greulich C, Braun D, Stroetges C, Rehage H, Siebers B, Köller M, Epple M. Silver-doped calcium phosphate nanoparticles: Synthesis, characterization, and toxic effects toward mammalian and prokaryotic cells. Colloids Surf B Biointerfaces 2013; 102:724-9. [DOI: 10.1016/j.colsurfb.2012.09.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 09/25/2012] [Accepted: 09/26/2012] [Indexed: 01/22/2023]
|
1011
|
Navarro-Navarro M, Ruiz-Bustos P, Valencia D, Robles-Zepeda R, Ruiz-Bustos E, Virués C, Hernandez J, Domínguez Z, Velazquez C. Antibacterial activity of Sonoran propolis and some of its constituents against clinically significant Vibrio species. Foodborne Pathog Dis 2013; 10:150-8. [PMID: 23351031 DOI: 10.1089/fpd.2012.1318] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The aim of the present study was to evaluate the anti-Vibrio activity of propolis collected from three different areas of the Sonoran Desert in northwestern, Mexico [Pueblo de Alamos (PAP), Ures (UP), and Caborca (CP)]. The anti-Vibrio spp. activity of Sonoran propolis was determined by the broth microdilution method. UP propolis showed the highest antibacterial activity [minimal inhibitory concentration (MIC(50))<50 μg mL(-1)] against Vibrio spp. (UP>CP>PAP). UP propolis significantly inhibited the growth of Vibrio cholerae O1 serotype Inaba (MIC(50)<50 μg mL(-1)), V. cholerae non-O1 (MIC(50)<50 μg mL(-1)), V. vulnificus (MIC(50)<50 μg mL(-1)), and V. cholerae O1 serotype Ogawa (MIC(50) 100 μg mL(-1)), in a concentration-dependent manner. The UP propolis constituents, galangin and caffeic acid phenethyl ester (CAPE), exhibited a potent growth inhibitory activity (MIC(50) 0.05-0.1 mmol l(-1)) against V. cholerae strains (non-O1 and serotype Ogawa). The strong anti-Vibrio activity of Sonoran propolis and some of its chemical constituents (galangin and CAPE) support further studies on the clinical applications of this natural bee product against different Vibrio spp., mainly V. cholerae.
Collapse
|
1012
|
Structures of a Na+-coupled, substrate-bound MATE multidrug transporter. Proc Natl Acad Sci U S A 2013; 110:2099-104. [PMID: 23341609 DOI: 10.1073/pnas.1219901110] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multidrug transporters belonging to the multidrug and toxic compound extrusion (MATE) family expel dissimilar lipophilic and cationic drugs across cell membranes by dissipating a preexisting Na(+) or H(+) gradient. Despite its clinical relevance, the transport mechanism of MATE proteins remains poorly understood, largely owing to a lack of structural information on the substrate-bound transporter. Here we report crystal structures of a Na(+)-coupled MATE transporter NorM from Neisseria gonorrheae in complexes with three distinct translocation substrates (ethidium, rhodamine 6G, and tetraphenylphosphonium), as well as Cs(+) (a Na(+) congener), all captured in extracellular-facing and drug-bound states. The structures revealed a multidrug-binding cavity festooned with four negatively charged amino acids and surprisingly limited hydrophobic moieties, in stark contrast to the general belief that aromatic amino acids play a prominent role in multidrug recognition. Furthermore, we discovered an uncommon cation-π interaction in the Na(+)-binding site located outside the drug-binding cavity and validated the biological relevance of both the substrate- and cation-binding sites by conducting drug resistance and transport assays. Additionally, we uncovered potential rearrangement of at least two transmembrane helices upon Na(+)-induced drug export. Based on our structural and functional analyses, we suggest that Na(+) triggers multidrug extrusion by inducing protein conformational changes rather than by directly competing for the substrate-binding amino acids. This scenario is distinct from the canonical antiport mechanism, in which both substrate and counterion compete for a shared binding site in the transporter. Collectively, our findings provide an important step toward a detailed and mechanistic understanding of multidrug transport.
Collapse
|
1013
|
Los FCO, Ha C, Aroian RV. Neuronal Goα and CAPS regulate behavioral and immune responses to bacterial pore-forming toxins. PLoS One 2013; 8:e54528. [PMID: 23349920 PMCID: PMC3547950 DOI: 10.1371/journal.pone.0054528] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 12/12/2012] [Indexed: 12/13/2022] Open
Abstract
Pore-forming toxins (PFTs) are abundant bacterial virulence factors that attack host cell plasma membranes. Host defense mechanisms against PFTs described to date all function in the host tissue that is directly attacked by the PFT. Here we characterize a rapid and fully penetrant cessation of feeding of Caenorhabditis elegans in response to PFT attack. We demonstrate via analyses of C. elegans mutants that inhibition of feeding by PFT requires the neuronal G protein Goα subunit goa-1, and that maintenance of this response requires neuronally expressed calcium activator for protein secretion (CAPS) homolog unc-31. Independently from their role in feeding cessation, we find that goa-1 and unc-31 are additionally required for immune protection against PFTs. We thus demonstrate that the behavioral and immune responses to bacterial PFT attack involve the cross-talk between the nervous system and the cells directly under attack.
Collapse
Affiliation(s)
- Ferdinand C. O. Los
- University of California San Diego, Division of Biological Sciences, Section of cell and developmental biology, La Jolla, California, United States of America
| | - Christine Ha
- University of California San Diego, Division of Biological Sciences, Section of cell and developmental biology, La Jolla, California, United States of America
| | - Raffi V. Aroian
- University of California San Diego, Division of Biological Sciences, Section of cell and developmental biology, La Jolla, California, United States of America
| |
Collapse
|
1014
|
Draft genome sequence of Paenibacillus sp. strain OSY-SE, a bacterium producing the novel broad-spectrum lipopeptide antibiotic paenibacterin. J Bacteriol 2013; 194:6306. [PMID: 23105053 DOI: 10.1128/jb.01506-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A strain of Paenibacillus sp., OSY-SE, was isolated from soil and found to produce a novel lipopeptide antibiotic. The antibiotic, paenibacterin, is active against Gram-negative and Gram-positive bacterial pathogens. Paenibacterin is biosynthesized by a nonribosomal peptide synthetase pathway. Here we report the draft genome sequence of Paenibacillus sp. OSY-SE.
Collapse
|
1015
|
Savoia D. Plant-derived antimicrobial compounds: alternatives to antibiotics. Future Microbiol 2013; 7:979-90. [PMID: 22913356 DOI: 10.2217/fmb.12.68] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The increasing incidence of drug-resistant pathogens has drawn the attention of the pharmaceutical and scientific communities towards studies on the potential antimicrobial activity of plant-derived substances, an untapped source of antimicrobial chemotypes, which are used in traditional medicine in different countries. The aim of this review is to provide recent insights regarding the possibilities of the most important natural antimicrobial compounds derived from plant sources containing a wide variety of secondary metabolites, which are useful as alternative strategies to control infectious diseases. This review will focus on natural plant products as a useful source of antimicrobial molecules, active in particular, on bacteria and fungi. When considering that many of these compounds, which have been used for centuries, are a source of new drugs and that there are ever-increasing technical breakthroughs, it can be envisaged that in the next years some different molecules discovered by ingenious screening programs and obtained from different plant oils and extracts will become useful therapeutic tools.
Collapse
Affiliation(s)
- Dianella Savoia
- Department of Clinical & Biological Sciences, University of Torino, Turin, Italy.
| |
Collapse
|
1016
|
Mitchell GJ, Wiesenfeld K, Nelson DC, Weitz JS. Critical cell wall hole size for lysis in Gram-positive bacteria. J R Soc Interface 2013; 10:20120892. [PMID: 23303219 PMCID: PMC3565739 DOI: 10.1098/rsif.2012.0892] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Gram-positive bacteria can transport molecules necessary for their survival through holes in their cell wall. The holes in cell walls need to be large enough to let critical nutrients pass through. However, the cell wall must also function to prevent the bacteria's membrane from protruding through a large hole into the environment and lysing the cell. As such, we hypothesize that there exists a range of cell wall hole sizes that allow for molecule transport but prevent membrane protrusion. Here, we develop and analyse a biophysical theory of the response of a Gram-positive cell's membrane to the formation of a hole in the cell wall. We predict a critical hole size in the range of 15-24 nm beyond which lysis occurs. To test our theory, we measured hole sizes in Streptococcus pyogenes cells undergoing enzymatic lysis via transmission electron microscopy. The measured hole sizes are in strong agreement with our theoretical prediction. Together, the theory and experiments provide a means to quantify the mechanisms of death of Gram-positive cells via enzymatically mediated lysis and provides insights into the range of cell wall hole sizes compatible with bacterial homeostasis.
Collapse
|
1017
|
Murphy RJ. Preventing multidrug-resistant gram-negative organisms in surgical patients. AORN J 2013; 96:315-29. [PMID: 22935259 DOI: 10.1016/j.aorn.2012.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 01/30/2012] [Accepted: 04/22/2012] [Indexed: 10/28/2022]
Abstract
Multidrug-resistant gram-positive infections have consumed the attention of health care organizations as well as the media, but recently multidrug-resistant gram-negative organisms (MDR-GNOs) have become more and more prevalent. Limited treatment options are available for MDR-GNO infections; thus, prevention is key. Patients who are at high risk for developing infections must be identified, and specialized prevention interventions must be developed for their care. Surgical patients are one subset of patients at high risk for developing MDR-GNOs. Advanced practice nurses must support nurses and other health care personnel in preventing MDR-GNO infections. Education should include active surveillance, contact precautions, cohorting patients and staff members, unit closures, reinforced hand hygiene practices, and environmental cleaning tailored to the care of surgical patients in regard to their preoperative, intraoperative, and postoperative care.
Collapse
|
1018
|
Palermo EF, Vemparala S, Kuroda K. Antimicrobial Polymers: Molecular Design as Synthetic Mimics of Host-Defense Peptides. ACS SYMPOSIUM SERIES 2013. [DOI: 10.1021/bk-2013-1135.ch019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Edmund F. Palermo
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Satyavani Vemparala
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kenichi Kuroda
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
1019
|
Chen YW, Wang H, Hupert M, Soper SA. Identification of methicillin-resistant Staphylococcus aureus using an integrated and modular microfluidic system. Analyst 2013; 138:1075-83. [DOI: 10.1039/c2an36430a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
1020
|
Cheng P, Clive DLJ, Fernandopulle S, Chen Z. Racemic marinopyrrole B by total synthesis. Chem Commun (Camb) 2013; 49:558-60. [DOI: 10.1039/c2cc37110c] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
1021
|
Shehzad A, Rehman G, Ul-Islam M, Khattak WA, Lee YS. Challenges in the development of drugs for the treatment of tuberculosis. Braz J Infect Dis 2013; 17:74-81. [PMID: 23287547 PMCID: PMC9427384 DOI: 10.1016/j.bjid.2012.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 10/02/2012] [Accepted: 10/03/2012] [Indexed: 11/17/2022] Open
Abstract
Tuberculosis infection is a serious human health threat and the early 21st century has seen a remarkable increase in global tuberculosis activity. The pathogen responsible for tuberculosis is Mycobacterium tuberculosis, which adopts diverse strategies in order to survive in a variety of host lesions. These survival mechanisms make the pathogen resistant to currently available drugs, a major contributing factor in the failure to control the spread of tuberculosis. Multiple drugs are available for clinical use and several potential compounds are being screened, synthesized, or evaluated in preclinical or clinical studies. Lasting and effective achievements in the development of anti-tuberculosis drugs will depend largely on the proper understanding of the complex interactions between the pathogen and its human host. Ample evidence exists to explain the characteristics of tuberculosis. In this study, we highlighted the challenges for the development of novel drugs with potent bacteriostatic or bactericidal activity, which reduce the minimum time required to cure tuberculosis infection.
Collapse
Affiliation(s)
- Adeeb Shehzad
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | | | | | | | | |
Collapse
|
1022
|
Jeong YC, Anwar M, Bikadi Z, Hazai E, Moloney MG. Natural product inspired antibacterial tetramic acid libraries with dual enzyme inhibition. Chem Sci 2013. [DOI: 10.1039/c2sc21713a] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
1023
|
Park SR, Park JW, Ban YH, Sohng JK, Yoon YJ. 2-Deoxystreptamine-containing aminoglycoside antibiotics: Recent advances in the characterization and manipulation of their biosynthetic pathways. Nat Prod Rep 2013. [DOI: 10.1039/c2np20092a] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
1024
|
Stokes NR, Baker N, Bennett JM, Berry J, Collins I, Czaplewski LG, Logan A, Macdonald R, Macleod L, Peasley H, Mitchell JP, Nayal N, Yadav A, Srivastava A, Haydon DJ. An improved small-molecule inhibitor of FtsZ with superior in vitro potency, drug-like properties, and in vivo efficacy. Antimicrob Agents Chemother 2013; 57:317-25. [PMID: 23114779 PMCID: PMC3535900 DOI: 10.1128/aac.01580-12] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 10/22/2012] [Indexed: 02/07/2023] Open
Abstract
The bacterial cell division protein FtsZ is an attractive target for small-molecule antibacterial drug discovery. Derivatives of 3-methoxybenzamide, including compound PC190723, have been reported to be potent and selective antistaphylococcal agents which exert their effects through the disruption of intracellular FtsZ function. Here, we report the further optimization of 3-methoxybenzamide derivatives towards a drug candidate. The in vitro and in vivo characterization of a more advanced lead compound, designated compound 1, is described. Compound 1 was potently antibacterial, with an average MIC of 0.12 μg/ml against all staphylococcal species, including methicillin- and multidrug-resistant Staphylococcus aureus and Staphylococcus epidermidis. Compound 1 inhibited an S. aureus strain carrying the G196A mutation in FtsZ, which confers resistance to PC190723. Like PC190723, compound 1 acted on whole bacterial cells by blocking cytokinesis. No interactions between compound 1 and a diverse panel of antibiotics were measured in checkerboard experiments. Compound 1 displayed suitable in vitro pharmaceutical properties and a favorable in vivo pharmacokinetic profile following intravenous and oral administration, with a calculated bioavailability of 82.0% in mice. Compound 1 demonstrated efficacy in a murine model of systemic S. aureus infection and caused a significant decrease in the bacterial load in the thigh infection model. A greater reduction in the number of S. aureus cells recovered from infected thighs, equivalent to 3.68 log units, than in those recovered from controls was achieved using a succinate prodrug of compound 1, which was designated compound 2. In summary, optimized derivatives of 3-methoxybenzamide may yield a first-in-class FtsZ inhibitor for the treatment of antibiotic-resistant staphylococcal infections.
Collapse
Affiliation(s)
- Neil R Stokes
- Biota Europe Ltd., Begbroke Science Park, Oxfordshire, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1025
|
Goswami S, Adhikari MD, Kar C, Thiyagarajan D, Das G, Ramesh A. Synthetic amphiphiles as therapeutic antibacterials: lessons on bactericidal efficacy and cytotoxicity and potential application as an adjuvant in antimicrobial chemotherapy. J Mater Chem B 2013; 1:2612-2623. [DOI: 10.1039/c3tb20226g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
1026
|
Bourne CR, Wakeham N, Nammalwar B, Tseitin V, Bourne PC, Barrow EW, Mylvaganam S, Ramnarayan K, Bunce RA, Berlin KD, Barrow WW. Structure-activity relationship for enantiomers of potent inhibitors of B. anthracis dihydrofolate reductase. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1834:46-52. [PMID: 22999981 PMCID: PMC3530638 DOI: 10.1016/j.bbapap.2012.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/09/2012] [Accepted: 09/04/2012] [Indexed: 01/07/2023]
Abstract
BACKGROUND Bacterial resistance to antibiotic therapies is increasing and new treatment options are badly needed. There is an overlap between these resistant bacteria and organisms classified as likely bioterror weapons. For example, Bacillus anthracis is innately resistant to the anti-folate trimethoprim due to sequence changes found in the dihydrofolate reductase enzyme. Development of new inhibitors provides an opportunity to enhance the current arsenal of anti-folate antibiotics while also expanding the coverage of the anti-folate class. METHODS We have characterized inhibitors of B. anthracis dihydrofolate reductase by measuring the K(i) and MIC values and calculating the energetics of binding. This series contains a core diaminopyrimidine ring, a central dimethoxybenzyl ring, and a dihydrophthalazine moiety. We have altered the chemical groups extended from a chiral center on the dihydropyridazine ring of the phthalazine moiety. The interactions for the most potent compounds were visualized by X-ray structure determination. RESULTS We find that the potency of individual enantiomers is divergent with clear preference for the S-enantiomer, while maintaining a high conservation of contacts within the binding site. The preference for enantiomers seems to be predicated largely by differential interactions with protein residues Leu29, Gln30 and Arg53. CONCLUSIONS These studies have clarified the activity of modifications and of individual enantiomers, and highlighted the role of the less-active R-enantiomer in effectively diluting the more active S-enantiomer in racemic solutions. This directly contributes to the development of new antimicrobials, combating trimethoprim resistance, and treatment options for potential bioterrorism agents.
Collapse
Affiliation(s)
- Christina R. Bourne
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078,Corresponding authors: CRB: phone +1 (405) 744-6737 fax +1 (405) 744-5275 , WWB: phone +1 (405) 744-1842 fax +1 (405) 744-3738
| | - Nancy Wakeham
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Baskar Nammalwar
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078
| | | | - Philip C. Bourne
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Esther W. Barrow
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078
| | | | | | - Richard A. Bunce
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078
| | - K. Darrell Berlin
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078
| | - William W. Barrow
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078,Corresponding authors: CRB: phone +1 (405) 744-6737 fax +1 (405) 744-5275 , WWB: phone +1 (405) 744-1842 fax +1 (405) 744-3738
| |
Collapse
|
1027
|
Abstract
The search for small molecules with activity against Mycobacterium tuberculosis increasingly uses -high-throughput screening and computational methods. Previously, we have analyzed recent studies in which computational tools were used for cheminformatics. We have now updated this analysis to illustrate how they may assist in finding desirable leads for tuberculosis drug discovery. We provide our thoughts on strategies for drug discovery efforts for neglected diseases.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations in Chemistry, Fuquay Varina, NC, USA
| | | |
Collapse
|
1028
|
Orlov AV, Khodakova JA, Nikitin MP, Shepelyakovskaya AO, Brovko FA, Laman AG, Grishin EV, Nikitin PI. Magnetic Immunoassay for Detection of Staphylococcal Toxins in Complex Media. Anal Chem 2012; 85:1154-63. [DOI: 10.1021/ac303075b] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Alexey V. Orlov
- A. M. Prokhorov General Physics
Institute, Russian Academy of Sciences,
38 Vavilova St. Moscow 119991 Russia
- Moscow Institute of Physics and Technology, Moscow, Russia
| | - Julia A. Khodakova
- A. M. Prokhorov General Physics
Institute, Russian Academy of Sciences,
38 Vavilova St. Moscow 119991 Russia
| | - Maxim P. Nikitin
- A. M. Prokhorov General Physics
Institute, Russian Academy of Sciences,
38 Vavilova St. Moscow 119991 Russia
- Moscow Institute of Physics and Technology, Moscow, Russia
- Shemyakin-Ovchinnikov Institute
of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anna O. Shepelyakovskaya
- Pushchino
Branch of Shemyakin-Ovchinnikov
Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Fedor A. Brovko
- Pushchino
Branch of Shemyakin-Ovchinnikov
Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Alexander G. Laman
- Pushchino
Branch of Shemyakin-Ovchinnikov
Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Evgeny V. Grishin
- Shemyakin-Ovchinnikov Institute
of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Petr I. Nikitin
- A. M. Prokhorov General Physics
Institute, Russian Academy of Sciences,
38 Vavilova St. Moscow 119991 Russia
| |
Collapse
|
1029
|
Kim PS, Shin NR, Kim JY, Yun JH, Hyun DW, Bae JW. Gibbsiella papilionis sp. nov., isolated from the intestinal tract of the butterfly Mycalesis gotama, and emended description of the genus Gibbsiella. Int J Syst Evol Microbiol 2012; 63:2607-2611. [PMID: 23264501 DOI: 10.1099/ijs.0.045880-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A novel Gram-negative, non-motile, facultative anaerobic and rod-shaped bacterium, designated strain LEN33(T), was isolated from the intestinal tract of a butterfly (Mycalesis gotama). Strain LEN33(T) grew optimally at 37 °C in the presence of 1 % (w/v) NaCl and at pH 9. The novel strain was oxidase-negative and catalase-positive. The major cellular fatty acids were C14 : 0, C16 : 0 and cyclo-C17 : 0. Strain LEN33(T) contained two unidentified lipids, three unidentified amino-phospholipids, phosphatidylethanolamine (PE) and phosphatidylglycerol (PG). The major isoprenoid quinone was ubiquinone-8 (Q-8). Phylogenetic analysis based on 16S rRNA gene sequences indicated that strain LEN33(T) was most closely related to Gibbsiella quercinecans FRB 97(T) and Gibbsiella dentisursi NUM 1720(T), with 98.7 % similarities. DNA-DNA hybridization experiments indicated less than 40.7 ± 2 % relatedness to the closest phylogenetic species, G. quercinecans FRB 97(T). The G+C content of genomic DNA was 58.7 mol%. Phenotypic, phylogenetic and genotypic analysis indicated that strain LEN33(T) represents a novel species within the genus Gibbsiella, for which the name Gibbsiella papilionis is proposed. The type strain is referred to as LEN33(T) ( = KACC 16707(T) = JCM 18389(T)). An emended description of the genus Gibbsiella is also proposed.
Collapse
Affiliation(s)
- Pil Soo Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul 130-701, Korea
| | - Na-Ri Shin
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul 130-701, Korea
| | - Joon Yong Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul 130-701, Korea
| | - Ji-Hyun Yun
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul 130-701, Korea
| | - Dong-Wook Hyun
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul 130-701, Korea
| | - Jin-Woo Bae
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul 130-701, Korea
| |
Collapse
|
1030
|
Moltzahn F, Haeni K, Birkhäuser FD, Roth B, Thalmann GN, Zehnder P. Peri-interventional antibiotic prophylaxis only vs continuous low-dose antibiotic treatment in patients with JJ stents: a prospective randomised trial analysing the effect on urinary tract infections and stent-related symptoms. BJU Int 2012; 111:289-95. [PMID: 23253774 DOI: 10.1111/j.1464-410x.2012.11592.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To evaluate the antibiotic treatment regime in patients with indwelling JJ stents, the benefits and disadvantages of a peri-interventional antibiotic prophylaxis were compared with those of a continuous low-dose antibiotic treatment in a prospective randomised trial. PATIENTS AND METHODS In all, 95 patients were randomised to either receive peri-interventional antibiotic prophylaxis during stent insertion only (group A, 44 patients) or to additionally receive a continuous low-dose antibiotic treatment until stent removal (group B, 51). Evaluations for urinary tract infections (UTI), stent-related symptoms (SRSs) and drug side-effects were performed before stent insertion and consecutively after 1, 2 and 4 weeks and/or at stent withdrawal. All patients received a peri-interventional antibiotic prophylaxis with 1.2 g amoxicillin/clavulanic acid. Amoxicillin/clavulanic acid (625 mg) once daily was administered for continuous low-dose treatment (group B). Primary endpoints were the overall rates of UTIs and SRSs. Secondary endpoints were the rates and severity of drug side-effects. RESULTS Neither the overall UTI rates (group A: 9% vs group B: 10%), nor the rates of febrile UTIs (group A: 7% vs group B: 6%) were different between the groups. Similarly, SRS rates did not differ (group A: 98% vs group B: 96%). Antibiotic side-effect symptoms were to be increased in patients treated with low-dose antibiotics. CONCLUSION A continuous antibiotic low-dose treatment during the entire JJ stent-indwelling time does not reduce the quantity or severity of UTIs and has no effect on SRSs either compared with a peri-interventional antibiotic prophylaxis only.
Collapse
|
1031
|
Lu J, Vlamis‐Gardikas A, Kandasamy K, Zhao R, Gustafsson TN, Engstrand L, Hoffner S, Engman L, Holmgren A. Inhibition of bacterial thioredoxin reductase: an antibiotic mechanism targeting bacteria lacking glutathione. FASEB J 2012; 27:1394-403. [DOI: 10.1096/fj.12-223305] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jun Lu
- Division of BiochemistryDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Alexios Vlamis‐Gardikas
- Division of BiochemistryDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Karuppasamy Kandasamy
- Division of BiochemistryDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Rong Zhao
- Division of BiochemistryDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Tomas N. Gustafsson
- Division of BiochemistryDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Lars Engstrand
- Microbiology and Tumor Biology CenterKarolinska InstitutetStockholmSweden
| | - Sven Hoffner
- Microbiology and Tumor Biology CenterKarolinska InstitutetStockholmSweden
- World Health Organization Supranational Tuberculosis Reference LaboratoryDepartment for PreparednessSwedish Institute for Communicable Disease ControlSolnaSweden
| | - Lars Engman
- Department of Biochemistry and Organic ChemistryUppsala UniversityUppsalaSweden
| | - Arne Holmgren
- Division of BiochemistryDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| |
Collapse
|
1032
|
Zakrzewski P, Medema MH, Gevorgyan A, Kierzek AM, Breitling R, Takano E. MultiMetEval: comparative and multi-objective analysis of genome-scale metabolic models. PLoS One 2012; 7:e51511. [PMID: 23272111 PMCID: PMC3522732 DOI: 10.1371/journal.pone.0051511] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/01/2012] [Indexed: 12/02/2022] Open
Abstract
Comparative metabolic modelling is emerging as a novel field, supported by the development of reliable and standardized approaches for constructing genome-scale metabolic models in high throughput. New software solutions are needed to allow efficient comparative analysis of multiple models in the context of multiple cellular objectives. Here, we present the user-friendly software framework Multi-Metabolic Evaluator (MultiMetEval), built upon SurreyFBA, which allows the user to compose collections of metabolic models that together can be subjected to flux balance analysis. Additionally, MultiMetEval implements functionalities for multi-objective analysis by calculating the Pareto front between two cellular objectives. Using a previously generated dataset of 38 actinobacterial genome-scale metabolic models, we show how these approaches can lead to exciting novel insights. Firstly, after incorporating several pathways for the biosynthesis of natural products into each of these models, comparative flux balance analysis predicted that species like Streptomyces that harbour the highest diversity of secondary metabolite biosynthetic gene clusters in their genomes do not necessarily have the metabolic network topology most suitable for compound overproduction. Secondly, multi-objective analysis of biomass production and natural product biosynthesis in these actinobacteria shows that the well-studied occurrence of discrete metabolic switches during the change of cellular objectives is inherent to their metabolic network architecture. Comparative and multi-objective modelling can lead to insights that could not be obtained by normal flux balance analyses. MultiMetEval provides a powerful platform that makes these analyses straightforward for biologists. Sources and binaries of MultiMetEval are freely available from https://github.com/PiotrZakrzewski/MetEval/downloads.
Collapse
Affiliation(s)
- Piotr Zakrzewski
- Department of Microbial Physiology, University of Groningen, Groningen, The Netherlands
- Groningen Bioinformatics Centre, University of Groningen, Groningen, The Netherlands
| | - Marnix H. Medema
- Department of Microbial Physiology, University of Groningen, Groningen, The Netherlands
- Groningen Bioinformatics Centre, University of Groningen, Groningen, The Netherlands
| | - Albert Gevorgyan
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Andrzej M. Kierzek
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Rainer Breitling
- Groningen Bioinformatics Centre, University of Groningen, Groningen, The Netherlands
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- * E-mail: (RB); (ET)
| | - Eriko Takano
- Department of Microbial Physiology, University of Groningen, Groningen, The Netherlands
- * E-mail: (RB); (ET)
| |
Collapse
|
1033
|
Bioengineering natural product biosynthetic pathways for therapeutic applications. Curr Opin Biotechnol 2012; 23:931-40. [DOI: 10.1016/j.copbio.2012.03.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 03/13/2012] [Indexed: 01/05/2023]
|
1034
|
Biotransformation and biocatalysis: roles and applications in the discovery of antimalarials. Future Med Chem 2012; 4:2325-36. [DOI: 10.4155/fmc.12.173] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Several strategies to discover new antimalarials have been proposed to augment and complement the conventional drug-discovery paradigm. One approach, which has not yet been fully exploited, is the use of drug biotransformation to identify new active molecules. This concept rests on the use of the biotransformation of drugs to their pharmacologically active metabolites. This approach has been used successfully in human chemotherapy, with the discovery and development of several metabolite-based drugs. This review looks at the contribution that biotransformations can play in antimalarial drug discovery.
Collapse
|
1035
|
Puckett SE, Reese KA, Mitev GM, Mullen V, Johnson RC, Pomraning KR, Mellbye BL, Tilley LD, Iversen PL, Freitag M, Geller BL. Bacterial resistance to antisense peptide phosphorodiamidate morpholino oligomers. Antimicrob Agents Chemother 2012; 56:6147-53. [PMID: 22985881 PMCID: PMC3497173 DOI: 10.1128/aac.00850-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 06/25/2012] [Indexed: 12/16/2022] Open
Abstract
Peptide phosphorodiamidate morpholino oligomers (PPMOs) are synthetic DNA mimics that bind cRNA and inhibit bacterial gene expression. The PPMO (RFF)(3)RXB-AcpP (where R is arginine, F, phenylalanine, X is 6-aminohexanoic acid, B is β-alanine, and AcpP is acyl carrier protein) is complementary to 11 bases of the essential gene acpP (which encodes acyl carrier protein). The MIC of (RFF)(3)RXB-AcpP was 2.5 μM (14 μg/ml) in Escherichia coli W3110. The rate of spontaneous resistance of E. coli to (RFF)(3)RXB-AcpP was 4 × 10(-7) mutations/cell division. A spontaneous (RFF)(3)RXB-AcpP-resistant mutant (PR200.1) was isolated. The MIC of (RFF)(3)RXB-AcpP was 40 μM (224 μg/ml) for PR200.1. The MICs of standard antibiotics for PR200.1 and W3110 were identical. The sequence of acpP was identical in PR200.1 and W3110. PR200.1 was also resistant to other PPMOs conjugated to (RFF)(3)RXB or peptides with a similar composition or pattern of cationic and nonpolar residues. Genomic sequencing of PR200.1 identified a mutation in sbmA, which encodes an active transport protein. In separate experiments, a (RFF)(3)RXB-AcpP-resistant isolate (RR3) was selected from a transposome library, and the insertion was mapped to sbmA. Genetic complementation of PR200.1 or RR3 with sbmA restored susceptibility to (RFF)(3)RXB-AcpP. Deletion of sbmA caused resistance to (RFF)(3)RXB-AcpP. We conclude that resistance to (RFF)(3)RXB-AcpP was linked to the peptide and not the phosphorodiamidate morpholino oligomer, dependent on the composition or repeating pattern of amino acids, and caused by mutations in sbmA. The data further suggest that (RFF)(3)R-XB PPMOs may be transported across the plasma membrane by SbmA.
Collapse
Affiliation(s)
| | | | | | | | | | - Kyle R. Pomraning
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | | | | | | | - Michael Freitag
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | - Bruce L. Geller
- Department of Microbiology
- AVI BioPharma, Inc., Corvallis, Oregon, USA
| |
Collapse
|
1036
|
Paknikar SS, Narayana S. Newer antibacterials in therapy and clinical trials. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2012. [PMID: 23181224 PMCID: PMC3503371 DOI: 10.4103/1947-2714.103312] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In order to deal with the rising problem of antibiotic resistance, newer antibacterials are being discovered and added to existing pool. Since the year 2000, however, only four new classes of antibacterials have been discovered. These include the oxazolidinones, glycolipopeptides, glycolipodepepsipeptide and pleuromutilins. Newer drugs were added to existing classes of antibiotics, such as streptogramins, quinolones, beta-lactam antibiotics, and macrolide-, tetracycline- and trimethoprim-related drugs. Most of the antibacterials are directed against resistant S. aureus infections, with very few against resistant gram-negative infections. The following article reviews the antibacterials approved by the FDA after the year 2000 as well as some of those in clinical trials. Data was obtained through a literature search via Pubmed and google as well as a detailed search of our library database.
Collapse
|
1037
|
Morita Y, Tomida J, Kawamura Y. MexXY multidrug efflux system of Pseudomonas aeruginosa. Front Microbiol 2012; 3:408. [PMID: 23233851 PMCID: PMC3516279 DOI: 10.3389/fmicb.2012.00408] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/13/2012] [Indexed: 01/01/2023] Open
Abstract
Anti-pseudomonas aminoglycosides, such as amikacin and tobramycin, are used in the treatment of Pseudomonas aeruginosa infections. However, their use is linked to the development of resistance. During the last decade, the MexXY multidrug efflux system has been comprehensively studied, and numerous reports of laboratory and clinical isolates have been published. This system has been increasingly recognized as one of the primary determinants of aminoglycoside resistance in P. aeruginosa. In P. aeruginosa cystic fibrosis isolates, upregulation of the pump is considered the most common mechanism of aminoglycoside resistance. Non-fermentative Gram-negative pathogens possessing very close MexXY orthologs such as Achromobacter xylosoxidans and various Burkholderia species (e.g., Burkholderia pseudomallei and B. cepacia complexes), but not B. gladioli, are intrinsically resistant to aminoglycosides. Here, we summarize the properties (e.g., discovery, mechanism, gene expression, clinical significance) of the P. aeruginosa MexXY pump and other aminoglycoside efflux pumps such as AcrD of Escherichia coli, AmrAB-OprA of B. pseudomallei, and AdeABC of Acinetobacter baumannii. MexXY inducibility of the PA5471 gene product, which is dependent on ribosome inhibition or oxidative stress, is noteworthy. Moreover, the discovery of the cognate outer membrane component (OprA) of MexXY in the multidrug-resistant clinical isolate PA7, serotype O12 deserves special attention.
Collapse
Affiliation(s)
- Yuji Morita
- Department of Microbiology, School of Pharmacy, Aichi Gakuin University Nagoya, Japan
| | | | | |
Collapse
|
1038
|
Shi Y, Bueno A, van der Donk WA. Heterologous production of the lantibiotic Ala(0)actagardine in Escherichia coli. Chem Commun (Camb) 2012; 48:10966-8. [PMID: 23034674 PMCID: PMC3485686 DOI: 10.1039/c2cc36336d] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We report the heterologous production of Ala(0)actagardine in E. coli by co-expression of the substrate peptide GarA and its modification enzymes GarM and GarO. The activity of GarO, a luciferase-like monooxygenase that introduces the unique sulfoxide group of actagardine, was also investigated in vitro.
Collapse
Affiliation(s)
- Yanxiang Shi
- Department of Chemistry and Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 S Mathews Ave, Urbana, IL 61801, USA. Fax: (217) 244-8533; Tel: (217) 244-5360
| | - Alejandro Bueno
- Department of Chemistry and Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 S Mathews Ave, Urbana, IL 61801, USA. Fax: (217) 244-8533; Tel: (217) 244-5360
| | - Wilfred A. van der Donk
- Department of Chemistry and Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 S Mathews Ave, Urbana, IL 61801, USA. Fax: (217) 244-8533; Tel: (217) 244-5360
| |
Collapse
|
1039
|
Keating TA, Newman JV, Olivier NB, Otterson LG, Andrews B, Boriack-Sjodin PA, Breen JN, Doig P, Dumas J, Gangl E, Green OM, Guler SY, Hentemann MF, Joseph-McCarthy D, Kawatkar S, Kutschke A, Loch JT, McKenzie AR, Pradeepan S, Prasad S, Martínez-Botella G. In vivo validation of thymidylate kinase (TMK) with a rationally designed, selective antibacterial compound. ACS Chem Biol 2012; 7:1866-72. [PMID: 22908966 DOI: 10.1021/cb300316n] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is an urgent need for new antibacterials that pinpoint novel targets and thereby avoid existing resistance mechanisms. We have created novel synthetic antibacterials through structure-based drug design that specifically target bacterial thymidylate kinase (TMK), a nucleotide kinase essential in the DNA synthesis pathway. A high-resolution structure shows compound TK-666 binding partly in the thymidine monophosphate substrate site, but also forming new induced-fit interactions that give picomolar affinity. TK-666 has potent, broad-spectrum Gram-positive microbiological activity (including activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus), bactericidal action with rapid killing kinetics, excellent target selectivity over the human ortholog, and low resistance rates. We demonstrate in vivo efficacy against S. aureus in a murine infected-thigh model. This work presents the first validation of TMK as a compelling antibacterial target and provides a rationale for pursuing novel clinical candidates for treating Gram-positive infections through TMK.
Collapse
Affiliation(s)
- Thomas A. Keating
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Joseph V. Newman
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Nelson B. Olivier
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive,
Waltham, Massachusetts 02451, United States
| | - Linda G. Otterson
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Beth Andrews
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - P. Ann Boriack-Sjodin
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive,
Waltham, Massachusetts 02451, United States
| | - John N. Breen
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive,
Waltham, Massachusetts 02451, United States
| | - Peter Doig
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive,
Waltham, Massachusetts 02451, United States
| | - Jacques Dumas
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Eric Gangl
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Oluyinka M. Green
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Satenig Y. Guler
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Martin F. Hentemann
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Diane Joseph-McCarthy
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Sameer Kawatkar
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Amy Kutschke
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - James T. Loch
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Andrew R. McKenzie
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Selvi Pradeepan
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Swati Prasad
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Gabriel Martínez-Botella
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| |
Collapse
|
1040
|
Xue X, Chen X, Mao X, Hou Z, Zhou Y, Bai H, Meng J, Da F, Sang G, Wang Y, Luo X. Amino-terminated generation 2 poly(amidoamine) dendrimer as a potential broad-spectrum, nonresistance-inducing antibacterial agent. AAPS JOURNAL 2012; 15:132-42. [PMID: 23135925 DOI: 10.1208/s12248-012-9416-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 09/18/2012] [Indexed: 11/30/2022]
Abstract
The treatment of septicemia caused by antibiotic-resistant bacteria is a great challenge in the clinic. Because traditional antibiotics inevitably induce bacterial resistance, which is responsible for many treatment failures, there is an urgent need to develop novel antibiotic drugs. Amino-terminated Poly(amidoamine) dendrimers (PAMAM-NH(2)) are reported to have antibacterial activities. However, previous studies focused on high generations of PAMAM-NH(2), which have been found to exhibit high toxicities. The present study aimed to clarify whether low generations of PAMAM-NH(2) could be used as novel antibacterial agents. We found that generation 2 (G2.0) PAMAM-NH(2) showed significant antibacterial effects against antibiotic-sensitive and antibiotic-resistant strains but exhibited little toxicity to human gastric epithelial cells and did not induce antibiotic resistance in bacteria. Scanning and transmission electron microscopy analyses suggested that G2.0 PAMAM-NH(2) might inhibit the growth of bacteria by destroying their cell membranes. The administration of G2.0 PAMAM-NH(2) dose-dependently improved the animal survival rate of mice infected with extended-spectrum beta lactamase-producing Escherichia coli (ESBL-EC) and of animals infected with a combination of ESBL-EC and methicillin-resistant Staphylococcus aureus. A treatment regimen of 10 mg/kg of G2.0 PAMAM-NH(2) starting 12 h before inoculation followed by 10 mg/kg at 0.5 h after inoculation rescued 100% of singly infected mice and 60% of multiply infected mice. The protective effects were associated with the reduction of the bacterial titers in the blood and with the morphological amelioration of infected tissues. These findings demonstrate that the G2.0 PAMAM-NH(2) is a potential broad-spectrum and nonresistance-inducing antibiotic agent with relatively low toxicity.
Collapse
Affiliation(s)
- Xiaoyan Xue
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, No.169, Changle West Road, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1041
|
Eadsforth TC, Maluf FV, Hunter WN. Acinetobacter baumannii FolD ligand complexes --potent inhibitors of folate metabolism and a re-evaluation of the structure of LY374571. FEBS J 2012; 279:4350-60. [PMID: 23050773 DOI: 10.1111/febs.12025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/07/2012] [Accepted: 10/04/2012] [Indexed: 01/26/2023]
Abstract
The bifunctional N(5),N(10)-methylenetetrahydrofolate dehydrogenase/cyclohydrolase (DHCH or FolD), which is widely distributed in prokaryotes and eukaryotes, is involved in the biosynthesis of folate cofactors that are essential for growth and cellular development. The enzyme activities represent a potential antimicrobial drug target. We have characterized the kinetic properties of FolD from the Gram-negative pathogen Acinetobacter baumanni and determined high-resolution crystal structures of complexes with a cofactor and two potent inhibitors. The data reveal new details with respect to the molecular basis of catalysis and potent inhibition. A unexpected finding was that our crystallographic data revealed a different structure for LY374571 (an inhibitor studied as an antifolate) than that previously published. The implications of this observation are discussed.
Collapse
Affiliation(s)
- Thomas C Eadsforth
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, UK
| | | | | |
Collapse
|
1042
|
Juhas M, Eberl L, Church GM. Essential genes as antimicrobial targets and cornerstones of synthetic biology. Trends Biotechnol 2012; 30:601-7. [DOI: 10.1016/j.tibtech.2012.08.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/02/2012] [Accepted: 08/02/2012] [Indexed: 11/15/2022]
|
1043
|
Agarwal V, Pierce E, McIntosh J, Schmidt E, Nair S. Structures of Cyanobactin Maturation Enzymes Define a Family of Transamidating Proteases. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.chembiol.2012.09.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
1044
|
Novel apidaecin 1b analogs with superior serum stabilities for treatment of infections by gram-negative pathogens. Antimicrob Agents Chemother 2012; 57:402-9. [PMID: 23114765 DOI: 10.1128/aac.01923-12] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Proline-rich antimicrobial peptides (PrAMPs) from insects and mammals have recently been evaluated for their pharmaceutical potential in treating systemic bacterial infections. Besides the native peptides, several shortened, modified, or even artificial sequences were highly effective in different murine infection models. Most recently, we showed that the 18-residue-long peptide Api88, an optimized version of apidaecin 1b, was efficient in two different animal infection models using the pathogenic Escherichia coli strains ATCC 25922 and Neumann, with a promising safety margin. Here, we show that Api88 is degraded relatively fast upon incubation with mouse serum, by cleavage of the C-terminal leucine residue. To improve its in vitro characteristics, we aimed to improve its serum stability. Replacing the C-terminal amide by the free acid or substituting Arg-17 with l-ornithine or l-homoarginine increased the serum stabilities by more than 20-fold (half-life, ∼4 to 6 h). These analogs were nontoxic to human embryonic kidney (HEK 293), human hepatoma (HepG2), SH-SY5Y, and HeLa cells and nonhemolytic to human erythrocytes. The binding constants of all three analogs with the chaperone DnaK, which is proposed as the bacterial target of PrAMPs, were very similar to that of Api88. Of all the analogs tested, Api137 (Gu-ONNRPVYIPRPRPPHPRL; Gu is N,N,N',N'-tetramethylguanidino) appeared most promising due to its high antibacterial activity, which was very similar to Api88. Positional alanine and d-amino acid scans of Api137 indicated that substitutions of residues 1 to 13 had only minor effects on the activity against an E. coli strain, whereas substitutions of residues 14 to 18 decreased the activity dramatically. Based on the significantly improved resistance to proteolysis, Api137 appears to be a very promising lead compound that should be even more efficient in vivo than Api88.
Collapse
|
1045
|
Draft genome sequence of Paenibacillus polymyxa OSY-DF, which coproduces a lantibiotic, paenibacillin, and polymyxin E1. J Bacteriol 2012; 194:4739-40. [PMID: 22887654 DOI: 10.1128/jb.00846-12] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Paenibacillus polymyxa OSY-DF is a Gram-positive rod-shaped bacterium isolated from a fermented vegetable food. This bacterial strain displays potent antimicrobial activities against Gram-positive and Gram-negative pathogenic bacteria, attributed to the production of the lantibiotic paenibacillin and the colistin peptide polymyxin E1. Here we report the draft genome sequence of Paenibacillus polymyxa OSY-DF.
Collapse
|
1046
|
Abstract
If discovery of new antibiotics continues to falter while resistance to drugs in clinical use continues to spread, society's medicine chest will soon lack effective treatments for many infections. Heritable antibiotic resistance emerges in bacteria from nonheritable resistance, also called phenotypic tolerance. This widespread phenomenon is closely linked to nonproliferative states in ways that scientists are just beginning to understand. A deeper understanding of the mechanisms of phenotypic tolerance may reveal new drug targets in the infecting organisms. At the same time, researchers must investigate ways to target the host in order to influence host-pathogen relationships. Government must reform the regulatory process for approval of new antibiotics. The private sector, government, and academia must undertake multiple, organized, multidisciplinary, parallel efforts to improve the ways in which antibiotics are discovered, tested, approved, and conserved, or it will be difficult to sustain the modern practice of medicine.
Collapse
Affiliation(s)
- Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
1047
|
Martínez-Botella G, Breen JN, Duffy JES, Dumas J, Geng B, Gowers IK, Green OM, Guler S, Hentemann MF, Hernandez-Juan FA, Joseph-McCarthy D, Kawatkar S, Larsen NA, Lazari O, Loch JT, Macritchie JA, McKenzie AR, Newman JV, Olivier NB, Otterson LG, Owens AP, Read J, Sheppard DW, Keating TA. Discovery of Selective and Potent Inhibitors of Gram-Positive Bacterial Thymidylate Kinase (TMK). J Med Chem 2012; 55:10010-21. [DOI: 10.1021/jm3011806] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gabriel Martínez-Botella
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - John N. Breen
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | | | - Jacques Dumas
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Bolin Geng
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Ian K. Gowers
- BioFocus, Chesterford Research Park, Saffron Walden
CB10 1XL, U.K
| | - Oluyinka M. Green
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Satenig Guler
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Martin F. Hentemann
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | | | - Diane Joseph-McCarthy
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Sameer Kawatkar
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Nicholas A. Larsen
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Ovadia Lazari
- BioFocus, Chesterford Research Park, Saffron Walden
CB10 1XL, U.K
| | - James T. Loch
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | | | - Andrew R. McKenzie
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Joseph V. Newman
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Nelson B. Olivier
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Linda G. Otterson
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | - Andrew P. Owens
- BioFocus, Chesterford Research Park, Saffron Walden
CB10 1XL, U.K
| | - Jon Read
- AstraZeneca Discovery Sciences, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| | | | - Thomas A. Keating
- AstraZeneca Infection Innovative Medicines, 35 Gatehouse Drive, Waltham, Massachusetts
02451, United States
| |
Collapse
|
1048
|
Bertelsen K, Dorosz J, Hansen SK, Nielsen NC, Vosegaard T. Mechanisms of peptide-induced pore formation in lipid bilayers investigated by oriented 31P solid-state NMR spectroscopy. PLoS One 2012; 7:e47745. [PMID: 23094079 PMCID: PMC3475706 DOI: 10.1371/journal.pone.0047745] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/17/2012] [Indexed: 01/30/2023] Open
Abstract
There is a considerable interest in understanding the function of antimicrobial peptides (AMPs), but the details of their mode of action is not fully understood. This motivates extensive efforts in determining structural and mechanistic parameters for AMP’s interaction with lipid membranes. In this study we show that oriented-sample 31P solid-state NMR spectroscopy can be used to probe the membrane perturbations and -disruption by AMPs. For two AMPs, alamethicin and novicidin, we observe that the majority of the lipids remain in a planar bilayer conformation but that a number of lipids are involved in the peptide anchoring. These lipids display reduced dynamics. Our study supports previous studies showing that alamethicin adopts a transmembrane arrangement without significant disturbance of the surrounding lipids, while novicidin forms toroidal pores at high concentrations leading to more extensive membrane disturbance.
Collapse
Affiliation(s)
- Kresten Bertelsen
- Center for Insoluble Protein Structures (inSPIN), Interdisciplinary Nanoscience Center (iNANO), Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - Jerzy Dorosz
- Center for Insoluble Protein Structures (inSPIN), Interdisciplinary Nanoscience Center (iNANO), Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - Sara Krogh Hansen
- Center for Insoluble Protein Structures (inSPIN), Interdisciplinary Nanoscience Center (iNANO), Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - Niels Chr. Nielsen
- Center for Insoluble Protein Structures (inSPIN), Interdisciplinary Nanoscience Center (iNANO), Department of Chemistry, University of Aarhus, Aarhus, Denmark
- * E-mail: (NCN); (TV)
| | - Thomas Vosegaard
- Center for Insoluble Protein Structures (inSPIN), Interdisciplinary Nanoscience Center (iNANO), Department of Chemistry, University of Aarhus, Aarhus, Denmark
- Department of Engineering, School of Engineering, University of Aarhus, Aarhus, Denmark
- * E-mail: (NCN); (TV)
| |
Collapse
|
1049
|
Kuroda K, Caputo GA. Antimicrobial polymers as synthetic mimics of host-defense peptides. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2012; 5:49-66. [PMID: 23076870 DOI: 10.1002/wnan.1199] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Antibiotic-resistant bacteria 'superbugs' are an emerging threat to public health due to the decrease in effective antibiotics as well as the slowed pace of development of new antibiotics to replace those that become ineffective. The need for new antimicrobial agents is a well-documented issue relating to world health. Tremendous efforts have been given to developing compounds that not only show high efficacy, but also those that are less susceptible to resistance development in the bacteria. However, the development of newer, stronger antibiotics which can overcome these acquired resistances is still a scientific challenge because a new mode of antimicrobial action is likely required. To that end, amphiphilic, cationic polymers have emerged as a promising candidate for further development as an antimicrobial agent with decreased potential for resistance development. These polymers are designed to mimic naturally occurring host-defense antimicrobial peptides which act on bacterial cell walls or membranes. Antimicrobial-peptide mimetic polymers display antibacterial activity against a broad spectrum of bacteria including drug-resistant strains and are less susceptible to resistance development in bacteria. These polymers also showed selective activity to bacteria over mammalian cells. Antimicrobial polymers provide a new molecular framework for chemical modification and adaptation to tune their biological functions. The peptide-mimetic design of antimicrobial polymers will be versatile, generating a new generation of antibiotics toward implementation of polymers in biomedical applications.
Collapse
Affiliation(s)
- Kenichi Kuroda
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA.
| | | |
Collapse
|
1050
|
Velho-Pereira S, Kamat NM. Antimicrobial Screening of Actinobacteria using a Modified Cross-Streak Method. Indian J Pharm Sci 2012; 73:223-8. [PMID: 22303068 PMCID: PMC3267309 DOI: 10.4103/0250-474x.91566] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 01/31/2011] [Accepted: 03/08/2011] [Indexed: 11/28/2022] Open
Abstract
Out of the 30 actinobacterial cultures screened for antimicrobial activity, 28 cultures were found to produce active products against various pathogenic microorganisms such as Gram-negative, Gram-positive bacteria and yeast, using a modified cross streak method. The modified method helped in easy quantification of results and also in ruling out probable mutual antibiosis. The actinobacterial strains that showed the ability to produce antimicrobial compounds belonged to Streptomyces (53%), Micromonospora (13%) and Actinomadura (10%) genera. Streptomyces sp. strain MMA-5 showed the highest multispecific antibiosis efficiency score value. Broad antibiotic spectrum activity was exhibited by Streptomyces sp. strain MMA-2 and Micromonospora sp. strain MMA-8. The multidrug resistant human pathogenic yeast strain Candida albicans was inhibited by 18 actinobacterial strains.
Collapse
|