151
|
Moreau MJJ, Morin I, Askin SP, Cooper A, Moreland NJ, Vasudevan SG, Schaeffer PM. Rapid determination of protein stability and ligand binding by differential scanning fluorimetry of GFP-tagged proteins. RSC Adv 2012. [DOI: 10.1039/c2ra22368f] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
152
|
Fimereli DK, Tsirigos KD, Litou ZI, Liakopoulos TD, Bagos PG, Hamodrakas SJ. CW-PRED: A HMM-Based Method for the Classification of Cell Wall-Anchored Proteins of Gram-Positive Bacteria. LECTURE NOTES IN COMPUTER SCIENCE 2012. [DOI: 10.1007/978-3-642-30448-4_36] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
153
|
Askin SP, Schaeffer PM. A universal immuno-PCR platform for comparative and ultrasensitive quantification of dual affinity-tagged proteins in complex matrices. Analyst 2012; 137:5193-6. [DOI: 10.1039/c2an35857c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
154
|
Paganelli FL, Willems RJ, Leavis HL. Optimizing future treatment of enterococcal infections: attacking the biofilm? Trends Microbiol 2011; 20:40-9. [PMID: 22169461 DOI: 10.1016/j.tim.2011.11.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/19/2011] [Accepted: 11/02/2011] [Indexed: 11/16/2022]
Abstract
Enterococcus faecalis and Enterococcus faecium are among the leading causative agents of nosocomial infections and are infamous for their resistance to many antibiotics. They cause difficult-to-treat infections, often originating from biofilm-mediated infections associated with implanted medical devices or endocarditis. Biofilms protect bacteria against antibiotics and phagocytosis, and physical removal of devices or infected tissue is often needed but is frequently not possible. Currently there are no clinically available compounds that disassemble biofilms. In this review we discuss all known structural and regulatory genes involved in enterococcal biofilm formation, the compounds directed against biofilm formation that have been studied, and potentially useful targets for future drugs to treat enterococcal biofilm-associated infections.
Collapse
Affiliation(s)
- Fernanda L Paganelli
- Department of Medical Microbiology, University Medical Center Utrecht, The Netherlands
| | | | | |
Collapse
|
155
|
Abstract
In Gram-positive bacteria proteins are displayed on the cell surface using sortase enzymes. These cysteine transpeptidases join proteins bearing an appropriate sorting signal to strategically positioned amino groups on the cell surface. Working alone, or in concert with other enzymes, sortases either attach proteins to the cross-bridge peptide of the cell wall or they link proteins together to form pili. Because surface proteins play a fundamental role in microbial physiology and are frequently virulence factors, sortase enzymes have been intensely studied since their discovery a little more than a decade ago. Based on their primary sequences and functions sortases can be partitioned into distinct families called class A to F enzymes. Most bacteria elaborate their surfaces using more than one type of sortase that function non-redundantly by recognizing unique sorting signals within their protein substrates. Here we review what is known about the functions of these enzymes and the molecular basis of catalysis. Particular emphasis is placed on 'pilin' specific class C sortases that construct structurally complex pili. Exciting new data have revealed that these enzymes are amazingly promiscuous in the substrates that they can employ and that there is a startling degree of diversity in their mechanism of action. We also review recent data that suggest that sortases are targeted to specific sites on the cell surface where they work with other sortases and accessory factors to properly function.
Collapse
Affiliation(s)
- Thomas Spirig
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
156
|
Functional characterization and localization of a Bacillus subtilis sortase and its substrate and use of this sortase system to covalently anchor a heterologous protein to the B. subtilis cell wall for surface display. J Bacteriol 2011; 194:161-75. [PMID: 22020651 DOI: 10.1128/jb.05711-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sortases catalyze the covalent anchoring of proteins to the cell surface on Gram-positive bacteria. Bioinformatic analysis suggests the presence of structural genes encoding sortases and their substrates in the Bacillus subtilis genome. In this study, a β-lactamase reporter was fused to the cell wall anchoring domain from a putative sortase substrate, YhcR. Covalent anchoring of this fusion protein to the cell wall was confirmed by using the eight-protease-deficient B. subtilis strain WB800 as the host. Inactivation of yhcS abolished the cell wall anchoring reaction. The amounts of fusion protein anchored to the cell wall were proportional to the levels of YhcS. These data demonstrate that YhcS and YhcR are the sortase and sortase substrate, respectively, in B. subtilis. Furthermore, yhcS is not essential for the survival of B. subtilis under the cultivation condition tested. YhcR fusions were distributed helically in the lateral cell wall. Interestingly, when viewed with an epifluorescence microscope, YhcS also appeared to form short helical arcs. This is the first report to illustrate such distribution of sortases in a rod-shaped bacterium. Models for the spatial distribution of both the sortase and its substrate are discussed. The amount of the reporters displayed on the surface was unambiguously quantified via a unique strategy. Under optimal conditions with the overproduction of YhcS, 47,300 YhcR fusions could be displayed per cell. Displayed reporters were biologically functional and surface accessible. Characterization of the sortase-substrate system allowed the successful development of a YhcR-based covalent surface display system. This system may have various biotechnological applications.
Collapse
|
157
|
Tian BX, Eriksson LA. Catalytic mechanism and roles of Arg197 and Thr183 in the Staphylococcus aureus sortase A enzyme. J Phys Chem B 2011; 115:13003-11. [PMID: 21950672 DOI: 10.1021/jp2058113] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The sortase A enzyme, which catalyzes the peptidoglycan cell wall anchoring reaction of LPXTG surface proteins, has been proposed to be a universal target for therapeutic agents against Gram-positive bacteria. The catalytic mechanism of the Staphylococcus aureus sortase A enzyme has been systematically studied using molecular dynamics simulations, ONIOM(DFT:MM) calculations, and QM/MM charge deletion analysis. The catalytic roles of Arg197 and Thr183 were analyzed. Our calculations show that Arg197 has several important roles in the mechanism. It is crucial for substrate binding, and is capable of reversible shift of its hydrogen bonds between the LP and TG carbonyls of the LPXTG substrate motif, depending on the protonation state of the catalytic Cys184-His120 dyad. Arg197 stabilizes the catalytic dyad in the active ion pair form but at the same time raises the barrier to acylation by approximately 8 kcal/mol. Thr183 is also essential for the catalytic reaction in that it correspondingly lowers the barrier by the same amount via electrostatic interactions. The catalytic mechanism proceeds via proton transfer from His120, followed by nucleophilic attack from the thiolate anion of Cys184. The data thus supports the proposed reverse protonation mechanism, and disproves the hypothesis of the Arg197 generating an oxyanion hole to stabilize the tetrahedral intermediate of the reaction.
Collapse
Affiliation(s)
- Bo-Xue Tian
- School of Chemistry, National University of Ireland-Galway, Galway, Ireland
| | | |
Collapse
|
158
|
Bien J, Sokolova O, Bozko P. Characterization of Virulence Factors of Staphylococcus aureus: Novel Function of Known Virulence Factors That Are Implicated in Activation of Airway Epithelial Proinflammatory Response. J Pathog 2011; 2011:601905. [PMID: 22567334 PMCID: PMC3335658 DOI: 10.4061/2011/601905] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 06/23/2011] [Accepted: 07/15/2011] [Indexed: 12/04/2022] Open
Abstract
Airway epithelial cells play a major role in initiating inflammation in response to bacterial pathogens. S. aureus is an important pathogen associated with activation of diverse types of infection characterized by inflammation dominated by polymorphonuclear leukocytes. This bacterium frequently causes lung infection, which is attributed to virulence factors. Many of virulence determinants associated with S. aureus-mediated lung infection have been known for several years. In this paper, we discuss recent advances in our understanding of known virulence factors implicated in pneumonia. We anticipate that better understanding of novel functions of known virulence factors could open the way to regulate inflammatory reactions of the epithelium and to develop effective strategies to treat S. aureus-induced airway diseases.
Collapse
Affiliation(s)
- Justyna Bien
- Witold Stefanski Institute of Parasitology of the Polish Academy of Sciences, Twarda Street 51/55, 00-818 Warsaw, Poland
| | | | | |
Collapse
|
159
|
Signoretto C, Canepari P, Stauder M, Vezzulli L, Pruzzo C. Functional foods and strategies contrasting bacterial adhesion. Curr Opin Biotechnol 2011; 23:160-7. [PMID: 21906930 DOI: 10.1016/j.copbio.2011.08.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 08/18/2011] [Accepted: 08/22/2011] [Indexed: 11/15/2022]
Abstract
Antibacterial strategies targeting bacterial adhesion to substrates are considered a valuable alternative to traditional antibiotic therapy, in view of the great advantage they bring in combating the infectious process at the very early stage without selecting for drug resistant cells. Amongst bioactive compounds with activity against bacterial adhesion, several are found in natural food and beverages, such as cranberry, tea, coffee, wine and milk. For the analysis of their anti-infective potential, successful experimental models can be conducted using different substrates from the oral cavity. Studies conducted so far in this field allowed the discovery of a variety of anti-adhesive fractions and compounds proven to be effective against bacterial traits involved in the development of oral pathologies such as caries and gingivitis/periodontitis. Discovering new anti-adhesive compounds from natural products, unravelling and testing their prophylactic and therapeutic values, and improving their use in the general population are promising new frontiers in the global fight against human infectious diseases.
Collapse
Affiliation(s)
- Caterina Signoretto
- Dipartimento di Patologia e Diagnostica, Sezione di Microbiologia, Università di Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | | | | | | | | |
Collapse
|
160
|
Chen P, Leung KP. Identification of the srtC1 transcription start site and catalytically essential residues required for Actinomyces oris T14V SrtC1 activity. FEMS Microbiol Lett 2011; 322:115-22. [DOI: 10.1111/j.1574-6968.2011.02338.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
161
|
Analysis and application of Bacillus subtilis sortases to anchor recombinant proteins on the cell wall. AMB Express 2011; 1:22. [PMID: 21906378 PMCID: PMC3222306 DOI: 10.1186/2191-0855-1-22] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 07/21/2011] [Indexed: 02/06/2023] Open
Abstract
Bacillus subtilis codes for two putative sortases, YhcS and YwpE, and two surface proteins, YhcR and YfkN, harboring sorting motifs supposed to be recognized by the putative sortase(s). However, there is no experimental evidence to show a direct link between these sortases and sorting sequences. To study the role of these two putative sortases on displaying YhcR and YfkN on the cell wall, expression of yhcS and ywpE was analyzed by transcriptional fusions and by Northern blot. It turned out that yhcS gene is expressed at a higher level during the late stationary phase from both experiments, while ywpE expression is not confirmed in the Northern blot analysis. Next, we constructed yhcS and ywpE single and double knockout strains and plasmids that express one or both genes to restore the functions of the knockout strains. It could be shown that display of YhcR and YfkN on the surface depended on the presence of YhcS while YwpE seems not to play a major role if any as a sortase. Finally, the putative sorting motif together with a 123-amino-acid spacer derived from YhcR and YfkN designated YhcR123 and YfkN123, respectively, were fused to an α-amylase reporter enzyme. The fusion protein YhcR123-AmyQ could be displayed on the surface at high amounts, while YfkN123-AmyQ could be hardly detected. We conclude that the sortase YhcS can recognize and anchor YhcR on the cell wall. This result further indicates that the YhcR sorting sequence can be used to display recombinant proteins on the surface of B. subtilis cells.
Collapse
|
162
|
Popp MWL, Ploegh HL. Bilden und Brechen von Peptidbindungen: Protein-Engineering mithilfe von Sortase. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201008267] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
163
|
Popp MWL, Ploegh HL. Making and breaking peptide bonds: protein engineering using sortase. Angew Chem Int Ed Engl 2011; 50:5024-32. [PMID: 21538739 DOI: 10.1002/anie.201008267] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Indexed: 01/31/2023]
Abstract
Sortases are a class of bacterial enzymes that possess transpeptidase activity. It is their ability to site-specifically break a peptide bond and then reform a new bond with an incoming nucleophile that makes sortase an attractive tool for protein engineering. This technique has been adopted for a range of applications, from chemistry-based to cell biology and technology. In this Minireview we provide a brief overview of the biology of sortase enzymes and current applications in protein engineering. We identify areas that lend themselves to further innovation and that suggest new applications.
Collapse
|
164
|
Recent advances in understanding the antibacterial properties of flavonoids. Int J Antimicrob Agents 2011; 38:99-107. [PMID: 21514796 DOI: 10.1016/j.ijantimicag.2011.02.014] [Citation(s) in RCA: 665] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 01/14/2023]
Abstract
Antibiotic resistance is a major global problem and there is a pressing need to develop new therapeutic agents. Flavonoids are a family of plant-derived compounds with potentially exploitable activities, including direct antibacterial activity, synergism with antibiotics, and suppression of bacterial virulence. In this review, recent advances towards understanding these properties are described. Information is presented on the ten most potently antibacterial flavonoids as well as the five most synergistic flavonoid-antibiotic combinations tested in the last 6 years (identified from PubMed and ScienceDirect). Top of these respective lists are panduratin A, with minimum inhibitory concentrations (MICs) of 0.06-2.0 μg/mL against Staphylococcus aureus, and epicatechin gallate, which reduces oxacillin MICs as much as 512-fold. Research seeking to improve such activity and understand structure-activity relationships is discussed. Proposed mechanisms of action are also discussed. In addition to direct and synergistic activities, flavonoids inhibit a number of bacterial virulence factors, including quorum-sensing signal receptors, enzymes and toxins. Evidence of these molecular effects at the cellular level include in vitro inhibition of biofilm formation, inhibition of bacterial attachment to host ligands, and neutralisation of toxicity towards cultured human cells. In vivo evidence of disruption of bacterial pathogenesis includes demonstrated efficacy against Helicobacter pylori infection and S. aureus α-toxin intoxication.
Collapse
|
165
|
Oh I, Yang WY, Chung SC, Kim TY, Oh KB, Shin J. In vitro sortase A inhibitory and antimicrobial activity of flavonoids isolated from the roots of Sophora flavescens. Arch Pharm Res 2011; 34:217-22. [PMID: 21380804 DOI: 10.1007/s12272-011-0206-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 10/05/2010] [Accepted: 11/01/2010] [Indexed: 02/01/2023]
Abstract
A series of flavonoids (1-14) was isolated from the roots of Sophora flavescens. We evaluated their ability to inhibit both microbial growth and sortase A, an enzyme that plays a key role in cell wall protein anchoring and virulence in Staphylococcus aureus. Most prenylated flavonoids (7-13) displayed potent inhibitory activity against gram-positive and gram-negative bacteria except E. coli, with minimum inhibitory concentrations values ranging from 4.40 to 27.7 μM, and weak or no activity against fungal strains tested. Kurarinol (6) was a potent inhibitor of sortase A, with an IC(50) value of 107.7 ± 6.6 μM. A preliminary structure-activity relationship, including essential structural requirements, is described.
Collapse
Affiliation(s)
- Ikhoon Oh
- College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
166
|
Tian B, Eriksson LA. Structural changes of Listeria monocytogenes Sortase A: a key to understanding the catalytic mechanism. Proteins 2011; 79:1564-72. [PMID: 21374720 DOI: 10.1002/prot.22983] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 12/03/2010] [Accepted: 12/30/2010] [Indexed: 11/08/2022]
Abstract
Listeria monocytogenes is one of the most virulent foodborne pathogens. L. monocytogenes Sortase A (SrtA) enzyme, which catalyzes the cell wall anchoring reaction of the leucine, proline, X, threonine, and glycine proteins (LPXTG, where X is any amino acid), is a target for the development of antilisteriosis drugs. In this study, the structure of the L. monocytogenes SrtA enzyme-substrate complex was obtained using homology modeling, molecular docking and molecular dynamics simulations. Explicit enzyme-substrate interactions in the inactive and active forms of the enzyme were compared, based on 30 ns simulations on each system. The active site arginine (Arg 197) was found to be able change its hydrogen donor interactions from the LP backbone carbonyl groups of the LPXTG substrate in the inactive form, to the TG backbone carbonyls in the active form, which could be of importance for holding the substrate in position for the catalytic process.
Collapse
Affiliation(s)
- Boxue Tian
- School of Chemistry, National University of Ireland, Galway, Ireland
| | | |
Collapse
|
167
|
Catalytic Asymmetric Synthesis of γ-Substituted Vinyl Sulfones. Chemistry 2011; 17:2450-7. [DOI: 10.1002/chem.201003177] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Indexed: 01/13/2023]
|
168
|
Crystal structure of Spy0129, a Streptococcus pyogenes class B sortase involved in pilus assembly. PLoS One 2011; 6:e15969. [PMID: 21264317 PMCID: PMC3019223 DOI: 10.1371/journal.pone.0015969] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 12/07/2010] [Indexed: 01/31/2023] Open
Abstract
Sortase enzymes are cysteine transpeptidases that mediate the covalent attachment of substrate proteins to the cell walls of Gram-positive bacteria, and thereby play a crucial role in virulence, infection and colonisation by pathogens. Many cell-surface proteins are anchored by the housekeeping sortase SrtA but other more specialised sortases exist that attach sub-sets of proteins or function in pilus assembly. The sortase Spy0129, or SrtC1, from the M1 SF370 strain of Streptococcus pyogenes is responsible for generating the covalent linkages between the pilin subunits in the pili of this organism. The crystal structure of Spy0129 has been determined at 2.3 Å resolution (R = 20.4%, Rfree = 26.0%). The structure shows that Spy0129 is a class B sortase, in contrast to other characterised pilin polymerases, which belong to class C. Spy0129 lacks a flap believed to function in substrate recognition in class C enzymes and instead has an elaborated β6/β7 loop. The two independent Spy0129 molecules in the crystal show differences in the positions and orientations of the catalytic Cys and His residues, Cys221 and His126, correlated with movements of the β7/β8 and β4/β5 loops that respectively follow these residues. Bound zinc ions stabilise these alternative conformations in the crystal. This conformational variability is likely to be important for function although there is no evidence that zinc is involved in vivo.
Collapse
|
169
|
Nelson JW, Chamessian AG, McEnaney PJ, Murelli RP, Kazmiercak BI, Spiegel DA. A biosynthetic strategy for re-engineering the Staphylococcus aureus cell wall with non-native small molecules. ACS Chem Biol 2010; 5:1147-55. [PMID: 20923200 PMCID: PMC3003768 DOI: 10.1021/cb100195d] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterial pathogen that has emerged as a major public health threat. Here we report that the cell wall of S. aureus can be covalently re-engineered to contain non-native small molecules. This process makes use of endogenous levels of the bacterial enzyme sortase A (SrtA), which ordinarily functions to incorporate proteins into the bacterial cell wall. Thus, incubation of wild-type bacteria with rationally designed SrtA substrates results in covalent incorporation of functional molecular handles (fluorescein, biotin, and azide) into cell wall peptidoglycan. These conclusions are supported by data obtained through a variety of experimental techniques (epifluorescence and electron microscopy, biochemical extraction, and mass spectrometry), and cell-wall-incorporated azide was exploited as a chemical handle to perform an azide-alkyne cycloaddition reaction on the bacterial cell surface. This report represents the first example of cell wall engineering of S. aureus or any other pathogenic Gram-positive bacteria and has the potential for widespread utility.
Collapse
Affiliation(s)
- James W. Nelson
- Department of Chemistry, Yale University, New Haven, Connecticut 06520
| | | | | | - Ryan P. Murelli
- Department of Chemistry, Yale University, New Haven, Connecticut 06520
| | - Barbara I. Kazmiercak
- Department of Medicine (Infectious Diseases), Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06520
| | - David A. Spiegel
- Department of Chemistry, Yale University, New Haven, Connecticut 06520
| |
Collapse
|
170
|
Newly discovered and characterized antivirulence compounds inhibit bacterial mono-ADP-ribosyltransferase toxins. Antimicrob Agents Chemother 2010; 55:983-91. [PMID: 21135177 DOI: 10.1128/aac.01164-10] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mono-ADP-ribosyltransferase toxins are bacterial virulence factors that contribute to many disease states in plants, animals, and humans. These toxins function as enzymes that target various host proteins and covalently attach an ADP-ribose moiety that alters target protein function. We tested compounds from a virtual screen of commercially available compounds combined with a directed poly(ADP-ribose) polymerase (PARP) inhibitor library and found several compounds that bind tightly and inhibit toxins from Pseudomonas aeruginosa and Vibrio cholerae. The most efficacious compounds completely protected human lung epithelial cells against the cytotoxicity of these bacterial virulence factors. Moreover, we determined high-resolution crystal structures of the best inhibitors in complex with cholix toxin to reveal important criteria for inhibitor binding and mechanism of action. These results provide new insight into development of antivirulence compounds for treating many bacterial diseases.
Collapse
|
171
|
Letek M, González P, MacArthur I, Rodríguez H, Freeman TC, Valero-Rello A, Blanco M, Buckley T, Cherevach I, Fahey R, Hapeshi A, Holdstock J, Leadon D, Navas J, Ocampo A, Quail MA, Sanders M, Scortti MM, Prescott JF, Fogarty U, Meijer WG, Parkhill J, Bentley SD, Vázquez-Boland JA. The genome of a pathogenic rhodococcus: cooptive virulence underpinned by key gene acquisitions. PLoS Genet 2010; 6:e1001145. [PMID: 20941392 PMCID: PMC2947987 DOI: 10.1371/journal.pgen.1001145] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 08/31/2010] [Indexed: 11/29/2022] Open
Abstract
We report the genome of the facultative intracellular parasite Rhodococcus equi, the only animal pathogen within the biotechnologically important actinobacterial genus Rhodococcus. The 5.0-Mb R. equi 103S genome is significantly smaller than those of environmental rhodococci. This is due to genome expansion in nonpathogenic species, via a linear gain of paralogous genes and an accelerated genetic flux, rather than reductive evolution in R. equi. The 103S genome lacks the extensive catabolic and secondary metabolic complement of environmental rhodococci, and it displays unique adaptations for host colonization and competition in the short-chain fatty acid-rich intestine and manure of herbivores--two main R. equi reservoirs. Except for a few horizontally acquired (HGT) pathogenicity loci, including a cytoadhesive pilus determinant (rpl) and the virulence plasmid vap pathogenicity island (PAI) required for intramacrophage survival, most of the potential virulence-associated genes identified in R. equi are conserved in environmental rhodococci or have homologs in nonpathogenic Actinobacteria. This suggests a mechanism of virulence evolution based on the cooption of existing core actinobacterial traits, triggered by key host niche-adaptive HGT events. We tested this hypothesis by investigating R. equi virulence plasmid-chromosome crosstalk, by global transcription profiling and expression network analysis. Two chromosomal genes conserved in environmental rhodococci, encoding putative chorismate mutase and anthranilate synthase enzymes involved in aromatic amino acid biosynthesis, were strongly coregulated with vap PAI virulence genes and required for optimal proliferation in macrophages. The regulatory integration of chromosomal metabolic genes under the control of the HGT-acquired plasmid PAI is thus an important element in the cooptive virulence of R. equi.
Collapse
Affiliation(s)
- Michal Letek
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| | - Patricia González
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Irish Equine Centre, Johnstown, Naas, Ireland
| | - Iain MacArthur
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Irish Equine Centre, Johnstown, Naas, Ireland
- Department of Pathobiology, University of Guelph, Guelph, Canada
| | - Héctor Rodríguez
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Irish Equine Centre, Johnstown, Naas, Ireland
| | - Tom C. Freeman
- Division of Genetics and Genomics, Roslin BioCentre, University of Edinburgh, Edinburgh, United Kingdom
| | - Ana Valero-Rello
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Irish Equine Centre, Johnstown, Naas, Ireland
| | - Mónica Blanco
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Irish Equine Centre, Johnstown, Naas, Ireland
| | - Tom Buckley
- Irish Equine Centre, Johnstown, Naas, Ireland
| | - Inna Cherevach
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Ruth Fahey
- School of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Alexia Hapeshi
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| | - Jolyon Holdstock
- Oxford Gene Technology, Begbroke Science Park, Oxford, United Kingdom
| | | | - Jesús Navas
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | | | - Michael A. Quail
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Mandy Sanders
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Mariela M. Scortti
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Departamento de Bioquímica y Biología Molecular IV, Universidad Complutense, Madrid, Spain
| | - John F. Prescott
- Department of Pathobiology, University of Guelph, Guelph, Canada
| | | | - Wim G. Meijer
- School of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Julian Parkhill
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Stephen D. Bentley
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - José A. Vázquez-Boland
- Microbial Pathogenesis Unit, Centres for Infectious Diseases and Immunity, Infection, and Evolution, University of Edinburgh, Edinburgh, United Kingdom
- Grupo de Patogenómica Bacteriana, Universidad de León, León, Spain
| |
Collapse
|
172
|
|
173
|
Weiner EM, Robson S, Marohn M, Clubb RT. The Sortase A enzyme that attaches proteins to the cell wall of Bacillus anthracis contains an unusual active site architecture. J Biol Chem 2010; 285:23433-43. [PMID: 20489200 PMCID: PMC2906334 DOI: 10.1074/jbc.m110.135434] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 05/14/2010] [Indexed: 11/06/2022] Open
Abstract
The pathogen Bacillus anthracis uses the Sortase A (SrtA) enzyme to anchor proteins to its cell wall envelope during vegetative growth. To gain insight into the mechanism of protein attachment to the cell wall in B. anthracis we investigated the structure, backbone dynamics, and function of SrtA. The NMR structure of SrtA has been determined with a backbone coordinate precision of 0.40 +/- 0.07 A. SrtA possesses several novel features not previously observed in sortase enzymes including the presence of a structurally ordered amino terminus positioned within the active site and in contact with catalytically essential histidine residue (His(126)). We propose that this appendage, in combination with a unique flexible active site loop, mediates the recognition of lipid II, the second substrate to which proteins are attached during the anchoring reaction. pK(a) measurements indicate that His(126) is uncharged at physiological pH compatible with the enzyme operating through a "reverse protonation" mechanism. Interestingly, NMR relaxation measurements and the results of a model building study suggest that SrtA recognizes the LPXTG sorting signal through a lock-in-key mechanism in contrast to the prototypical SrtA enzyme from Staphylococcus aureus.
Collapse
Affiliation(s)
| | - Scott Robson
- From the
Department of Chemistry and Biochemistry
| | | | - Robert T. Clubb
- From the
Department of Chemistry and Biochemistry
- UCLA-DOE Institute of Genomics and Proteomics, and
- Molecular Biology Institute, University of California, Los Angeles, California 90095-1570
| |
Collapse
|
174
|
Oh KB, Nam KW, Ahn H, Shin J, Kim S, Mar W. Therapeutic effect of (Z)-3-(2,5-dimethoxyphenyl)-2-(4-methoxyphenyl) acrylonitrile (DMMA) against Staphylococcus aureus infection in a murine model. Biochem Biophys Res Commun 2010; 396:440-4. [PMID: 20433810 DOI: 10.1016/j.bbrc.2010.04.113] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 04/20/2010] [Indexed: 02/02/2023]
|
175
|
A Bacillus anthracis S-layer homology protein that binds heme and mediates heme delivery to IsdC. J Bacteriol 2010; 192:3503-11. [PMID: 20435727 DOI: 10.1128/jb.00054-10] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The sequestration of iron by mammalian hosts represents a significant obstacle to the establishment of a bacterial infection. In response, pathogenic bacteria have evolved mechanisms to acquire iron from host heme. Bacillus anthracis, the causative agent of anthrax, utilizes secreted hemophores to scavenge heme from host hemoglobin, thereby facilitating iron acquisition from extracellular heme pools and delivery to iron-regulated surface determinant (Isd) proteins covalently attached to the cell wall. However, several Gram-positive pathogens, including B. anthracis, contain genes that encode near iron transporter (NEAT) proteins that are genomically distant from the genetically linked Isd locus. NEAT domains are protein modules that partake in several functions related to heme transport, including binding heme and hemoglobin. This finding raises interesting questions concerning the relative role of these NEAT proteins, relative to hemophores and the Isd system, in iron uptake. Here, we present evidence that a B. anthracis S-layer homology (SLH) protein harboring a NEAT domain binds and directionally transfers heme to the Isd system via the cell wall protein IsdC. This finding suggests that the Isd system can receive heme from multiple inputs and may reflect an adaptation of B. anthracis to changing iron reservoirs during an infection. Understanding the mechanism of heme uptake in pathogenic bacteria is important for the development of novel therapeutics to prevent and treat bacterial infections.
Collapse
|
176
|
Kline KA, Dodson KW, Caparon MG, Hultgren SJ. A tale of two pili: assembly and function of pili in bacteria. Trends Microbiol 2010; 18:224-32. [PMID: 20378353 DOI: 10.1016/j.tim.2010.03.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 11/25/2009] [Accepted: 03/05/2010] [Indexed: 10/19/2022]
Abstract
Bacterial pili have long been recognized as mediators of initial host-pathogen interactions important for the progression of Gram-negative bacterial diseases. An appreciation of the role of pili on virulence in Gram-positive bacteria and the unique properties of their biogenesis is a rapidly emerging area of research. In this review, we focus on recent advances in one of the longest-studied Gram-negative pilus systems, the chaperone/usher assembled pili, along with the newcomer to the field, the sortase-assembled pili of Gram-positive bacteria. In both systems, a wealth of new structural and molecular details has emerged recently. In light of this, we explore similarities between chaperone/usher and sortase-assembled pilus biogenesis and highlight paradigms unique to each, with the goal of using knowledge of each system to raise new questions and inform future studies of the other.
Collapse
Affiliation(s)
- Kimberly A Kline
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
177
|
Solis N, Larsen MR, Cordwell SJ. Improved accuracy of cell surface shaving proteomics in Staphylococcus aureus
using a false-positive control. Proteomics 2010; 10:2037-49. [DOI: 10.1002/pmic.200900564] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
178
|
El Mortaji L, Terrasse R, Dessen A, Vernet T, Di Guilmi AM. Stability and assembly of pilus subunits of Streptococcus pneumoniae. J Biol Chem 2010; 285:12405-15. [PMID: 20147289 DOI: 10.1074/jbc.m109.082776] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pili are surface-exposed virulence factors involved in bacterial adhesion to host cells. The Streptococcus pneumoniae pilus is composed of three structural proteins, RrgA, RrgB, and RrgC and three transpeptidase enzymes, sortases SrtC-1, SrtC-2, and SrtC-3. To gain insights into the mechanism of pilus formation we have exploited biochemical approaches using recombinant proteins expressed in Escherichia coli. Using site-directed mutagenesis, mass spectrometry, limited proteolysis, and thermal stability measurements, we have identified isopeptide bonds in RrgB and RrgC and demonstrate their role in protein stabilization. Co-expression in E. coli of RrgB together with RrgC and SrtC-1 leads to the formation of a covalent RrgB-RrgC complex. Inactivation of SrtC-1 by mutation of the active site cysteine impairs RrgB-RrgC complex formation, indicating that the association between RrgB and RrgC is specifically catalyzed by SrtC-1. Mass spectrometry analyses performed on purified samples of the RrgB-RrgC complex show that the complex has 1:1 stoichiometry. The deletion of the IPQTG RrgB sorting signal, but not the corresponding sequence in RrgC, abolishes complex formation, indicating that SrtC-1 recognizes exclusively the sorting motif of RrgB. Finally, we show that the intramolecular bonds that stabilize RrgB may play a role in its efficient recognition by SrtC-1. The development of a methodology to generate covalent pilin complexes in vitro, facilitating the study of sortase specificity and the importance of isopeptide bond formation for pilus biogenesis, provide key information toward the understanding of this complex macromolecular process.
Collapse
Affiliation(s)
- Lamya El Mortaji
- Laboratoire d'Ingénierie des Macromolécules, Institut de Biologie Structurale Jean-Pierre Ebel, UMR 5075 (CEA, CNRS, UJF), Grenoble, France
| | | | | | | | | |
Collapse
|
179
|
Leahy SC, Kelly WJ, Altermann E, Ronimus RS, Yeoman CJ, Pacheco DM, Li D, Kong Z, McTavish S, Sang C, Lambie SC, Janssen PH, Dey D, Attwood GT. The genome sequence of the rumen methanogen Methanobrevibacter ruminantium reveals new possibilities for controlling ruminant methane emissions. PLoS One 2010; 5:e8926. [PMID: 20126622 PMCID: PMC2812497 DOI: 10.1371/journal.pone.0008926] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 12/07/2009] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Methane (CH(4)) is a potent greenhouse gas (GHG), having a global warming potential 21 times that of carbon dioxide (CO(2)). Methane emissions from agriculture represent around 40% of the emissions produced by human-related activities, the single largest source being enteric fermentation, mainly in ruminant livestock. Technologies to reduce these emissions are lacking. Ruminant methane is formed by the action of methanogenic archaea typified by Methanobrevibacter ruminantium, which is present in ruminants fed a wide variety of diets worldwide. To gain more insight into the lifestyle of a rumen methanogen, and to identify genes and proteins that can be targeted to reduce methane production, we have sequenced the 2.93 Mb genome of M. ruminantium M1, the first rumen methanogen genome to be completed. METHODOLOGY/PRINCIPAL FINDINGS The M1 genome was sequenced, annotated and subjected to comparative genomic and metabolic pathway analyses. Conserved and methanogen-specific gene sets suitable as targets for vaccine development or chemogenomic-based inhibition of rumen methanogens were identified. The feasibility of using a synthetic peptide-directed vaccinology approach to target epitopes of methanogen surface proteins was demonstrated. A prophage genome was described and its lytic enzyme, endoisopeptidase PeiR, was shown to lyse M1 cells in pure culture. A predicted stimulation of M1 growth by alcohols was demonstrated and microarray analyses indicated up-regulation of methanogenesis genes during co-culture with a hydrogen (H(2)) producing rumen bacterium. We also report the discovery of non-ribosomal peptide synthetases in M. ruminantium M1, the first reported in archaeal species. CONCLUSIONS/SIGNIFICANCE The M1 genome sequence provides new insights into the lifestyle and cellular processes of this important rumen methanogen. It also defines vaccine and chemogenomic targets for broad inhibition of rumen methanogens and represents a significant contribution to worldwide efforts to mitigate ruminant methane emissions and reduce production of anthropogenic greenhouse gases.
Collapse
Affiliation(s)
- Sinead C. Leahy
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - William J. Kelly
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Eric Altermann
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Ron S. Ronimus
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Carl J. Yeoman
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Diana M. Pacheco
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Dong Li
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Zhanhao Kong
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Sharla McTavish
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Carrie Sang
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Suzanne C. Lambie
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Peter H. Janssen
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Debjit Dey
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Graeme T. Attwood
- Rumen Microbial Genomics, Food Metabolism and Microbiology Section, Food and Textiles Group, AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| |
Collapse
|
180
|
Clancy KW, Melvin JA, McCafferty DG. Sortase transpeptidases: insights into mechanism, substrate specificity, and inhibition. Biopolymers 2010; 94:385-96. [PMID: 20593474 PMCID: PMC4648256 DOI: 10.1002/bip.21472] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gram-positive bacteria pose a serious healthcare threat. The growing antibiotic resistance epidemic creates a dire need for new antibiotic targets. The sortase family of enzymes is a promising target for antimicrobial therapy. This review covers the current knowledge of the mechanism, substrate specificity, and inhibitory studies of the Gram-positive bacterial [corrected] enzyme sortase.
Collapse
Affiliation(s)
| | | | - Dewey G. McCafferty
- Department of Chemistry, Duke University, Durham, NC
- Department of Biochemistry, Duke University, Durham, NC
| |
Collapse
|
181
|
Speziale P, Pietrocola G, Rindi S, Provenzano M, Provenza G, Di Poto A, Visai L, Arciola CR. Structural and functional role of Staphylococcus aureus surface components recognizing adhesive matrix molecules of the host. Future Microbiol 2009; 4:1337-52. [DOI: 10.2217/fmb.09.102] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a versatile and harmful human pathogen in both hospital- and community-acquired infections. S. aureus can initiate host infection by adhering to components of the extracellular matrix. Adherence is mediated by a variety of protein adhesins of the microbial surface component recognizing adhesive matrix molecule (MSCRAMM) family. In this article, we describe these MSCRAMMs in terms of structural organization and ligand-binding capacity and discuss their role as a possible target for immunotherapy.
Collapse
Affiliation(s)
- Pietro Speziale
- Department of Biochemistry, Viale Taramelli 3/b 27100 Pavia, Italy
| | | | - Simonetta Rindi
- Department of Biochemistry, Viale Taramelli 3/b 27100 Pavia, Italy
| | - Maria Provenzano
- Department of Biochemistry, Viale Taramelli 3/b 27100 Pavia, Italy
| | - Giulio Provenza
- Department of Biochemistry, Viale Taramelli 3/b 27100 Pavia, Italy
| | | | - Livia Visai
- Department of Biochemistry, Viale Taramelli 3/b 27100 Pavia, Italy and Center for Tissue Engineering (CIT), Via Ferrata 1, 27100 Pavia, Italy
| | - Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy and Experimental Pathology Department, University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy
| |
Collapse
|
182
|
Fabian M, Solomaha E, Olson JS, Maresso AW. Heme transfer to the bacterial cell envelope occurs via a secreted hemophore in the Gram-positive pathogen Bacillus anthracis. J Biol Chem 2009; 284:32138-46. [PMID: 19759022 PMCID: PMC2797284 DOI: 10.1074/jbc.m109.040915] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 08/28/2009] [Indexed: 11/06/2022] Open
Abstract
To initiate and sustain an infection in mammals, bacterial pathogens must acquire host iron. However, the host's compartmentalization of large amounts of iron in heme, which is bound primarily by hemoglobin in red blood cells, acts as a barrier to bacterial iron assimilation. Bacillus anthracis, the causative agent of the disease anthrax, secretes two NEAT (near iron transporter) proteins, IsdX1 and IsdX2, which scavenge heme from host hemoglobin and promote growth under low iron conditions. The mechanism of heme transfer from these hemophores to the bacterial cell is not known. We present evidence that the heme-bound form of IsdX1 rapidly and directionally transfers heme to IsdC, a NEAT protein covalently attached to the cell wall, as well as to IsdX2. In both cases, the transfer of heme is mediated by a physical association between the donor and recipient. Unlike Staphylococcus aureus, whose NEAT proteins acquire heme from hemoglobin directly at the bacterial surface, B. anthracis secretes IsdX1 to capture heme in the extracellular milieu and relies on NEAT-NEAT interactions to deliver the bound heme to the envelope via IsdC. Understanding the mechanism of NEAT-mediated iron transport into pathogenic Gram-positive bacteria may provide an avenue for the development of therapeutics to combat infection.
Collapse
Affiliation(s)
- Marian Fabian
- From the Department of Biochemistry & Cell Biology, Rice University, Houston, Texas 77096
| | - Elena Solomaha
- the Biophysics Core Facility, The University of Chicago, Chicago, Illinois 60637, and
| | - John S. Olson
- From the Department of Biochemistry & Cell Biology, Rice University, Houston, Texas 77096
| | - Anthony W. Maresso
- the Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
183
|
Interaction of sortase A and lipase 2 in the inhibition of Staphylococcus aureus biofilm formation. Arch Microbiol 2009; 191:879-84. [PMID: 19838678 DOI: 10.1007/s00203-009-0520-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 09/28/2009] [Accepted: 10/05/2009] [Indexed: 10/20/2022]
Abstract
Recombinant sortase A (SrtA) was used to immune rabbit, and the inhibitory activity of anti-SrtA serum on Staphylococcus aureus biofilm formation was tested. Biofilm formation was inhibited by anti-SrtA rabbit serum in S. aureus ATCC25923 and two clinical isolated strains. The antiserum was separated into two fractions, and the main component with the inhibitory activity was demonstrated to be the IgG fraction. Two proteins interact with the IgG fraction were identified by using an in vitro pull-down assay and were confirmed to be lipase 2 and gamma-hemolysin by mass spectrometry. Cross-interaction between SrtA and lipase 2 was further confirmed by Western blotting. Addition of anti-lipase 2 serum in the culture medium also showed inhibitory effect against biofilm formation. Together, our study suggests anti-SrtA serum inhibits S. aureus biofilm formation and lipase 2 is one of the targets of anti-SrtA serum in this inhibition process. This is the first study to demonstrate the roles of antisera against SrtA and lipase 2 in the inhibition of biofilm formation in S. aureus.
Collapse
|
184
|
Suree N, Yi SW, Thieu W, Marohn M, Damoiseaux R, Chan A, Jung ME, Clubb RT. Discovery and structure-activity relationship analysis of Staphylococcus aureus sortase A inhibitors. Bioorg Med Chem 2009; 17:7174-85. [PMID: 19781950 PMCID: PMC2888031 DOI: 10.1016/j.bmc.2009.08.067] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 08/22/2009] [Accepted: 08/29/2009] [Indexed: 01/29/2023]
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) is a major health problem that has created a pressing need for new antibiotics. Compounds that inhibit the S. aureus SrtA sortase may function as potent anti-infective agents as this enzyme attaches virulence factors to the cell wall. Using high-throughput screening, we have identified several compounds that inhibit the enzymatic activity of the SrtA. A structure-activity relationship (SAR) analysis led to the identification of several pyridazinone and pyrazolethione analogs that inhibit SrtA with IC(50) values in the sub-micromolar range. Many of these molecules also inhibit the sortase enzyme from Bacillus anthracis suggesting that they may be generalized sortase inhibitors.
Collapse
Affiliation(s)
- Nuttee Suree
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
- Molecular Biology Institute, University of California, Los Angeles, CA 90095-1570, United States
| | - Sung Wook Yi
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
| | - William Thieu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
| | - Melanie Marohn
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
| | - Robert Damoiseaux
- Molecular Screening Shared Resource, University of California, Los Angeles, CA 90095-1570, United States
| | - Albert Chan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
- Molecular Biology Institute, University of California, Los Angeles, CA 90095-1570, United States
| | - Michael E. Jung
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
| | - Robert T. Clubb
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1570, United States
- Molecular Biology Institute, University of California, Los Angeles, CA 90095-1570, United States
- UCLA-Department of Energy Institute for Genomics and Proteomics, University of California, Los Angeles, CA 90095-1570, United States
| |
Collapse
|
185
|
Barczak AK, Hung DT. Productive steps toward an antimicrobial targeting virulence. Curr Opin Microbiol 2009; 12:490-6. [PMID: 19631578 PMCID: PMC2763366 DOI: 10.1016/j.mib.2009.06.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 06/05/2009] [Indexed: 01/19/2023]
Abstract
Targeting virulence factors has gained increasing attention as a potential approach to new antibiotics. Small molecule inhibitors of virulence have been shown to change the course of disease in whole organism infection models. Recently, key advances in the field include the identification of novel targets within cell signaling pathways, a new class of anti-virulence compounds that target bacterial defenses against host immunity, and a growing body of in vivo data to support the general approach of anti-virulence therapies. Additionally, there has been a distinct trend toward developing broader spectrum anti-virulence compounds, in particular agents with activity against diverse Gram-negative organisms. Herein we provide an update on the status of the field with a focus on recent advancements.
Collapse
Affiliation(s)
- Amy K Barczak
- Division of Infectious Diseases, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114, USA
| | | |
Collapse
|
186
|
Suree N, Liew CK, Villareal VA, Thieu W, Fadeev EA, Clemens JJ, Jung ME, Clubb RT. The structure of the Staphylococcus aureus sortase-substrate complex reveals how the universally conserved LPXTG sorting signal is recognized. J Biol Chem 2009; 284:24465-77. [PMID: 19592495 PMCID: PMC2782039 DOI: 10.1074/jbc.m109.022624] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 06/16/2009] [Indexed: 02/01/2023] Open
Abstract
In Gram-positive bacteria, sortase enzymes assemble surface proteins and pili in the cell wall envelope. Sortases catalyze a transpeptidation reaction that joins a highly conserved LPXTG sorting signal within their polypeptide substrate to the cell wall or to other pilin subunits. The molecular basis of transpeptidation and sorting signal recognition are not well understood, because the intermediates of catalysis are short lived. We have overcome this problem by synthesizing an analog of the LPXTG signal whose stable covalent complex with the enzyme mimics a key thioacyl catalytic intermediate. Here we report the solution structure and dynamics of its covalent complex with the Staphylococcus aureus SrtA sortase. In marked contrast to a previously reported crystal structure, we show that SrtA adaptively recognizes the LPXTG sorting signal by closing and immobilizing an active site loop. We have also used chemical shift mapping experiments to localize the binding site for the triglycine portion of lipid II, the second substrate to which surface proteins are attached. We propose a unified model of the transpeptidation reaction that explains the functions of key active site residues. Since the sortase-catalyzed anchoring reaction is required for the virulence of a number of bacterial pathogens, the results presented here may facilitate the development of new anti-infective agents.
Collapse
Affiliation(s)
- Nuttee Suree
- From the Department of Chemistry and Biochemistry and
- the UCLA-Department of Energy Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095-1570
| | - Chu Kong Liew
- From the Department of Chemistry and Biochemistry and
| | - Valerie A. Villareal
- From the Department of Chemistry and Biochemistry and
- the UCLA-Department of Energy Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095-1570
| | - William Thieu
- From the Department of Chemistry and Biochemistry and
- the UCLA-Department of Energy Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095-1570
| | - Evgeny A. Fadeev
- From the Department of Chemistry and Biochemistry and
- the UCLA-Department of Energy Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095-1570
| | | | | | - Robert T. Clubb
- From the Department of Chemistry and Biochemistry and
- the UCLA-Department of Energy Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095-1570
| |
Collapse
|
187
|
Race PR, Bentley ML, Melvin JA, Crow A, Hughes RK, Smith WD, Sessions RB, Kehoe MA, McCafferty DG, Banfield MJ. Crystal structure of Streptococcus pyogenes sortase A: implications for sortase mechanism. J Biol Chem 2009; 284:6924-33. [PMID: 19129180 PMCID: PMC2652338 DOI: 10.1074/jbc.m805406200] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 12/05/2008] [Indexed: 01/29/2023] Open
Abstract
Sortases are a family of Gram-positive bacterial transpeptidases that anchor secreted proteins to bacterial cell surfaces. These include many proteins that play critical roles in the virulence of Gram-positive bacterial pathogens such that sortases are attractive targets for development of novel antimicrobial agents. All Gram-positive pathogens express a "housekeeping" sortase that recognizes the majority of secreted proteins containing an LPXTG wall-sorting motif and covalently attaches these to bacterial cell wall peptidoglycan. Many Gram-positive pathogens also express additional sortases that link a small number of proteins, often with variant wall-sorting motifs, to either other surface proteins or peptidoglycan. To better understand the mechanisms of catalysis and substrate recognition by the housekeeping sortase produced by the important human pathogen Streptococcus pyogenes, the crystal structure of this protein has been solved and its transpeptidase activity established in vitro. The structure reveals a novel arrangement of key catalytic residues in the active site of a sortase, the first that is consistent with kinetic analysis. The structure also provides a complete description of residue positions surrounding the active site, overcoming the limitation of localized disorder in previous structures of sortase A-type proteins. Modification of the active site Cys through oxidation to its sulfenic acid form or by an alkylating reagent supports a role for a reactive thiol/thiolate in the catalytic mechanism. These new insights into sortase structure and function could have important consequences for inhibitor design.
Collapse
Affiliation(s)
- Paul R Race
- Institute for Cell and Molecular Biosciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Law B, Tung CH. Proteolysis: A Biological Process Adapted in Drug Delivery, Therapy, and Imaging. Bioconjug Chem 2009; 20:1683-95. [DOI: 10.1021/bc800500a] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Benedict Law
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, and The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, Texas 77030
| | - Ching-Hsuan Tung
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, and The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, Texas 77030
| |
Collapse
|
189
|
Liu MH, Otsuka N, Noyori K, Shiota S, Ogawa W, Kuroda T, Hatano T, Tsuchiya T. Synergistic Effect of Kaempferol Glycosides Purified from Laurus nobilis and Fluoroquinolones on Methicillin-Resistant Staphylococcus aureus. Biol Pharm Bull 2009; 32:489-92. [DOI: 10.1248/bpb.32.489] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Mei-Hua Liu
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Nao Otsuka
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Kumiko Noyori
- Laboratory of Pathogenic Microbiology, School of Pharmacy, Shujitsu University
| | - Sumiko Shiota
- Laboratory of Pathogenic Microbiology, School of Pharmacy, Shujitsu University
| | - Wakano Ogawa
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Teruo Kuroda
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Tsutomu Hatano
- Department of Pharmacognosy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Tomofusa Tsuchiya
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| |
Collapse
|
190
|
Abstract
Many of the greatest challenges in medicine and public health involve the evolution of drug resistance by pathogens. Recent advances in the theory of natural selection suggest that there are two broad classes of pathogen traits that can be targeted by drugs or vaccines. The first class, consisting of traits that benefit the individual organisms bearing them, causes a strong evolutionary response and the rapid emergence of drug resistance. The second class, consisting of traits that benefit groups of pathogen organisms including the individual provider, causes a weaker evolutionary response and less drug resistance. Although most previous drug development has targeted the first class, it would be advantageous to focus on the second class as targets for drug and vaccine development. Specific examples and test cases are discussed.
Collapse
Affiliation(s)
- John W Pepper
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, Arizona 85721, USA.
| |
Collapse
|
191
|
Antivirulence as a new antibacterial approach for chemotherapy. Curr Opin Chem Biol 2008; 12:400-8. [DOI: 10.1016/j.cbpa.2008.06.022] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 06/19/2008] [Accepted: 06/20/2008] [Indexed: 12/11/2022]
|