101
|
Tran HNT, Tran P, Deuis JR, Agwa AJ, Zhang AH, Vetter I, Schroeder CI. Enzymatic Ligation of a Pore Blocker Toxin and a Gating Modifier Toxin: Creating Double-Knotted Peptides with Improved Sodium Channel NaV1.7 Inhibition. Bioconjug Chem 2019; 31:64-73. [DOI: 10.1021/acs.bioconjchem.9b00744] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hue N. T. Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alan H. Zhang
- Center for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
102
|
Trachsel L, Johnbosco C, Lang T, Benetti EM, Zenobi-Wong M. Double-Network Hydrogels Including Enzymatically Crosslinked Poly-(2-alkyl-2-oxazoline)s for 3D Bioprinting of Cartilage-Engineering Constructs. Biomacromolecules 2019; 20:4502-4511. [DOI: 10.1021/acs.biomac.9b01266] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Lucca Trachsel
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Castro Johnbosco
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Thamar Lang
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Edmondo M. Benetti
- Laboratory for Surface Science and Technology, Department of Materials, ETH Zürich, CH-8093 Zürich, Switzerland
- Biointerfaces, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014 St. Gallen, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, CH-8093 Zürich, Switzerland
| |
Collapse
|
103
|
Abstract
Subtiligase-catalyzed peptide ligation is a powerful approach for site-specific protein bioconjugation, synthesis and semisynthesis of proteins and peptides, and chemoproteomic analysis of cellular N termini. Here, we provide a comprehensive review of the subtiligase technology, including its development, applications, and impacts on protein science. We highlight key advantages and limitations of the tool and compare it to other peptide ligase enzymes. Finally, we provide a perspective on future applications and challenges and how they may be addressed.
Collapse
Affiliation(s)
- Amy M Weeks
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94143, United States
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94143, United States.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
104
|
Hong H, Zhou Z, Zhou K, Liu S, Guo Z, Wu Z. Site-specific C-terminal dinitrophenylation to reconstitute the antibody Fc functions for nanobodies. Chem Sci 2019; 10:9331-9338. [PMID: 32110296 PMCID: PMC7006623 DOI: 10.1039/c9sc03840j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
Nanobodies are a class of camelid-derived single-domain antibodies that have many potential advantages over conventional antibodies and have been utilized to develop new therapeutic strategies for cancer and other diseases. However, nanobodies lack the Fc region of a conventional antibody, which possesses many functions, e.g., eliciting antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), essential for effective immunotherapy. The small molecular size of nanobodies also leads to poor pharmacokinetics, such as short in vivo half-life. To address these deficiencies, an endogenous antibody-based strategy to reconstitute the Fc functions for nanobodies was developed. As a proof-of-principle, an anti-human EGFR nanobody, 7D12, was selected to conduct C-terminal modification with the dinitrophenyl (DNP) hapten through Sortase A-mediated site-specific ligation. It was expected that the DNP motif would recruit endogenous human anti-DNP antibodies to indirectly reinstate the Fc functions. The resultant nanobody-DNP conjugates were shown to exhibit specific and high affinity binding to human EGFR expressed on target cancer cells. It was further proved that in the presence of anti-DNP antibody, these conjugates could mediate potent ADCC and CDC in vitro and exhibit significantly elongated half-life in vivo. Ultimately, it was proven in severe combined immunodeficiency (SCID) mice that treatment with the nanobody 7D12-DNP conjugate and anti-DNP mouse serum could inhibit xenograft tumor growth efficiently. In view of the abundance of anti-DNP and other endogenous antibodies in the human blood system, this could be a generally applicable approach employed to reconstitute the Fc functions for nanobodies and develop nanobody-based cancer immunotherapy and other therapies.
Collapse
Affiliation(s)
- Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology , Ministry of Education , School of Biotechnology , Jiangnan University , Wuxi , 214122 , China .
| | - Zhifang Zhou
- Key Laboratory of Carbohydrate Chemistry & Biotechnology , Ministry of Education , School of Biotechnology , Jiangnan University , Wuxi , 214122 , China .
| | - Kun Zhou
- Key Laboratory of Carbohydrate Chemistry & Biotechnology , Ministry of Education , School of Biotechnology , Jiangnan University , Wuxi , 214122 , China .
| | - Shaozhong Liu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology , Ministry of Education , School of Biotechnology , Jiangnan University , Wuxi , 214122 , China .
| | - Zhongwu Guo
- Department of Chemistry , University of Florida , 214 Leigh Hall , Gainesville , Florida 32611 , USA .
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology , Ministry of Education , School of Biotechnology , Jiangnan University , Wuxi , 214122 , China .
| |
Collapse
|
105
|
Jing X, Jin K. A gold mine for drug discovery: Strategies to develop cyclic peptides into therapies. Med Res Rev 2019; 40:753-810. [PMID: 31599007 DOI: 10.1002/med.21639] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/05/2019] [Accepted: 09/26/2019] [Indexed: 12/19/2022]
Abstract
As a versatile therapeutic modality, peptides attract much attention because of their great binding affinity, low toxicity, and the capability of targeting traditionally "undruggable" protein surfaces. However, the deficiency of cell permeability and metabolic stability always limits the success of in vitro bioactive peptides as drug candidates. Peptide macrocyclization is one of the most established strategies to overcome these limitations. Over the past decades, more than 40 cyclic peptide drugs have been clinically approved, the vast majority of which are derived from natural products. The de novo discovered cyclic peptides on the basis of rational design and in vitro evolution, have also enabled the binding with targets for which nature provides no solutions. The current review summarizes different classes of cyclic peptides with diverse biological activities, and presents an overview of various approaches to develop cyclic peptide-based drug candidates, drawing upon series of examples to illustrate each strategy.
Collapse
Affiliation(s)
- Xiaoshu Jing
- Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Kang Jin
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Jinan, Shandong, China
| |
Collapse
|
106
|
Structural studies of Staphylococcus aureus Sortase inhibiton via Conus venom peptides. Arch Biochem Biophys 2019; 671:87-102. [DOI: 10.1016/j.abb.2019.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
|
107
|
Schneewind O, Missiakas DM. Staphylococcal Protein Secretion and Envelope Assembly. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0070-2019. [PMID: 31267890 PMCID: PMC7028390 DOI: 10.1128/microbiolspec.gpp3-0070-2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
The highly cross-linked peptidoglycan represents the rigid layer of the bacterial envelope and protects bacteria from osmotic lysis. In Gram-positive bacteria, peptidoglycan also functions as a scaffold for the immobilization of capsular polysaccharide, wall teichoic acid (WTA), and surface proteins. This chapter captures recent development on the assembly of the envelope of Staphylococcus aureus including mechanisms accounting for immobilization of molecules to peptidoglycan as well as hydrolysis of peptidoglycan for the specific release of bound molecules, facilitation of protein secretion across the envelope and cell division. Peptidoglycan, WTA and capsular polysaccharide are directly synthesized onto undecaprenol. Surface proteins are anchored by Sortase A, a membrane-embedded transpeptidase that scans secreted polypeptides for the C-terminal LPXTG motif of sorting signals. The resulting acyl enzyme intermediate is resolved by lipid II, the undecaprenol-bound peptidoglycan precursor. While these pathways share membrane diffusible undecaprenol, assembly of these molecules occurs either at the cross-walls or the cell poles. In S. aureus, the cross-wall represents the site of de novo peptidoglycan synthesis which is eventually split to complete the cell cycle yielding newly divided daughter cells. Peptidoglycan synthesized at the cross-wall is initially devoid of WTA. Conversely, lipoteichoic acid (LTA) synthesis which does not require bactoprenol is seemingly restricted to septal membranes. Similarly, S. aureus distinguishes two types of surface protein precursors. Polypeptides with canonical signal peptides are deposited at the cell poles, whereas precursors with conserved YSIRK-GXXS motif signal peptides traffic to the cross-wall. A model for protein trafficking in the envelope and uneven distribution of teichoic acids is discussed.
Collapse
Affiliation(s)
- Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, IL 60637
| | | |
Collapse
|
108
|
Fischetti VA. Surface Proteins on Gram-Positive Bacteria. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0012-2018. [PMID: 31373270 PMCID: PMC6684298 DOI: 10.1128/microbiolspec.gpp3-0012-2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Indexed: 12/14/2022] Open
Abstract
Surface proteins are critical for the survival of gram-positive bacteria both in the environment and to establish an infection. Depending on the organism, their surface proteins are evolutionarily tailored to interact with specific ligands on their target surface, be it inanimate or animate. Most surface molecules on these organisms are covalently anchored to the peptidoglycan through an LPxTG motif found at the C-terminus. These surface molecules are generally modular with multiple binding or enzymatic domains designed for a specific survival function. For example, some molecules will bind serum proteins like fibronectin or fibrinogen in one domain and have a separate function in another domain. In addition, enzymes such as those responsible for the production of ATP may be generally found on some bacterial surfaces, but when or how they are used in the life of these bacteria is currently unknown. While surface proteins are required for pathogenicity but not viability, targeting the expression of these molecules on the bacterial surface would prevent infection but not death of the organism. Given that the number of different surface proteins could be in the range of two to three dozen, each with two or three separate functional domains (with hundreds to thousands of each protein on a given organism), exemplifies the complexity that exists on the bacterial surface. Because of their number, we could not adequately describe the characteristics of all surface proteins in this chapter. However, since the streptococcal M protein was one of the first gram-positive surface protein to be completely sequenced, and perhaps one of the best studied, we will use M protein as a model for surface proteins in general, pointing out differences with other surface molecules when necessary.
Collapse
Affiliation(s)
- Vincent A Fischetti
- Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, NY 10065
| |
Collapse
|
109
|
Anderl A, Ferlemann C, Muth M, Henkel-Gupalo A, Ebenig A, Brenner-Weiß G, Kolmar H, Fuchsbauer HL. Biochemical study of sortase E2 from Streptomyces mobaraensis and determination of transglutaminase cross-linking sites. FEBS Lett 2019; 593:1944-1956. [PMID: 31155711 DOI: 10.1002/1873-3468.13466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022]
Abstract
Distinct streptomycetes such as Streptomyces mobaraensis produce the protein cross-linking enzyme transglutaminase. Bioinformatic analysis predicted the occurrence of seven sortases exerting transpeptidation reactions similarly to transglutaminase. Here, we report the production and characterization of sortase E2 (Sm-SrtE2) solubilized by removal of its membrane anchor domain. Sm-SrtE2 activity was measured using pentapeptides predicted to be cell wall sorting signals of putative sortase substrate proteins. Preferred linkage to Gly3 by Sm-SrtE2 was in the order LAETG>>LAHTG>>LAQTG~LANTG>LARTG. Chaplin 1 from S. mobaraensis was further demonstrated to be an excellent substrate of both the intrinsic Sm-SrtE2 and transglutaminase. The unexpected discovery showing Gln-62 and Gln-65 of Δ1-50 -Sm-SrtE2 as transglutaminase cross-linking sites suggests that low enzyme stability might be due to anchor domain truncation and a disordered N terminus.
Collapse
Affiliation(s)
- Anita Anderl
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Germany.,Department of Chemistry, Technische Universität Darmstadt, Germany
| | - Cathrin Ferlemann
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Germany
| | - Marius Muth
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Germany.,Bioengineering and Biosystems, Institute of Functional Interfaces, Karlsruhe Institute of Technology, Germany
| | - Antonina Henkel-Gupalo
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Germany
| | - Aileen Ebenig
- Department of Chemistry, Technische Universität Darmstadt, Germany
| | - Gerald Brenner-Weiß
- Bioengineering and Biosystems, Institute of Functional Interfaces, Karlsruhe Institute of Technology, Germany
| | - Harald Kolmar
- Department of Chemistry, Technische Universität Darmstadt, Germany
| | - Hans-Lothar Fuchsbauer
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Germany
| |
Collapse
|
110
|
Abstract
Bacteria in the genus Staphylococcus are important targets for phage therapy due to their prevalence as pathogens and increasing antibiotic resistance. Here we review Staphylococcus outer surface features and specific phage resistance mechanisms that define the host range, the set of strains that an individual phage can potentially infect. Phage infection goes through five distinct phases: attachment, uptake, biosynthesis, assembly, and lysis. Adsorption inhibition, encompassing outer surface teichoic acid receptor alteration, elimination, or occlusion, limits successful phage attachment and entry. Restriction-modification systems (in particular, type I and IV systems), which target phage DNA inside the cell, serve as the major barriers to biosynthesis as well as transduction and horizontal gene transfer between clonal complexes and species. Resistance to late stages of infection occurs through mechanisms such as assembly interference, in which staphylococcal pathogenicity islands siphon away superinfecting phage proteins to package their own DNA. While genes responsible for teichoic acid biosynthesis, capsule, and restriction-modification are found in most Staphylococcus strains, a variety of other host range determinants (e.g., clustered regularly interspaced short palindromic repeats, abortive infection, and superinfection immunity) are sporadic. The fitness costs of phage resistance through teichoic acid structure alteration could make staphylococcal phage therapies promising, but host range prediction is complex because of the large number of genes involved, and the roles of many of these are unknown. In addition, little is known about the genetic determinants that contribute to host range expansion in the phages themselves. Future research must identify host range determinants, characterize resistance development during infection and treatment, and examine population-wide genetic background effects on resistance selection.
Collapse
Affiliation(s)
- Abraham G Moller
- Program in Microbiology and Molecular Genetics (MMG), Graduate Division of Biological and Biomedical Sciences (GDBBS), Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jodi A Lindsay
- Institute of Infection and Immunity, St. George's, University of London, London, United Kingdom
| | - Timothy D Read
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
111
|
A comprehensive in silico analysis of sortase superfamily. J Microbiol 2019; 57:431-443. [DOI: 10.1007/s12275-019-8545-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 12/22/2022]
|
112
|
Sortase-Dependent Proteins Promote Gastrointestinal Colonization by Enterococci. Infect Immun 2019; 87:IAI.00853-18. [PMID: 30804098 DOI: 10.1128/iai.00853-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
The human gastrointestinal tract (GIT) is inhabited by a dense microbial community of symbionts. Enterococci are among the earliest members of this community and remain core members of the GIT microbiota throughout life. Enterococci have also recently emerged as opportunistic pathogens and major causes of nosocomial infections. Although recognized as a prerequisite for infection, colonization of the GIT by enterococci remains poorly understood. One way that bacteria adapt to dynamic ecosystems like the GIT is through the use of their surface proteins to sense and interact with components of their immediate environment. In Gram-positive bacteria, a subset of surface proteins relies on an enzyme called sortase for covalent attachment to the cell wall. Here, we show that the housekeeping sortase A (SrtA) enzyme promotes intestinal colonization by enterococci. Furthermore, we show that the enzymatic activity of SrtA is key to the ability of Enterococcus faecalis to bind mucin (a major component of the GIT mucus). We also report the GIT colonization phenotypes of E. faecalis mutants lacking selected sortase-dependent proteins (SDPs). Further examination of the mucin binding ability of these mutants suggests that adhesion to mucin contributes to intestinal colonization by E. faecalis.
Collapse
|
113
|
Hsu YP, Hall E, Booher G, Murphy B, Radkov AD, Yablonowski J, Mulcahey C, Alvarez L, Cava F, Brun YV, Kuru E, VanNieuwenhze MS. Fluorogenic D-amino acids enable real-time monitoring of peptidoglycan biosynthesis and high-throughput transpeptidation assays. Nat Chem 2019; 11:335-341. [PMID: 30804500 PMCID: PMC6444347 DOI: 10.1038/s41557-019-0217-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 01/10/2019] [Indexed: 01/09/2023]
Abstract
Peptidoglycan (PG) is an essential cell wall component that maintains the morphology and viability of nearly all bacteria. Its biosynthesis requires periplasmic transpeptidation reactions which construct peptide cross-linkages between polysaccharide chains to endow mechanical strength. However, tracking transpeptidation reaction in vivo and in vitro is challenging, mainly due to the lack of efficient, biocompatible probes. Here, we report the design, synthesis, and application of rotor-fluorogenic D-amino acids (RfDAAs) enabling real-time, continuous tracking of transpeptidation reactions. These probes enable monitoring PG biosynthesis in real time through visualizing transpeptidase reactions in live cells, as well as real-time activity assays of D,D-, L,D-transpeptidases, and sortases in vitro. The unique ability of RfDAAs to become fluorescent when incorporated into PG provides a powerful new tool to study PG biosynthesis with high temporal resolution and prospectively enable high-throughput screening for inhibitors of PG biosynthesis.
Collapse
Affiliation(s)
- Yen-Pang Hsu
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Edward Hall
- Department of Chemistry, Indiana University, Bloomington, IN, USA.,Department of Chemistry, Hanover College, Hanover, IN, USA
| | - Garrett Booher
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Brennan Murphy
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Atanas D Radkov
- Department of Chemistry, Indiana University, Bloomington, IN, USA.,Department of Biophysics and Biochemistry, University of California San Francisco, San Francisco, CA, USA
| | - Jacob Yablonowski
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Caitlyn Mulcahey
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Laura Alvarez
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Felipe Cava
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Yves V Brun
- Department of Biology, Indiana University, Bloomington, IN, USA. .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Pavillon Roger-Gaudry, C.P. 6128, Succursale Centre-ville, Montréal, Canada.
| | - Erkin Kuru
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA. .,Department of Genetics, Harvard Medical School, Boston, MA, USA.
| | - Michael S VanNieuwenhze
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA. .,Department of Chemistry, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
114
|
Astilbin Inhibits the Activity of Sortase A from Streptococcus mutans. Molecules 2019; 24:molecules24030465. [PMID: 30696091 PMCID: PMC6384703 DOI: 10.3390/molecules24030465] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 11/17/2022] Open
Abstract
Streptococcus mutans (S. mutans) is the primary etiological agent of dental caries. The S. mutans enzyme sortase A (SrtA) is responsible for anchoring bacterial cell wall surface proteins involved in host cell attachment and biofilm formation. Thus, SrtA is an attractive target for inhibiting dental caries caused by S. mutans-associated acid fermentation. In this study, we observed that astilbin, a flavanone compound extracted from Rhizoma Smilacis Glabrae, has potent inhibitory activity against the S. mutans SrtA, with an IC50 of 7.5 μg/mL. In addition, astilbin was proven to reduce the formation of biofilm while without affecting the growth of S. mutans. The results of a molecular dynamics simulation and a mutation analysis revealed that the Arg213, Leu111, and Leu116 of SrtA are important for the interaction between SrtA and astilbin. The results of this study demonstrate the potential of using astilbin as a nonbactericidal agent to modulate pathogenicity of S. mutans by inhibiting the activity of SrtA.
Collapse
|
115
|
Gébleux R, Briendl M, Grawunder U, Beerli RR. Sortase A Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody-Drug Conjugates. Methods Mol Biol 2019; 2012:1-13. [PMID: 31161500 DOI: 10.1007/978-1-4939-9546-2_1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Antibody-drug conjugates (ADCs) are highly potent targeted anticancer therapies. They rely on the linking of a selectively targeting antibody moiety with potent cytotoxic payloads to effect antitumoral activity. In recent years, one focus in the ADC field was to create novel methods for site-specifically conjugating payloads to antibodies. The method presented here is based on the S. aureus sortase A-mediated transpeptidation reaction. This method requires antibodies to be engineered in such a way that they possess the sortase recognition pentapeptide motif LPETG on the C-terminus of the immunoglobulin heavy and/or light chains. In addition, the toxin must contain an oligoglycine motif in order to make it a suitable substrate for sortase A. Here we describe a detailed method to conjugate a pentaglycine-modified toxin to the C-termini of LPETG-tagged antibody heavy and light chains using sortase-mediated antibody conjugation (SMAC-Technology™). Highly homogenous, site-specifically conjugated ADCs with controlled drug to antibody ratio and improved overall properties can be obtained with this method.
Collapse
|
116
|
Schneewind O, Missiakas D. Sortases, Surface Proteins, and Their Roles in Staphylococcus aureus Disease and Vaccine Development. Microbiol Spectr 2019; 7:10.1128/microbiolspec.psib-0004-2018. [PMID: 30737913 PMCID: PMC6386163 DOI: 10.1128/microbiolspec.psib-0004-2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Indexed: 12/27/2022] Open
Abstract
Sortases cleave short peptide motif sequences at the C-terminal end of secreted surface protein precursors and either attach these polypeptides to the peptidoglycan of Gram-positive bacteria or promote their assembly into pilus structures that are also attached to peptidoglycan. Sortase A, the enzyme first identified in the human pathogen Staphylococcus aureus, binds LPXTG motif sorting signals, cleaves between threonine (T) and glycine (G) residues, and forms an acyl enzyme between its active-site cysteine thiol and the carboxyl group of threonine (T). Sortase A acyl enzyme is relieved by the nucleophilic attack of the cross bridge amino group within lipid II, thereby generating surface protein linked to peptidoglycan precursor. Such products are subsequently incorporated into the cell wall envelope by enzymes of the peptidoglycan synthesis pathway. Surface proteins linked to peptidoglycan may be released from the bacterial envelope to diffuse into host tissues and fulfill specific biological functions. S. aureus sortase A is essential for host colonization and for the pathogenesis of invasive diseases. Staphylococcal sortase-anchored surface proteins fulfill key functions during the infectious process, and vaccine-induced antibodies targeting surface proteins may provide protection against S. aureus. Alternatively, small-molecule inhibitors of sortase may be useful agents for the prevention of S. aureus colonization and invasive disease.
Collapse
Affiliation(s)
- Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, IL 60637
| | | |
Collapse
|
117
|
Cheng X, Hong H, Zhou Z, Wu Z. Enzymatic On-Resin Peptide Cleavage and in Situ Cyclization One-Pot Strategy for the Synthesis of Cyclopeptide and Cyclotide. J Org Chem 2018; 83:14078-14083. [PMID: 30277068 DOI: 10.1021/acs.joc.8b02032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A one-pot strategy combining sortase A mediated on-resin peptide cleavage and in situ cyclization was developed for the synthesis of cyclic peptides. This strategy was applied to synthesize head-to-tail cyclic antibacterial bovine lactoferricin peptide LFcinB20-35 in a yield of 67%. The one-pot strategy was compatible with an oxidative folding reaction, and complex cyclotides containing one or two disulfide bonds, such as sunflower trypsin inhibitors-1 and α-conotoxin MII, were successfully synthesized in one pot in a yield of 77% and 61%, respectively.
Collapse
Affiliation(s)
- Xiaozhong Cheng
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , Wuxi 214122 , China
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , Wuxi 214122 , China
| | - Zhifang Zhou
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , Wuxi 214122 , China
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , Wuxi 214122 , China
| |
Collapse
|
118
|
Ca2+ binding induced sequential allosteric activation of sortase A: An example for ion-triggered conformational selection. PLoS One 2018; 13:e0205057. [PMID: 30321208 PMCID: PMC6188747 DOI: 10.1371/journal.pone.0205057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/19/2018] [Indexed: 12/11/2022] Open
Abstract
The allosteric activation of the intrinsically disordered enzyme Staphylococcus aureus sortase A is initiated via binding of a Ca2+ ion. Although Ca2+ binding was shown to initiate structural changes inducing disorder-to-order transitions, the details of the allosteric activation mechanism remain elusive. We performed long-term molecular dynamics simulations of sortase A without (3 simulations of 1.6 μs) and with bound Ca2+ (simulations of 1.6 μs, 1.8 μs, and 2.5 μs). Our results show that Ca2+ binding causes not only ordering of the disordered β6/β7 loop of the protein, but also modulates hinge motions in the dynamic β7/β8 loop, which is important for the catalytic activity of the enzyme. Cation binding triggers signal transmission from the Ca2+ binding site to the dynamic β7/β8 loop via the repetitive folding/unfolding of short helical stretches of the disordered β6/β7 loop. These correlated structural rearrangements lead to several distinct conformational states of the binding groove, which show optimal binding features for the sorting signal motif and feature binding energies up to 20 kcal/mol more favorable than observed for the sortase A without Ca2+. The presented results indicate a highly correlated, conformational selection-based activation mechanism of the enzyme triggered by cation binding. They also demonstrate the importance of the dynamics of intrinsically disordered regions for allosteric regulation.
Collapse
|
119
|
Identification of potential inhibitors of sortase A: Binding studies, in-silico docking and protein-protein interaction studies of sortase A from Enterococcus faecalis. Int J Biol Macromol 2018; 120:1906-1916. [PMID: 30268755 DOI: 10.1016/j.ijbiomac.2018.09.174] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022]
Abstract
Enterococcus faecalis (Ef) is a Gram positive multidrug resistant (MDR) bacterium contributing about 70% of total enterococcal infections. In Ef, a membrane anchored transpeptidase Sortase A plays a major role in biofilm formation. Therefore, it has been recognized as an ideal drug target against Ef. In this regard to identify the potential inhibitors of Ef Sortase A (EfSrtA∆59), we have cloned, expressed and purified EfSrtA∆59. We have also done the in-silico docking studies to identify lead molecules interacting with EfSrtA∆59. Furthermore, the binding studies of these identified lead molecules were performed with EfSrtA∆59 using fluorescence and CD spectroscopic studies. We also identified the interaction partner of EfSrtA∆59 using STRING. Protein-protein docking studies were also performed. Docking experiment revealed that benzylpenicillin, cefotaxime, pantoprazole and valsartan were bound to same site on the protein with similar interactions. Binding studies using fluorescence spectroscopic studies confirmed the binding of all the ligands to EfSrtA∆59, which was further validated by far and near-UV CD experiments. Thermo stability experiments validate the stability-activity trade-off hypothesis. Sequence based interaction studies identified that EfSrtA∆59 interact with the Ef_1091, Ef_1093 and Ef_2658 proteins. Homology model of Ef_1091 and Ef_1093 was docked with modeled EfSrtA∆59 and their interactions are also discussed.
Collapse
|
120
|
Erianin against Staphylococcus aureus Infection via Inhibiting Sortase A. Toxins (Basel) 2018; 10:toxins10100385. [PMID: 30249042 PMCID: PMC6215257 DOI: 10.3390/toxins10100385] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/08/2018] [Accepted: 09/20/2018] [Indexed: 02/04/2023] Open
Abstract
With continuous emergence and widespread of multidrug-resistant Staphylococcus aureus infections, common antibiotics have become ineffective in treating these infections in the clinical setting. Anti-virulence strategies could be novel, effective therapeutic strategies against drug-resistant bacterial infections. Sortase A (srtA), a transpeptidase in gram-positive bacteria, can anchor surface proteins that play a vital role in pathogenesis of these bacteria. SrtA is known as a potential antivirulent drug target to treat bacterial infections. In this study, we found that erianin, a natural bibenzyl compound, could inhibit the activity of srtA in vitro (half maximal inhibitory concentration-IC50 = 20.91 ± 2.31 μg/mL, 65.7 ± 7.2 μM) at subminimum inhibitory concentrations (minimum inhibitory concentrations-MIC = 512 μg/mL against S. aureus). The molecular mechanism underlying the inhibition of srtA by erianin was identified using molecular dynamics simulation: erianin binds to srtA residues Ile182, Val193, Trp194, Arg197, and Ile199, forming a stable bond via hydrophobic interactions. In addition, the activities of S. aureus binding to fibronectin and biofilm formation were inhibited by erianin, when co-culture with S. aureus. In vivo, erianin could improve the survival in mice that infected with S. aureus by tail vein injection. Experimental results showed that erianin is a potential novel therapeutic compound against S. aureus infections via affecting srtA.
Collapse
|
121
|
Stanborough T, Suryadinata R, Fegan N, Powell SM, Tamplin M, Nuttall SD, Chandry PS. Characterisation of the Brochothrix thermosphacta sortase A enzyme. FEMS Microbiol Lett 2018; 365:5056718. [PMID: 30052925 DOI: 10.1093/femsle/fny184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
Gram-positive bacteria utilise class A sortases to coat the surface of their cells with a diversity of proteins that facilitate interactions with their environment and play fundamental roles in cell physiology and virulence. A putative sortase A gene was identified in the genome of the poorly studied meat spoilage bacterium Brochothrix thermosphacta. To understand how this bacterium mediates interactions with its environment, an N-terminal truncated, His-tagged variant of this protein (His6-BtSrtA) was expressed and purified. Catalytic activity of recombinant His6-BtSrtA was investigated, including sorting motif recognition of target proteins and bioconjugation activity. Further, the B. thermosphacta genome was examined for the presence of sortase A (SrtA) protein substrates. His6-BtSrtA readily formed intermediate complexes with LPXTG-tagged proteins. Although the reaction was inefficient, nucleophilic attack of the resultant thioacyl intermediates by tri-glycine was observed. Genome examination identified 11 potential SrtA substrates, two of which contained protein domains associated with adherence of pathogens to host extracellular matrix proteins and cells, suggesting the B. thermosphacta SrtA may be indirectly involved in its attachment to meat surfaces. Thus, further work in this area could provide crucial insight into molecular mechanisms involved in the colonisation of meat by B. thermosphacta.
Collapse
Affiliation(s)
- Tamsyn Stanborough
- CSIRO Agriculture and Food, Werribee, VIC 3030, Australia.,CSIRO Manufacturing, Parkville, VIC 3030, Australia
| | | | - Narelle Fegan
- CSIRO Agriculture and Food, Werribee, VIC 3030, Australia
| | - Shane M Powell
- Tasmanian Institute of Agriculture, University of Tasmania, Hobart, TAS 7001, Australia
| | - Mark Tamplin
- Tasmanian Institute of Agriculture, University of Tasmania, Hobart, TAS 7001, Australia
| | | | | |
Collapse
|
122
|
Agwa AJ, Blomster LV, Craik DJ, King GF, Schroeder CI. Efficient Enzymatic Ligation of Inhibitor Cystine Knot Spider Venom Peptides: Using Sortase A To Form Double-Knottins That Probe Voltage-Gated Sodium Channel NaV1.7. Bioconjug Chem 2018; 29:3309-3319. [DOI: 10.1021/acs.bioconjchem.8b00505] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Linda V. Blomster
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J. Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
123
|
Bartels L, Ploegh HL, Spits H, Wagner K. Preparation of bispecific antibody-protein adducts by site-specific chemo-enzymatic conjugation. Methods 2018; 154:93-101. [PMID: 30081077 DOI: 10.1016/j.ymeth.2018.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/25/2018] [Accepted: 07/31/2018] [Indexed: 11/29/2022] Open
Abstract
Historically, bispecific antibodies have been constructed through the genetic fusion of additional binding domains to the constant domains of the antibody heavy- or light chains. We present an alternative method for the introduction of additional functional domains to an antibody: site-specific chemo-enzymatic conjugation. This method relies on the combination of site-specific transpeptidases and bioorthogonal chemistry. Transpeptidases are used to site-specifically introduce chemical handles, which can then be used to couple new functional groups by means of a bioorthogonal chemical reaction. We demonstrate site-specific chemo-enzymatic linkage using the transpeptidase sortase (hereafter: sortase) and either a strain-promoted alkyne-azide cycloaddition (SPAAC) or an inverse-electron demand Diels-Alder reaction. Other transpeptidases and bioorthogonal reactions suitable for this purpose exist. Site-specific chemo-enzymatic linkage is a modular method. After introduction of a chemical handle in the antibody, any functional group of interest may then be attached. The modularity of this conjugation method allows for a 'plug-and-play' approach to prepare new antibody conjugates, thus bypassing the need for (potentially) laborious genetic fusions. Moreover, as sortase is used to specifically modify the exact C-termini of the antibody chains, the final product will be fused in a C-to-C orientation, which is impossible to achieve by genetic manipulations alone. Here we demonstrate the utility of site-specific chemo-enzymatic conjugation to prepare antibody heterodimers, bispecific T-cell engager antibodies, and immunocytokines, discussing purification methods and describing possible pitfalls.
Collapse
Affiliation(s)
- Lina Bartels
- AIMM Therapeutics, Amsterdam, Netherlands; Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Boston, MA, United States
| | - Hergen Spits
- AIMM Therapeutics, Amsterdam, Netherlands; Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | | |
Collapse
|
124
|
Miljkovic M, Marinkovic P, Novovic K, Jovcic B, Terzic-Vidojevic A, Kojic M. AggLr, a novel aggregation factor in Lactococcus raffinolactis BGTRK10-1: its role in surface adhesion. BIOFOULING 2018; 34:685-698. [PMID: 30027759 DOI: 10.1080/08927014.2018.1481956] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/16/2018] [Indexed: 06/08/2023]
Abstract
The ability of lactic acid bacteria to form multi-cellular aggregates via self-aggregation is regarded as an important mechanism for stress tolerance, adhesion, colonization and genetic material exchange. The novel aggLr gene encoding for the auto-aggregation promoting protein (AggLr) of Lactococcus raffinolactis BGTRK10-1 was cloned. Heterologous expression of AggLr enabled auto-aggregation, higher hydrophobicity and collagen and fibronectin binding of the carrier strains. Domain analysis and the type of aggregates formed by cells expressing AggLr confirmed that this aggregation factor belongs to the family of high molecular weight proteins that the authors propose to be called Snow-flake Forming Collagen Binding Aggregation Factors (SFCBAF). An additional feature of SFCBAF is that they are rich in threonine and lysine and are free of cysteine in all of the aggregation factors described so far. In contrast to previously discovered SFCBAF, the gene encoding for AggLr is located on the chromosome in the strain BGTRK10-1.
Collapse
Affiliation(s)
- Marija Miljkovic
- a Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering , University of Belgrade , Belgrade , Serbia
| | - Pavle Marinkovic
- a Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering , University of Belgrade , Belgrade , Serbia
| | - Katarina Novovic
- a Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering , University of Belgrade , Belgrade , Serbia
| | - Branko Jovcic
- a Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering , University of Belgrade , Belgrade , Serbia
- b Faculty of Biology , University of Belgrade , Belgrade , Serbia
| | - Amarela Terzic-Vidojevic
- a Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering , University of Belgrade , Belgrade , Serbia
| | - Milan Kojic
- a Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering , University of Belgrade , Belgrade , Serbia
| |
Collapse
|
125
|
|
126
|
|
127
|
Chang C, Amer BR, Osipiuk J, McConnell SA, Huang IH, Hsieh V, Fu J, Nguyen HH, Muroski J, Flores E, Ogorzalek Loo RR, Loo JA, Putkey JA, Joachimiak A, Das A, Clubb RT, Ton-That H. In vitro reconstitution of sortase-catalyzed pilus polymerization reveals structural elements involved in pilin cross-linking. Proc Natl Acad Sci U S A 2018; 115:E5477-E5486. [PMID: 29844180 PMCID: PMC6004493 DOI: 10.1073/pnas.1800954115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Covalently cross-linked pilus polymers displayed on the cell surface of Gram-positive bacteria are assembled by class C sortase enzymes. These pilus-specific transpeptidases located on the bacterial membrane catalyze a two-step protein ligation reaction, first cleaving the LPXTG motif of one pilin protomer to form an acyl-enzyme intermediate and then joining the terminal Thr to the nucleophilic Lys residue residing within the pilin motif of another pilin protomer. To date, the determinants of class C enzymes that uniquely enable them to construct pili remain unknown. Here, informed by high-resolution crystal structures of corynebacterial pilus-specific sortase (SrtA) and utilizing a structural variant of the enzyme (SrtA2M), whose catalytic pocket has been unmasked by activating mutations, we successfully reconstituted in vitro polymerization of the cognate major pilin (SpaA). Mass spectrometry, electron microscopy, and biochemical experiments authenticated that SrtA2M synthesizes pilus fibers with correct Lys-Thr isopeptide bonds linking individual pilins via a thioacyl intermediate. Structural modeling of the SpaA-SrtA-SpaA polymerization intermediate depicts SrtA2M sandwiched between the N- and C-terminal domains of SpaA harboring the reactive pilin and LPXTG motifs, respectively. Remarkably, the model uncovered a conserved TP(Y/L)XIN(S/T)H signature sequence following the catalytic Cys, in which the alanine substitutions abrogated cross-linking activity but not cleavage of LPXTG. These insights and our evidence that SrtA2M can terminate pilus polymerization by joining the terminal pilin SpaB to SpaA and catalyze ligation of isolated SpaA domains in vitro provide a facile and versatile platform for protein engineering and bio-conjugation that has major implications for biotechnology.
Collapse
Affiliation(s)
- Chungyu Chang
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, TX 77030
| | - Brendan R Amer
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - Jerzy Osipiuk
- Center for Structural Genomics of Infectious Diseases, Argonne National Laboratory, Argonne, IL 60439
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Scott A McConnell
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - I-Hsiu Huang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Van Hsieh
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - Janine Fu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - Hong H Nguyen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - John Muroski
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - Erika Flores
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, TX 77030
| | - Rachel R Ogorzalek Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - John A Putkey
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX 77030
| | - Andrzej Joachimiak
- Center for Structural Genomics of Infectious Diseases, Argonne National Laboratory, Argonne, IL 60439
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Asis Das
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030
| | - Robert T Clubb
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095;
- University of California, Los Angeles-US Department of Energy Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095
| | - Hung Ton-That
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, TX 77030;
| |
Collapse
|
128
|
Yu W, Missiakas D, Schneewind O. Septal secretion of protein A in Staphylococcus aureus requires SecA and lipoteichoic acid synthesis. eLife 2018; 7:34092. [PMID: 29757141 PMCID: PMC5962339 DOI: 10.7554/elife.34092] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/05/2018] [Indexed: 12/26/2022] Open
Abstract
Surface proteins of Staphylococcus aureus are secreted across septal membranes for assembly into the bacterial cross-wall. This localized secretion requires the YSIRK/GXXS motif signal peptide, however the mechanisms supporting precursor trafficking are not known. We show here that the signal peptide of staphylococcal protein A (SpA) is cleaved at the YSIRK/GXXS motif. A SpA signal peptide mutant defective for YSIRK/GXXS cleavage is also impaired for septal secretion and co-purifies with SecA, SecDF and LtaS. SecA depletion blocks precursor targeting to septal membranes, whereas deletion of secDF diminishes SpA secretion into the cross-wall. Depletion of LtaS blocks lipoteichoic acid synthesis and abolishes SpA precursor trafficking to septal membranes. We propose a model whereby SecA directs SpA precursors to lipoteichoic acid-rich septal membranes for YSIRK/GXXS motif cleavage and secretion into the cross-wall.
Collapse
Affiliation(s)
- Wenqi Yu
- Department of Microbiology, University of Chicago, Chicago, United States
| | | | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, United States
| |
Collapse
|
129
|
Zou Z, Mate DM, Rübsam K, Jakob F, Schwaneberg U. Sortase-Mediated High-Throughput Screening Platform for Directed Enzyme Evolution. ACS COMBINATORIAL SCIENCE 2018; 20:203-211. [PMID: 29363945 DOI: 10.1021/acscombsci.7b00153] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sortase-catalyzed ligations have emerged as powerful tools for the site-specific ligation of peptides and proteins in material science and biocatalysis. In this work, a directed sortase evolution strategy (SortEvolve) has been developed as a general high-throughput screening (HTS) platform to improve activity of sortase A (application 1) and to perform directed laccase evolution through a semipurification process in 96-well microtiter plate (MTP) (application 2). A semipurification process in polypropylene MTP (PP-MTP) is achieved through the anchor peptide LCI, which acts as adhesion promoter. To validate the SortEvolve screening platform for both applications, three site-saturation mutagenesis (SSM) libraries of sortase A (Sa-SrtA) from Staphylococcus aureus (application 1) and two SSM libraries of the copper efflux oxidase (CueO laccase) from Escherichia coli (application 2) were generated at literature reported positions. After screening and rescreening, an array of Sa-SrtA variants (including the previously reported P94S, D160N, and D165A) and CueO variants (including the previously reported D439A and P444A) were identified. Further recombinant Sa-SrtA variant P94T/D160L/D165Q and CueO variant D439V/P444V were characterized with 22-fold and 103-fold improvements in catalytic efficiency compared with corresponding wild-types, respectively. An important advantage of the SortEvolve screening platform in comparison to many MTP-based screening systems is that the background noise was minimized (decreased 20-fold; application 2) due to the employed semipurification process. In essence, SortEvolve provides a universal surface-functionalized screening platform for sortases and enzymes in which especially background activity can be minimized to enable successful directed evolution campaigns.
Collapse
Affiliation(s)
- Zhi Zou
- DWI—Leibniz-Institute for Interactive Materials, Forckenbeckstraße 50, 52074 Aachen, Germany
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Diana M. Mate
- DWI—Leibniz-Institute for Interactive Materials, Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Kristin Rübsam
- DWI—Leibniz-Institute for Interactive Materials, Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Felix Jakob
- DWI—Leibniz-Institute for Interactive Materials, Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Ulrich Schwaneberg
- DWI—Leibniz-Institute for Interactive Materials, Forckenbeckstraße 50, 52074 Aachen, Germany
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| |
Collapse
|
130
|
Niu X, Gao Y, Yu Y, Yang Y, Wang G, Sun L, Wang H. Molecular Modelling reveals the inhibition mechanism and structure-activity relationship of curcumin and its analogues to Staphylococcal aureus Sortase A. J Biomol Struct Dyn 2018; 37:1220-1230. [PMID: 29546799 DOI: 10.1080/07391102.2018.1453380] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Previous studies found that the activity of Sortase A, a bacterial surface protein from Staphylococcus aureus, was inhibited by curcumin and its analogues. To explore this inhibitory mechanism, Sortase A and its inhibitors in complex systems were studied by molecular docking, molecular modelling, binding energy decomposition calculation and steered molecular dynamics simulations. Energy decomposition analysis indicated that PRO-163, LEU-169, GLN-172, ILE-182 and ILE-199 are key residues in Sortase A-inhibitor complexes. Furthermore, interactions between the methoxyl group on the benzene ring in the conjugated molecule (curcumin, demethoxycurcumin, bisdemethoxycurcumin) and VAL-168, LEU-169 and GLN-172 induce the inhibitory activity based on the energy decomposition and distance analyses between the whole residues and inhibitors. However, because of its coiled structure, the non-conjugated molecule, tetrahydrocurcumin, with key residues in the binding sites of Sortase A, interacted weakly with SrtA, leading to the loss of inhibitory activity. Based on these results, the methoxyl group on the benzene ring in the conjugated molecule largely influenced the inhibitory activity of the Sortase A inhibitors.
Collapse
Affiliation(s)
- Xiaodi Niu
- a College of Food Science and Engineering , Jilin University , Changchun , China
| | - Yawen Gao
- a College of Food Science and Engineering , Jilin University , Changchun , China
| | - Yiding Yu
- a College of Food Science and Engineering , Jilin University , Changchun , China
| | - Yanan Yang
- a College of Food Science and Engineering , Jilin University , Changchun , China
| | - Guizhen Wang
- a College of Food Science and Engineering , Jilin University , Changchun , China
| | - Lin Sun
- a College of Food Science and Engineering , Jilin University , Changchun , China
| | - Hongsu Wang
- a College of Food Science and Engineering , Jilin University , Changchun , China
| |
Collapse
|
131
|
Wu Z, Wang G, Wang W, Pan D, Peng L, Lian L. Proteomics Analysis of the Adhesion Activity ofLactobacillus acidophilusATCC 4356 Upon Growth in an Intestine-Like pH Environment. Proteomics 2018; 18:e1700308. [DOI: 10.1002/pmic.201700308] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/22/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Zhen Wu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province; Marine Science School; Ningbo University; Ningbo P. R. China
| | - Gang Wang
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province; Marine Science School; Ningbo University; Ningbo P. R. China
| | - Wenwen Wang
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province; Marine Science School; Ningbo University; Ningbo P. R. China
| | - Daodong Pan
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province; Marine Science School; Ningbo University; Ningbo P. R. China
- Department of Food Science and Nutrition; Jinling College; Nanjing Normal University; Nanjing P.R. China
| | - Liuyang Peng
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province; Marine Science School; Ningbo University; Ningbo P. R. China
| | | |
Collapse
|
132
|
Abdul Halim MF, Rodriguez R, Stoltzfus JD, Duggin IG, Pohlschroder M. Conserved residues are critical for Haloferax volcanii archaeosortase catalytic activity: Implications for convergent evolution of the catalytic mechanisms of non-homologous sortases from archaea and bacteria. Mol Microbiol 2018; 108:276-287. [PMID: 29465796 DOI: 10.1111/mmi.13935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2018] [Indexed: 11/29/2022]
Abstract
Proper protein anchoring is key to the biogenesis of prokaryotic cell surfaces, dynamic, resilient structures that play crucial roles in various cell processes. A novel surface protein anchoring mechanism in Haloferax volcanii depends upon the peptidase archaeosortase A (ArtA) processing C-termini of substrates containing C-terminal tripartite structures and anchoring mature substrates to the cell membrane via intercalation of lipid-modified C-terminal amino acid residues. While this membrane protein lacks clear homology to soluble sortase transpeptidases of Gram-positive bacteria, which also process C-termini of substrates whose C-terminal tripartite structures resemble those of ArtA substrates, archaeosortases do contain conserved cysteine, arginine and arginine/histidine/asparagine residues, reminiscent of His-Cys-Arg residues of sortase catalytic sites. The study presented here shows that ArtAWT -GFP expressed in trans complements ΔartA growth and motility phenotypes, while alanine substitution mutants, Cys173 (C173A), Arg214 (R214A) or Arg253 (R253A), and the serine substitution mutant for Cys173 (C173S), fail to complement these phenotypes. Consistent with sortase active site replacement mutants, ArtAC173A -GFP, ArtAC173S -GFP and ArtAR214A -GFP cannot process substrates, while replacement of the third residue, ArtAR253A -GFP retains some processing activity. These findings support the view that similarities between certain aspects of the structures and functions of the sortases and archaeosortases are the result of convergent evolution.
Collapse
Affiliation(s)
| | - Ronald Rodriguez
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Iain G Duggin
- The ithree Institute, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | | |
Collapse
|
133
|
Evans ED, Pentelute BL. Discovery of a 29-Amino-Acid Reactive Abiotic Peptide for Selective Cysteine Arylation. ACS Chem Biol 2018; 13:527-532. [PMID: 29283243 DOI: 10.1021/acschembio.7b00520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The regio- and chemoselective modification of proteins or peptides with chemical reagents is often challenging. One approach to overcome this problem involves identifying abiotic polypeptide sequences that react with specific small molecules. Toward this goal, we profiled ∼5 × 1013 randomized 30-mer peptides using mRNA display and high-throughput sequencing in search of polypeptides that can undergo cysteine arylation with a water-soluble perfluoroarene. Within this vast chemical space, we discovered a cysteine-containing sequence with a second-order rate constant of 0.29 M-1 s-1 for arylation. An N- and C-terminal truncation reduced the reaction rate, as did the addition of denaturants. When the reactive peptide was covalently fused to the enzyme Sortase A, we observed regiospecific arylation at a single cysteine site, leaving the enzyme's active site cysteine unchanged. Taken together, these results demonstrate that long polypeptides of defined sequence, when matched with the appropriate reactive group, can be used for selective arylation of cysteine in water.
Collapse
Affiliation(s)
- Ethan D. Evans
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley L. Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
134
|
Yang M, Hong H, Liu S, Zhao X, Wu Z. Immobilization of Staphylococcus aureus Sortase A on Chitosan Particles and Its Applications in Peptide-to-Peptide Ligation and Peptide Cyclization. Molecules 2018; 23:molecules23010192. [PMID: 29351256 PMCID: PMC6017383 DOI: 10.3390/molecules23010192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/11/2018] [Accepted: 01/14/2018] [Indexed: 11/16/2022] Open
Abstract
Chitosan macro-particles prepared by the neutralization method were applied to Sortase A (SrtA) immobilization using glutaraldehyde as a crosslinking agent. The particles were characterized by Fourier transform infrared spectroscopy (FTIR) and scanning electron microscopy (SEM). Response surface methodology (RSM) was employed to optimize the immobilization process. An average specific activity of 3142 U (mg protein)-1 was obtained under optimized immobilization conditions (chitosan concentration 3%, SrtA concentration 0.5 mg·mL-1, glutaraldehyde concentration 0.5%, crosslinking and immobilization at 20 °C, crosslinking for 3 h, and an immobilization time of 8 h). The transpeptidase activity of immobilized SrtA was proved by a peptide-to-peptide ligation with a conversion yield approximately at 80%, and the immobilized catalyst was successfully reused for five cycles without obvious activity loss. Moreover, the scale-up capability of using immobilized SrtA to catalyze a head-to-tail peptide cyclization was investigated in a batch reaction and the conversion yield was more than 95% when using 20 mg of peptide as a substrate.
Collapse
Affiliation(s)
- Min Yang
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Shaozhong Liu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Xinrui Zhao
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
135
|
Wang-Lin SX, Balthasar JP. Pharmacokinetic and Pharmacodynamic Considerations for the Use of Monoclonal Antibodies in the Treatment of Bacterial Infections. Antibodies (Basel) 2018; 7:antib7010005. [PMID: 31544858 PMCID: PMC6698815 DOI: 10.3390/antib7010005] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/01/2018] [Accepted: 01/02/2018] [Indexed: 12/17/2022] Open
Abstract
Antibiotic-resistant bacterial pathogens are increasingly implicated in hospital- and community-acquired infections. Recent advances in monoclonal antibody (mAb) production and engineering have led to renewed interest in the development of antibody-based therapies for treatment of drug-resistant bacterial infections. Currently, there are three antibacterial mAb products approved by the Food and Drug Administration (FDA) and at least nine mAbs are in clinical trials. Antibacterial mAbs are typically developed to kill bacteria or to attenuate bacterial pathological activity through neutralization of bacterial toxins and virulence factors. Antibodies exhibit distinct pharmacological mechanisms from traditional antimicrobials and, hence, cross-resistance between small molecule antimicrobials and antibacterial mAbs is unlikely. Additionally, the long biological half-lives typically found for mAbs may allow convenient dosing and vaccine-like prophylaxis from infection. However, the high affinity of mAbs and the involvement of the host immune system in their pharmacological actions may lead to complex and nonlinear pharmacokinetics and pharmacodynamics. In this review, we summarize the pharmacokinetics and pharmacodynamics of the FDA-approved antibacterial mAbs and those are currently in clinical trials. Challenges in the development of antibacterial mAbs are also discussed.
Collapse
Affiliation(s)
- Shun Xin Wang-Lin
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA.
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA.
| |
Collapse
|
136
|
Trachsel L, Broguiere N, Rosenboom JG, Zenobi-Wong M, Benetti EM. Enzymatically crosslinked poly(2-alkyl-2-oxazoline) networks for 3D cell culture. J Mater Chem B 2018; 6:7568-7572. [DOI: 10.1039/c8tb02382d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cellularized poly(2-alkyl-2-oxazoline) hydrogels fabricated by sortase-mediated crosslinking feature tunable mechanical properties and enable extremely high cell viability.
Collapse
Affiliation(s)
- Lucca Trachsel
- Tissue Engineering + Biofabrication
- Department of Health Sciences and Technology
- ETH Zürich
- Zürich
- Switzerland
| | - Nicolas Broguiere
- Tissue Engineering + Biofabrication
- Department of Health Sciences and Technology
- ETH Zürich
- Zürich
- Switzerland
| | - Jan-Georg Rosenboom
- Institute of Chemical and Bioengineering
- Department of Chemistry and Applied Biosciences
- ETH Zürich
- Zürich
- Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication
- Department of Health Sciences and Technology
- ETH Zürich
- Zürich
- Switzerland
| | - Edmondo M. Benetti
- Polymer Surfaces Group
- Laboratory for Surface Science and Technology
- Department of Materials
- ETH Zürich
- Zürich
| |
Collapse
|
137
|
Schoonen L, van Hest JCM. Modification of CCMV Nanocages for Enzyme Encapsulation. Methods Mol Biol 2018; 1798:69-83. [PMID: 29868952 DOI: 10.1007/978-1-4939-7893-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In cellular systems, compartmentalization plays an important role in the protection and regulation of enzymes. Controlled encapsulation of enzymes in nanocompartments is crucial in understanding biocatalytic processes in the cellular environment. We have recently described an enzymatic method to covalently attach enzymes, equipped with a small recognition peptide, to the interior of viral capsids. Viral capsids are especially interesting in this respect, as they form very well-defined nanoparticles with a uniform size and shape. Here, we describe the relevant experimental procedures to encapsulate a model enzyme into the interior of a viral capsid, purify the resulting viral capsids, and measure the catalytic activity of the encapsulated enzymes.
Collapse
Affiliation(s)
- Lise Schoonen
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | | |
Collapse
|
138
|
Kane TL, Carothers KE, Lee SW. Virulence Factor Targeting of the Bacterial Pathogen Staphylococcus aureus for Vaccine and Therapeutics. Curr Drug Targets 2018; 19:111-127. [PMID: 27894236 PMCID: PMC5957279 DOI: 10.2174/1389450117666161128123536] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Staphylococcus aureus is a major bacterial pathogen capable of causing a range of infections in humans from gastrointestinal disease, skin and soft tissue infections, to severe outcomes such as sepsis. Staphylococcal infections in humans can be frequent and recurring, with treatments becoming less effective due to the growing persistence of antibiotic resistant S. aureus strains. Due to the prevalence of antibiotic resistance, and the current limitations on antibiotic development, an active and highly promising avenue of research has been to develop strategies to specifically inhibit the activity of virulence factors produced S. aureus as an alternative means to treat disease. OBJECTIVE In this review we specifically highlight several major virulence factors produced by S. aureus for which recent advances in antivirulence approaches may hold promise as an alternative means to treating diseases caused by this pathogen. Strategies to inhibit virulence factors can range from small molecule inhibitors, to antibodies, to mutant and toxoid forms of the virulence proteins. CONCLUSION The major prevalence of antibiotic resistant strains of S. aureus combined with the lack of new antibiotic discoveries highlight the need for vigorous research into alternative strategies to combat diseases caused by this highly successful pathogen. Current efforts to develop specific antivirulence strategies, vaccine approaches, and alternative therapies for treating severe disease caused by S. aureus have the potential to stem the tide against the limitations that we face in the post-antibiotic era.
Collapse
Affiliation(s)
- Trevor L. Kane
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Katelyn E. Carothers
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shaun W. Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
139
|
Nikghalb KD, Horvath NM, Prelesnik JL, Banks OGB, Filipov PA, Row RD, Roark TJ, Antos JM. Expanding the Scope of Sortase-Mediated Ligations by Using Sortase Homologues. Chembiochem 2017; 19:185-195. [PMID: 29124839 DOI: 10.1002/cbic.201700517] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Indexed: 02/04/2023]
Abstract
Sortase-catalyzed transacylation reactions are widely used for the construction of non-natural protein derivatives. However, the most commonly used enzyme for these strategies (sortase A from Staphylococcus aureus) is limited by its narrow substrate scope. To expand the range of substrates compatible with sortase-mediated reactions, we characterized the in vitro substrate preferences of eight sortase A homologues. From these studies, we identified sortase A enzymes that recognize multiple substrates that are unreactive toward sortase A from S. aureus. We further exploited the ability of sortase A from Streptococcus pneumoniae to recognize an LPATS substrate to perform a site-specific modification of the N-terminal serine residue in the naturally occurring antimicrobial peptide DCD-1L. Finally, we unexpectedly observed that certain substrates (LPATXG, X=Nle, Leu, Phe, Tyr) were susceptible to transacylation at alternative sites within the substrate motif, and sortase A from S. pneumoniae was capable of forming oligomers. Overall, this work provides a foundation for the further development of sortase enzymes for use in protein modification.
Collapse
Affiliation(s)
- Keyvan D Nikghalb
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - Nicholas M Horvath
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - Jesse L Prelesnik
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - Orion G B Banks
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - Pavel A Filipov
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - R David Row
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - Travis J Roark
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| | - John M Antos
- Department of Chemistry, Western Washington University, 516 High Street, Bellingham, WA, 98225, USA
| |
Collapse
|
140
|
Jeong HJ, Abhiraman GC, Story CM, Ingram JR, Dougan SK. Generation of Ca2+-independent sortase A mutants with enhanced activity for protein and cell surface labeling. PLoS One 2017; 12:e0189068. [PMID: 29200433 PMCID: PMC5714338 DOI: 10.1371/journal.pone.0189068] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/17/2017] [Indexed: 11/22/2022] Open
Abstract
Sortase A, a calcium-dependent transpeptidase derived from Staphylococcus aureus, is used in a broad range of applications, such as the conjugation of fluorescent dyes and other moieties to proteins or to the surface of eukaryotic cells. In vivo and cell-based applications of sortase have been somewhat limited by the large range of calcium concentrations, as well as by the often transient nature of protein-protein interactions in living systems. In order to use sortase A for cell labeling applications, we generated a new sortase A variant by combining multiple mutations to yield an enzyme that was both calcium-independent and highly active. This variant has enhanced activity for both N- and C-terminal labeling, as well as for cell surface modification under physiological conditions.
Collapse
Affiliation(s)
- Hee-Jin Jeong
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gita C. Abhiraman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Craig M. Story
- Department of Biology, Gordon College, Wenham, Massachusetts, United States of America
| | - Jessica R. Ingram
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephanie K. Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
141
|
Ming D, Wang D, Cao F, Xiang H, Mu D, Cao J, Li B, Zhong L, Dong X, Zhong X, Wang L, Wang T. Kaempferol Inhibits the Primary Attachment Phase of Biofilm Formation in Staphylococcus aureus. Front Microbiol 2017; 8:2263. [PMID: 29187848 PMCID: PMC5694784 DOI: 10.3389/fmicb.2017.02263] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/02/2017] [Indexed: 11/13/2022] Open
Abstract
The ability to form biofilms on surfaces makes Staphylococcus aureus the main pathogenic factor in implanted medical device infections. The aim of this study was to discover a biofilm inhibitor distinct from the antibiotics used to prevent infections resulting from S. aureus biofilms. Here, we describe kaempferol, a small molecule with anti-biofilm activity that specifically inhibited the formation of S. aureus biofilms. Crystal violet (CV) staining and fluorescence microscopy clearly showed that 64 μg/ml kaempferol inhibited biofilm formation by 80%. Meanwhile, the minimum inhibitory concentration (MIC) and growth curve results indicated that kaempferol had no antibacterial activity against the tested bacterial strain. Kaempferol inhibited the primary attachment phase of biofilm formation, as determined by a fibrinogen-binding assay. Moreover, a fluorescence resonance energy transfer (FRET) assay and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) analyses revealed that kaempferol reduced the activity of S. aureus sortaseA (SrtA) and the expression of adhesion-related genes. Based on these results, kaempferol provides a starting point for the development of novel anti-biofilm drugs, which may decrease the risk of bacterial drug resistance, to prevent S. aureus biofilm-related infections.
Collapse
Affiliation(s)
- Di Ming
- College of Animal Science, Jilin University, Changchun, China
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Fengjiao Cao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Hua Xiang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Dan Mu
- College of Animal Science, Jilin University, Changchun, China
| | - Junjie Cao
- College of Animal Science, Jilin University, Changchun, China
| | - Bangbang Li
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Ling Zhong
- College of Animal Science, Jilin University, Changchun, China
| | - Xiaoyun Dong
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Xiaobo Zhong
- College of Animal Science, Jilin University, Changchun, China
| | - Lin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tiedong Wang
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
142
|
Kadam RU, Juraszek J, Brandenburg B, Buyck C, Schepens WBG, Kesteleyn B, Stoops B, Vreeken RJ, Vermond J, Goutier W, Tang C, Vogels R, Friesen RHE, Goudsmit J, van Dongen MJP, Wilson IA. Potent peptidic fusion inhibitors of influenza virus. Science 2017; 358:496-502. [PMID: 28971971 DOI: 10.1126/science.aan0516] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 09/18/2017] [Indexed: 01/02/2023]
Abstract
Influenza therapeutics with new targets and mechanisms of action are urgently needed to combat potential pandemics, emerging viruses, and constantly mutating strains in circulation. We report here on the design and structural characterization of potent peptidic inhibitors of influenza hemagglutinin. The peptide design was based on complementarity-determining region loops of human broadly neutralizing antibodies against the hemagglutinin (FI6v3 and CR9114). The optimized peptides exhibit nanomolar affinity and neutralization against influenza A group 1 viruses, including the 2009 H1N1 pandemic and avian H5N1 strains. The peptide inhibitors bind to the highly conserved stem epitope and block the low pH-induced conformational rearrangements associated with membrane fusion. These peptidic compounds and their advantageous biological properties should accelerate the development of new small molecule- and peptide-based therapeutics against influenza virus.
Collapse
Affiliation(s)
- Rameshwar U Kadam
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | - Christophe Buyck
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium
| | - Wim B G Schepens
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium
| | - Bart Kesteleyn
- Janssen Infectious Diseases and Vaccines, Beerse, Belgium
| | - Bart Stoops
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium
| | - Rob J Vreeken
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium
| | - Jan Vermond
- Janssen Prevention Center, Leiden, Netherlands
| | | | - Chan Tang
- Janssen Prevention Center, Leiden, Netherlands
| | | | | | - Jaap Goudsmit
- Janssen Prevention Center, Leiden, Netherlands.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Maria J P van Dongen
- Janssen Prevention Center, Leiden, Netherlands. .,Discovery Sciences, Janssen Research & Development, Beerse, Belgium
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. .,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
143
|
Identification of Staphylococcus aureus Cellular Pathways Affected by the Stilbenoid Lead Drug SK-03-92 Using a Microarray. Antibiotics (Basel) 2017; 6:antibiotics6030017. [PMID: 28892020 PMCID: PMC5617981 DOI: 10.3390/antibiotics6030017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/25/2017] [Accepted: 09/07/2017] [Indexed: 12/16/2022] Open
Abstract
The mechanism of action for a new lead stilbene compound coded SK-03-92 with bactericidal activity against methicillin-resistant Staphylococcus aureus (MRSA) is unknown. To gain insight into the killing process, transcriptional profiling was performed on SK-03-92 treated vs. untreated S. aureus. Fourteen genes were upregulated and 38 genes downregulated by SK-03-92 treatment. Genes involved in sortase A production, protein metabolism, and transcriptional regulation were upregulated, whereas genes encoding transporters, purine synthesis proteins, and a putative two-component system (SACOL2360 (MW2284) and SACOL2361 (MW2285)) were downregulated by SK-03-92 treatment. Quantitative real-time polymerase chain reaction analyses validated upregulation of srtA and tdk as well as downregulation of the MW2284/MW2285 and purine biosynthesis genes in the drug-treated population. A quantitative real-time polymerase chain reaction analysis of MW2284 and MW2285 mutants compared to wild-type cells demonstrated that the srtA gene was upregulated by both putative two-component regulatory gene mutants compared to the wild-type strain. Using a transcription profiling technique, we have identified several cellular pathways regulated by SK-03-92 treatment, including a putative two-component system that may regulate srtA and other genes that could be tied to the SK-03-92 mechanism of action, biofilm formation, and drug persisters.
Collapse
|
144
|
Zhang B, Teng Z, Li X, Lu G, Deng X, Niu X, Wang J. Chalcone Attenuates Staphylococcus aureus Virulence by Targeting Sortase A and Alpha-Hemolysin. Front Microbiol 2017; 8:1715. [PMID: 28932220 PMCID: PMC5592744 DOI: 10.3389/fmicb.2017.01715] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus (S.aureus) resistance, considered a dilemma for the clinical treatment of this bacterial infection, is becoming increasingly intractable. Novel anti-virulence strategies will undoubtedly provide a path forward in combating these resistant bacterial infections. Sortase A (SrtA), an enzyme responsible for anchoring virulence-related surface proteins, and alpha-hemolysin (Hla), a pore-forming cytotoxin, have aroused great scientific interest, as they have been regarded as targets for promising agents against S. aureus infection. In this study, we discovered that chalcone, a natural small compound with little anti-S. aureus activity, could significantly inhibit SrtA activity with an IC50 of 53.15 μM and Hla hemolysis activity with an IC50 of 17.63 μM using a fluorescence resonance energy transfer (FRET) assay and a hemolysis assay, respectively. In addition, chalcone was proven to reduce protein A (SpA) display in intact bacteria, binding to fibronectin, formation of biofilm and S. aureus invasion. Chalcone could down-regulate the transcriptional levels of the hla gene and the agrA gene, thus leading to a reduction in the expression of Hla and significant protection against Hla-mediated A549 cell injury; more importantly, chalcone could also reduce mortality in infected mice. Additionally, molecular dynamics simulations and mutagenesis assays were used to identify the mechanism of chalcone against SrtA, which implied that the inhibitory activity lies in the bond between chalcone and SrtA residues Val168, Ile182, and Arg197. Taken together, the in vivo and in vitro experiments suggest that chalcone is a potential novel therapeutic compound for S. aureus infection via targeting SrtA and Hla.
Collapse
Affiliation(s)
- Bing Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China.,Center of Infection and Immunity, The First Hospital, Jilin UniversityChangchun, China
| | - Zihao Teng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Xianhe Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Gejin Lu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Xuming Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China.,Center of Infection and Immunity, The First Hospital, Jilin UniversityChangchun, China
| | - Xiaodi Niu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Jianfeng Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China.,Center of Infection and Immunity, The First Hospital, Jilin UniversityChangchun, China
| |
Collapse
|
145
|
Gau E, Mate DM, Zou Z, Oppermann A, Töpel A, Jakob F, Wöll D, Schwaneberg U, Pich A. Sortase-Mediated Surface Functionalization of Stimuli-Responsive Microgels. Biomacromolecules 2017; 18:2789-2798. [DOI: 10.1021/acs.biomac.7b00720] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Elisabeth Gau
- Functional
and Interactive Polymers, Institute of Technical and Macromolecular
Chemistry, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
| | - Diana M. Mate
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
| | - Zhi Zou
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
- Institute
for Biotechnology, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Alex Oppermann
- Institute
of Physical Chemistry, RWTH Aachen University, Landoltweg 2, 52074, Aachen, Germany
| | - Alexander Töpel
- Functional
and Interactive Polymers, Institute of Technical and Macromolecular
Chemistry, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
| | - Felix Jakob
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
| | - Dominik Wöll
- Institute
of Physical Chemistry, RWTH Aachen University, Landoltweg 2, 52074, Aachen, Germany
| | - Ulrich Schwaneberg
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
- Institute
for Biotechnology, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Andrij Pich
- Functional
and Interactive Polymers, Institute of Technical and Macromolecular
Chemistry, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
- DWI − Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074, Aachen, Germany
| |
Collapse
|
146
|
Anchoring of LPXTG-Like Proteins to the Gram-Positive Cell Wall Envelope. Curr Top Microbiol Immunol 2017; 404:159-175. [PMID: 27097813 DOI: 10.1007/82_2016_8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In Gram-positive bacteria, protein precursors with a signal peptide and a cell wall sorting signal (CWSS)-which begins with an LPXTG motif, followed by a hydrophobic domain and a tail of positively charged residues-are targeted to the cell envelope by a transpeptidase enzyme call sortase. Evolution and selective pressure gave rise to six classes of sortase, i.e., SrtA-F. Only class C sortases are capable of polymerizing substrates harboring the pilin motif and CWSS into protein polymers known as pili or fimbriae, whereas the others perform cell wall anchoring functions. Regardless of the products generated from these sortases, the basic principle of sortase-catalyzed transpeptidation is the same. It begins with the cleavage of the LPXTG motif, followed by the cross-linking of this cleaved product at the threonine residue to a nucleophile, i.e., an active amino group of the peptidoglycan stem peptide or the lysine residue of the pilin motif. This chapter will summarize the efforts to identify and characterize sortases and their associated pathways with emphasis on the cell wall anchoring function.
Collapse
|
147
|
Zhao X, Hong H, Cheng X, Liu S, Deng T, Guo Z, Wu Z. One-step purification and immobilization of extracellularly expressed sortase A by magnetic particles to develop a robust and recyclable biocatalyst. Sci Rep 2017; 7:6561. [PMID: 28747746 PMCID: PMC5529518 DOI: 10.1038/s41598-017-06856-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/19/2017] [Indexed: 12/21/2022] Open
Abstract
Sortase A (SrtA) is a transpeptidase widely used to site-specifically modify peptides and proteins and shows promise for industrial applications. In this study, a novel strategy was developed for constructing immobilized-SrtA as a robust and recyclable enzyme via direct immobilization of extracellularly expressed SrtA in the fermentation supernatant using magnetic particles. Efficient extracellular SrtA expression was achieved in Escherichia coli through molecular engineering, including manipulation of the protein transport pathway, codon optimization, and co-expression of molecular chaperones to promote expressed SrtA secretion into the medium at high levels. Subsequently, a simple one-step protocol was established for the purification and immobilization of SrtA containing a His-tag from the fermentation supernatant onto a nickel-modified magnetic particle. The immobilized SrtA was proved to retain full enzymatic activity for peptide-to-peptide ligation and protein modification, and was successfully reused for five cycles without obvious activity loss.
Collapse
Affiliation(s)
- Xinrui Zhao
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiaozhong Cheng
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Shaozhong Liu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Tao Deng
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida, 32611, United States of America
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
| |
Collapse
|
148
|
Innovative Solutions to Sticky Situations: Antiadhesive Strategies for Treating Bacterial Infections. Microbiol Spectr 2017; 4. [PMID: 27227305 DOI: 10.1128/microbiolspec.vmbf-0023-2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial adherence to host tissue is an essential process in pathogenesis, necessary for invasion and colonization and often required for the efficient delivery of toxins and other bacterial effectors. As existing treatment options for common bacterial infections dwindle, we find ourselves rapidly approaching a tipping point in our confrontation with antibiotic-resistant strains and in desperate need of new treatment options. Bacterial strains defective in adherence are typically avirulent and unable to cause infection in animal models. The importance of this initial binding event in the pathogenic cascade highlights its potential as a novel therapeutic target. This article seeks to highlight a variety of strategies being employed to treat and prevent infection by targeting the mechanisms of bacterial adhesion. Advancements in this area include the development of novel antivirulence therapies using small molecules, vaccines, and peptides to target a variety of bacterial infections. These therapies target bacterial adhesion through a number of mechanisms, including inhibition of pathogen receptor biogenesis, competition-based strategies with receptor and adhesin analogs, and the inhibition of binding through neutralizing antibodies. While this article is not an exhaustive description of every advancement in the field, we hope it will highlight several promising examples of the therapeutic potential of antiadhesive strategies.
Collapse
|
149
|
Schmohl L, Bierlmeier J, von Kügelgen N, Kurz L, Reis P, Barthels F, Mach P, Schutkowski M, Freund C, Schwarzer D. Identification of sortase substrates by specificity profiling. Bioorg Med Chem 2017; 25:5002-5007. [PMID: 28684010 DOI: 10.1016/j.bmc.2017.06.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 01/31/2023]
Abstract
Sortases catalyze the attachment of surface proteins to the peptidoglycan layer of gram-positive bacteria and further represent powerful tools of protein chemistry. During catalysis sortases cleave a donor substrate containing the LPxTG (x=any amino acid) sorting motif under formation of an enzyme-bound thioester and ligate this intermediate to an acceptor protein containing an N-terminal glycine residue. In addition to the well-established sortase A of Staphylococcus aureus several homologs of this enzyme have been identified in the genomes of gram-positive bacteria. We have profiled the specificity of seven sortases of Staphylococci and Streptococci origin and observed that sortases of the latter class displayed a more relaxed specificity for donor and acceptor substrates than their Staphylococci counterparts. Streptococci sortases prefer an LPKLG donor substrate sequence compared to the canonical sorting motif LPKTG. These findings might facilitate the use of Streptococci sortases as tools of protein chemistry.
Collapse
Affiliation(s)
- Lena Schmohl
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Jan Bierlmeier
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Nicolai von Kügelgen
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Leonie Kurz
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Pascal Reis
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Fabian Barthels
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Pia Mach
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Mike Schutkowski
- Institut für Biochemie and Biotechnologie, Martin-Luther-Universität Halle-Wittenberg, Kurt-Mothes-Str. 3, D-06120 Halle (Saale), Germany
| | - Christian Freund
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Thielallee 63, D-14195 Berlin, Germany
| | - Dirk Schwarzer
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany.
| |
Collapse
|
150
|
Electron Transport Chain Is Biochemically Linked to Pilus Assembly Required for Polymicrobial Interactions and Biofilm Formation in the Gram-Positive Actinobacterium Actinomyces oris. mBio 2017. [PMID: 28634238 PMCID: PMC5478893 DOI: 10.1128/mbio.00399-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The Gram-positive actinobacteria Actinomyces spp. are key colonizers in the development of oral biofilms due to the inherent ability of Actinomyces to adhere to receptor polysaccharides on the surface of oral streptococci and host cells. This receptor-dependent bacterial interaction, or coaggregation, requires a unique sortase-catalyzed pilus consisting of the pilus shaft FimA and the coaggregation factor CafA forming the pilus tip. While the essential role of the sortase machine SrtC2 in pilus assembly, biofilm formation, and coaggregation has been established, little is known about trans-acting factors contributing to these processes. We report here a large-scale Tn5 transposon screen for mutants defective in Actinomyces oris coaggregation with Streptococcus oralis. We obtained 33 independent clones, 13 of which completely failed to aggregate with S. oralis, and the remainder of which exhibited a range of phenotypes from severely to weakly defective coaggregation. The former had Tn5 insertions in fimA, cafA, or srtC2, as expected; the latter were mapped to genes coding for uncharacterized proteins and various nuo genes encoding the NADH dehydrogenase subunits. Electron microscopy and biochemical analyses of mutants with nonpolar deletions of nuo genes and ubiE, a menaquinone C-methyltransferase-encoding gene downstream of the nuo locus, confirmed the pilus and coaggregation defects. Both nuoA and ubiE mutants were defective in oxidation of MdbA, the major oxidoreductase required for oxidative folding of pilus proteins. Furthermore, supplementation of the ubiE mutant with exogenous menaquinone-4 rescued the cell growth and pilus defects. Altogether, we propose that the A. oris electron transport chain is biochemically linked to pilus assembly via oxidative protein folding. The Gram-positive actinobacterium A. oris expresses adhesive pili, or fimbriae, that are essential to biofilm formation and Actinomyces interactions with other bacteria, termed coaggregation. While the critical role of the conserved sortase machine in pilus assembly and the disulfide bond-forming catalyst MdbA in oxidative folding of pilins has been established, little is known about other trans-acting factors involved in these processes. Using a Tn5 transposon screen for mutants defective in coaggregation with Streptococcus oralis, we found that genetic disruption of the NADH dehydrogenase and menaquinone biosynthesis detrimentally alters pilus assembly. Further biochemical characterizations determined that menaquinone is important for reactivation of MdbA. This study supports the notion that the electron transport chain is biochemically linked to pilus assembly in A. oris via oxidative folding of pilin precursors.
Collapse
|