1
|
Martian PC, Tertis M, Leonte D, Hadade N, Cristea C, Crisan O. Cyclic peptides: A powerful instrument for advancing biomedical nanotechnologies and drug development. J Pharm Biomed Anal 2025; 252:116488. [PMID: 39388867 DOI: 10.1016/j.jpba.2024.116488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Cyclic peptides have emerged as an essential tool in the advancement of biomedical nanotechnologies, offering unique structural and functional advantages over linear peptides. This review article aims to highlight the roles of cyclic peptides in the development of biomedical fields, with a particular focus on their application in drug discovery and delivery. Cyclic peptides exhibit exceptional stability, bioavailability, and binding specificity, making them ideal candidates for therapeutic and diagnostic applications. We explore the synthesis and design strategies that enable the precise control of cyclic peptide structures, leading to enhanced performance in targeting specific cellular pathways. The article also highlights recent breakthroughs in the use of cyclic peptides for creating innovative drug delivery systems, including nanoparticle conjugates and peptide-drug conjugates, which have shown promise in improving the efficacy and safety profiles of existing traditional treatments. The integration of cyclic peptides into nanotechnological frameworks holds significant promise for addressing unmet medical needs, providing a foundation for future advancements in personalized medicine and targeted drug delivery.
Collapse
Affiliation(s)
- Paul Cristian Martian
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania
| | - Mihaela Tertis
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania
| | - Denisa Leonte
- Department of Organic Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 28 Victor Babes Street, Cluj-Napoca 400023, Romania
| | - Niculina Hadade
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babes Bolyai University, 11 Arany Janos Street, Cluj-Napoca 400028, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania.
| | - Ovidiu Crisan
- Department of Organic Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 28 Victor Babes Street, Cluj-Napoca 400023, Romania
| |
Collapse
|
2
|
Koo BM, Todor H, Sun J, van Gestel J, Hawkins JS, Hearne CC, Banta AB, Huang KC, Peters JM, Gross CA. Comprehensive double-mutant analysis of the Bacillus subtilis envelope using double-CRISPRi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.608006. [PMID: 39185233 PMCID: PMC11343205 DOI: 10.1101/2024.08.14.608006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Understanding bacterial gene function remains a major biological challenge. Double-mutant genetic interaction (GI) analysis addresses this challenge by uncovering the functional partners of targeted genes, allowing us to associate genes of unknown function with novel pathways and unravel connections between well-studied pathways, but is difficult to implement at the genome-scale. Here, we develop and use double-CRISPRi to systematically quantify genetic interactions at scale in the Bacillus subtilis envelope, including essential genes. We discover > 1000 known and novel genetic interactions. Our analysis pipeline and experimental follow-ups reveal the distinct roles of paralogous genes such as the mreB and mbl actin homologs, and identify new genes involved in the well-studied process of cell division. Overall, our study provides valuable insights into gene function and demonstrates the utility of double-CRISPRi for high-throughput dissection of bacterial gene networks, providing a blueprint for future studies in diverse bacterial species.
Collapse
Affiliation(s)
- Byoung-Mo Koo
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Horia Todor
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Jiawei Sun
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Jordi van Gestel
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - John S. Hawkins
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Cameron C. Hearne
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Amy B. Banta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jason M. Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Carol A. Gross
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
- California Institute of Quantitative Biology, University of California, San Francisco, San Francisco, CA, USA
- Lead Contact
| |
Collapse
|
3
|
Voitsekhovskaia I, Ho YTC, Klatt C, Müller A, Machell DL, Tan YJ, Triesman M, Bingel M, Schittenhelm RB, Tailhades J, Kulik A, Maier ME, Otting G, Wohlleben W, Schneider T, Cryle M, Stegmann E. Altering glycopeptide antibiotic biosynthesis through mutasynthesis allows incorporation of fluorinated phenylglycine residues. RSC Chem Biol 2024:d4cb00140k. [PMID: 39247680 PMCID: PMC11376024 DOI: 10.1039/d4cb00140k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/10/2024] [Indexed: 09/10/2024] Open
Abstract
Glycopeptide antibiotics (GPAs) are peptide natural products used as last resort treatments for antibiotic resistant bacterial infections. They are produced by the sequential activities of a linear nonribosomal peptide synthetase (NRPS), which assembles the heptapeptide core of GPAs, and cytochrome P450 (Oxy) enzymes, which perform a cascade of cyclisation reactions. The GPAs contain proteinogenic and nonproteinogenic amino acids, including phenylglycine residues such as 4-hydroxyphenylglycine (Hpg). The ability to incorporate non-proteinogenic amino acids in such peptides is a distinctive feature of the modular architecture of NRPSs, with each module selecting and incorporating a desired amino acid. Here, we have exploited this ability to produce and characterise GPA derivatives containing fluorinated phenylglycine (F-Phg) residues through a combination of mutasynthesis, biochemical, structural and bioactivity assays. Our data indicate that the incorporation of F-Phg residues is limited by poor acceptance by the NRPS machinery, and that the phenol moiety normally present on Hpg residues is essential to ensure both acceptance by the NRPS and the sequential cyclisation activity of Oxy enzymes. The principles learnt here may prove useful for the future production of GPA derivatives with more favourable properties through mixed feeding mutasynthesis approaches.
Collapse
Affiliation(s)
- Irina Voitsekhovskaia
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen Tübingen Germany
| | - Y T Candace Ho
- Department of Biochemistry and Molecular Biology, The Monash Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
- EMBL Australia, Monash University Clayton VIC 3800 Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
| | - Christoph Klatt
- Institute of Organic Chemistry, University of Tübingen Tübingen Germany
| | - Anna Müller
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn Bonn Germany
| | - Daniel L Machell
- Department of Biochemistry and Molecular Biology, The Monash Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
- EMBL Australia, Monash University Clayton VIC 3800 Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
| | - Yi Jiun Tan
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
- Research School of Chemistry, The Australian National University Acton ACT 2601 Australia
| | - Maxine Triesman
- Department of Biochemistry and Molecular Biology, The Monash Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
- EMBL Australia, Monash University Clayton VIC 3800 Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
| | - Mara Bingel
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn Bonn Germany
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash University Clayton VIC 3800 Australia
| | - Julien Tailhades
- Department of Biochemistry and Molecular Biology, The Monash Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
- EMBL Australia, Monash University Clayton VIC 3800 Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
| | - Andreas Kulik
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen Tübingen Germany
| | - Martin E Maier
- Institute of Organic Chemistry, University of Tübingen Tübingen Germany
| | - Gottfried Otting
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
- Research School of Chemistry, The Australian National University Acton ACT 2601 Australia
| | - Wolfgang Wohlleben
- Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen Tübingen Germany
| | - Tanja Schneider
- Institute of Organic Chemistry, University of Tübingen Tübingen Germany
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn Bonn Germany
| | - Max Cryle
- Department of Biochemistry and Molecular Biology, The Monash Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
- EMBL Australia, Monash University Clayton VIC 3800 Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
| | - Evi Stegmann
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen Tübingen Germany
- ARC Centre of Excellence for Innovations in Peptide and Protein Science Australia
- German Centre for Infection Research (DZIF), Partner Site Tübingen Tübingen Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections' (CMFI), University of Tübingen Tübingen Germany
| |
Collapse
|
4
|
van Groesen E, Mons E, Kotsogianni I, Arts M, Tehrani KHME, Wade N, Lysenko V, Stel FM, Zwerus JT, De Benedetti S, Bakker A, Chakraborty P, van der Stelt M, Scheffers DJ, Gooskens J, Smits WK, Holden K, Gilmour PS, Willemse J, Hitchcock CA, van Hasselt JGC, Schneider T, Martin NI. Semisynthetic guanidino lipoglycopeptides with potent in vitro and in vivo antibacterial activity. Sci Transl Med 2024; 16:eabo4736. [PMID: 39110780 DOI: 10.1126/scitranslmed.abo4736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/23/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024]
Abstract
Gram-positive bacterial infections present a major clinical challenge, with methicillin- and vancomycin-resistant strains continuing to be a cause for concern. In recent years, semisynthetic vancomycin derivatives have been developed to overcome this problem as exemplified by the clinically used telavancin, which exhibits increased antibacterial potency but has also raised toxicity concerns. Thus, glycopeptide antibiotics with enhanced antibacterial activities and improved safety profiles are still necessary. We describe the development of a class of highly potent semisynthetic glycopeptide antibiotics, the guanidino lipoglycopeptides, which contain a positively charged guanidino moiety bearing a variable lipid group. These glycopeptides exhibited enhanced in vitro activity against a panel of Gram-positive bacteria including clinically relevant methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant strains, showed minimal toxicity toward eukaryotic cells, and had a low propensity for resistance selection. Mechanistically, guanidino lipoglycopeptides engaged with bacterial cell wall precursor lipid II with a higher binding affinity than vancomycin. Binding to both wild-type d-Ala-d-Ala lipid II and the vancomycin-resistant d-Ala-d-Lac variant was confirmed, providing insight into the enhanced activity of guanidino lipoglycopeptides against vancomycin-resistant isolates. The in vivo efficacy of guanidino lipoglycopeptide EVG7 was evaluated in a S. aureus murine thigh infection model and a 7-day sepsis survival study, both of which demonstrated superiority to vancomycin. Moreover, the minimal to mild kidney effects at supratherapeutic doses of EVG7 indicate an improved therapeutic safety profile compared with vancomycin. These findings position guanidino lipoglycopeptides as candidates for further development as antibacterial agents for the treatment of clinically relevant multidrug-resistant Gram-positive infections.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Elma Mons
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Kamaleddin H M E Tehrani
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Nicola Wade
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Vladyslav Lysenko
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Florence M Stel
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Jordy T Zwerus
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Stefania De Benedetti
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Alexander Bakker
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Parichita Chakraborty
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Dirk-Jan Scheffers
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Jairo Gooskens
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Wiep Klaas Smits
- Experimental Bacteriology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Kirsty Holden
- Evotec (U.K.) Ltd., Alderley Park, Macclesfield, Cheshire, SK10 4TG UK
| | | | - Joost Willemse
- Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | | | - J G Coen van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, Netherlands
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| |
Collapse
|
5
|
George NL, Bennett EC, Orlando BJ. Guarding the walls: the multifaceted roles of Bce modules in cell envelope stress sensing and antimicrobial resistance. J Bacteriol 2024; 206:e0012324. [PMID: 38869304 PMCID: PMC11270860 DOI: 10.1128/jb.00123-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Bacteria have developed diverse strategies for defending their cell envelopes from external threats. In Firmicutes, one widespread strategy is to use Bce modules-membrane protein complexes that unite a peptide-detoxifying ABC transporter with a stress response coordinating two-component system. These modules provide specific, front-line defense for a wide variety of antimicrobial peptides and small molecule antibiotics as well as coordinate responses for heat, acid, and oxidative stress. Because of these abilities, Bce modules play important roles in virulence and the development of antibiotic resistance in a variety of pathogens, including Staphylococcus, Streptococcus, and Enterococcus species. Despite their importance, Bce modules are still poorly understood, with scattered functional data in only a small number of species. In this review, we will discuss Bce module structure in light of recent cryo-electron microscopy structures of the B. subtilis BceABRS module and explore the common threads and variations-on-a-theme in Bce module mechanisms across species. We also highlight the many remaining questions about Bce module function. Understanding these multifunctional membrane complexes will enhance our understanding of bacterial stress sensing and may point toward new therapeutic targets for highly resistant pathogens.
Collapse
Affiliation(s)
- Natasha L. George
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan, USA
| | - Ellen C. Bennett
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan, USA
| | - Benjamin J. Orlando
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
6
|
Buijs NP, Vlaming HC, Kotsogianni I, Arts M, Willemse J, Duan Y, Alexander FM, Cochrane SA, Schneider T, Martin NI. A classic antibiotic reimagined: Rationally designed bacitracin variants exhibit potent activity against vancomycin-resistant pathogens. Proc Natl Acad Sci U S A 2024; 121:e2315310121. [PMID: 38990944 PMCID: PMC11260088 DOI: 10.1073/pnas.2315310121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Bacitracin is a macrocyclic peptide antibiotic that is widely used as a topical treatment for infections caused by gram-positive bacteria. Mechanistically, bacitracin targets bacteria by specifically binding to the phospholipid undecaprenyl pyrophosphate (C55PP), which plays a key role in the bacterial lipid II cycle. Recent crystallographic studies have shown that when bound to C55PP, bacitracin adopts a highly ordered amphipathic conformation. In doing so, all hydrophobic side chains align on one face of the bacitracin-C55PP complex, presumably interacting with the bacterial cell membrane. These insights led us to undertake structure-activity investigations into the individual contribution of the nonpolar amino acids found in bacitracin. To achieve this we designed, synthesized, and evaluated a series of bacitracin analogues, a number of which were found to exhibit significantly enhanced antibacterial activity against clinically relevant, drug-resistant pathogens. As for the natural product, these next-generation bacitracins were found to form stable complexes with C55PP. The structure-activity insights thus obtained serve to inform the design of C55PP-targeting antibiotics, a key and underexploited antibacterial strategy.
Collapse
Affiliation(s)
- Ned P. Buijs
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden2333 BE, The Netherlands
| | - Halana C. Vlaming
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden2333 BE, The Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden2333 BE, The Netherlands
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn53115, Germany
| | - Joost Willemse
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden2333 BE, The Netherlands
| | - Yunhao Duan
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden2333 BE, The Netherlands
| | - Francesca M. Alexander
- School of Chemistry and Chemical Engineering, Queen’s University, BelfastBT9 5AG, United Kingdom
| | - Stephen A. Cochrane
- School of Chemistry and Chemical Engineering, Queen’s University, BelfastBT9 5AG, United Kingdom
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn53115, Germany
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden2333 BE, The Netherlands
| |
Collapse
|
7
|
Morris SM, Wiens L, Rose O, Fritz G, Rogers T, Gebhard S. Regulatory interactions between daptomycin- and bacitracin-responsive pathways coordinate the cell envelope antibiotic resistance response of Enterococcus faecalis. Mol Microbiol 2024; 121:1148-1163. [PMID: 38646792 DOI: 10.1111/mmi.15264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/23/2024]
Abstract
Enterococcal infections frequently show high levels of antibiotic resistance, including to cell envelope-acting antibiotics like daptomycin (DAP). While we have a good understanding of the resistance mechanisms, less is known about the control of such resistance genes in enterococci. Previous work unveiled a bacitracin resistance network, comprised of the sensory ABC transporter SapAB, the two-component system (TCS) SapRS and the resistance ABC transporter RapAB. Interestingly, components of this system have recently been implicated in DAP resistance, a role usually regulated by the TCS LiaFSR. To better understand the regulation of DAP resistance and how this relates to mutations observed in DAP-resistant clinical isolates of enterococci, we here explored the interplay between these two regulatory pathways. Our results show that SapR regulates an additional resistance operon, dltXABCD, a known DAP resistance determinant, and show that LiaFSR regulates the expression of sapRS. This regulatory structure places SapRS-target genes under dual control, where expression is directly controlled by SapRS, which itself is up-regulated through LiaFSR. The network structure described here shows how Enterococcus faecalis coordinates its response to cell envelope attack and can explain why clinical DAP resistance often emerges via mutations in regulatory components.
Collapse
Affiliation(s)
- Sali M Morris
- Life Sciences Department, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Laura Wiens
- Institute of Molecular Physiology, Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Olivia Rose
- Life Sciences Department, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Georg Fritz
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Tim Rogers
- Department of Mathematical Sciences, University of Bath, Bath, UK
| | - Susanne Gebhard
- Life Sciences Department, Milner Centre for Evolution, University of Bath, Bath, UK
- Institute of Molecular Physiology, Johannes-Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
8
|
Willdigg JR, Patel Y, Arquilevich BE, Subramanian C, Frank MW, Rock CO, Helmann JD. The Bacillus subtilis cell envelope stress-inducible ytpAB operon modulates membrane properties and contributes to bacitracin resistance. J Bacteriol 2024; 206:e0001524. [PMID: 38323910 PMCID: PMC10955860 DOI: 10.1128/jb.00015-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/08/2024] Open
Abstract
Antibiotics that inhibit peptidoglycan synthesis trigger the activation of both specific and general protective responses. σM responds to diverse antibiotics that inhibit cell wall synthesis. Here, we demonstrate that cell wall-inhibiting drugs, such as bacitracin and cefuroxime, induce the σM-dependent ytpAB operon. YtpA is a predicted hydrolase previously proposed to generate the putative lysophospholipid antibiotic bacilysocin (lysophosphatidylglycerol), and YtpB is the branchpoint enzyme for the synthesis of membrane-localized C35 terpenoids. Using targeted lipidomics, we reveal that YtpA is not required for the production of lysophosphatidylglycerol. Nevertheless, ytpA was critical for growth in a mutant strain defective for homeoviscous adaptation due to a lack of genes for the synthesis of branched chain fatty acids and the Des phospholipid desaturase. Consistently, overexpression of ytpA increased membrane fluidity as monitored by fluorescence anisotropy. The ytpA gene contributes to bacitracin resistance in mutants additionally lacking the bceAB or bcrC genes, which directly mediate bacitracin resistance. These epistatic interactions support a model in which σM-dependent induction of the ytpAB operon helps cells tolerate bacitracin stress, either by facilitating the flipping of the undecaprenyl phosphate carrier lipid or by impacting the assembly or function of membrane-associated complexes involved in cell wall homeostasis.IMPORTANCEPeptidoglycan synthesis inhibitors include some of our most important antibiotics. In Bacillus subtilis, peptidoglycan synthesis inhibitors induce the σM regulon, which is critical for intrinsic antibiotic resistance. The σM-dependent ytpAB operon encodes a predicted hydrolase (YtpA) and the enzyme that initiates the synthesis of C35 terpenoids (YtpB). Our results suggest that YtpA is critical in cells defective in homeoviscous adaptation. Furthermore, we find that YtpA functions cooperatively with the BceAB and BcrC proteins in conferring intrinsic resistance to bacitracin, a peptide antibiotic that binds tightly to the undecaprenyl-pyrophosphate lipid carrier that sustains peptidoglycan synthesis.
Collapse
Affiliation(s)
| | - Yesha Patel
- Department of Microbiology, Cornell University, Ithaca, New York, USA
| | | | - Chitra Subramanian
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Matthew W. Frank
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Charles O. Rock
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - John D. Helmann
- Department of Microbiology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
9
|
Kubiak X, Polsinelli I, Chavas LMG, Fyfe CD, Guillot A, Fradale L, Brewee C, Grimaldi S, Gerbaud G, Thureau A, Legrand P, Berteau O, Benjdia A. Structural and mechanistic basis for RiPP epimerization by a radical SAM enzyme. Nat Chem Biol 2024; 20:382-391. [PMID: 38158457 DOI: 10.1038/s41589-023-01493-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/30/2023] [Indexed: 01/03/2024]
Abstract
D-Amino acid residues, found in countless peptides and natural products including ribosomally synthesized and post-translationally modified peptides (RiPPs), are critical for the bioactivity of several antibiotics and toxins. Recently, radical S-adenosyl-L-methionine (SAM) enzymes have emerged as the only biocatalysts capable of installing direct and irreversible epimerization in RiPPs. However, the mechanism underpinning this biochemical process is ill-understood and the structural basis for this post-translational modification remains unknown. Here we report an atomic-resolution crystal structure of a RiPP-modifying radical SAM enzyme in complex with its substrate properly positioned in the active site. Crystallographic snapshots, size-exclusion chromatography-small-angle x-ray scattering, electron paramagnetic resonance spectroscopy and biochemical analyses reveal how epimerizations are installed in RiPPs and support an unprecedented enzyme mechanism for peptide epimerization. Collectively, our study brings unique perspectives on how radical SAM enzymes interact with RiPPs and catalyze post-translational modifications in natural products.
Collapse
Affiliation(s)
- Xavier Kubiak
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | - Ivan Polsinelli
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | | | - Cameron D Fyfe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | - Alain Guillot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | - Laura Fradale
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | - Clémence Brewee
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | | | | | - Aurélien Thureau
- Synchrotron SOLEIL, HelioBio Group, L'Orme des Merisiers, Saint-Aubin, France
| | - Pierre Legrand
- Synchrotron SOLEIL, HelioBio Group, L'Orme des Merisiers, Saint-Aubin, France
| | - Olivier Berteau
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France.
| | - Alhosna Benjdia
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France.
| |
Collapse
|
10
|
Rosenzweig AF, Wang Z, Morales-Amador A, Spotton K, Brady SF. A Family of Antibiotics That Evades Resistance by Binding Polyprenyl Phosphates. ACS Infect Dis 2023; 9:2394-2400. [PMID: 37937847 PMCID: PMC10904333 DOI: 10.1021/acsinfecdis.3c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Cilagicin is a Gram-positive active antibiotic that has a dual polyprenyl phosphate binding mechanism that impedes resistance development. Here we bioinformatically screened predicted non-ribosomal polypeptide synthetase encoded structures to search for antibiotics that might similarly avoid resistance development. Synthesis and bioactivity screening of the predicted structures that we identified led to three antibiotics that are active against multidrug-resistant Gram-positive pathogens, two of which, paenilagicin and virgilagicin, did not lead to resistance even after prolonged antibiotic exposure.
Collapse
Affiliation(s)
- Adam F Rosenzweig
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Zongqiang Wang
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Adrián Morales-Amador
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Kaylyn Spotton
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| |
Collapse
|
11
|
Comparison of Phenotype and Genotype Virulence and Antimicrobial Factors of Salmonella Typhimurium Isolated from Human Milk. Int J Mol Sci 2023; 24:ijms24065135. [PMID: 36982209 PMCID: PMC10048834 DOI: 10.3390/ijms24065135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023] Open
Abstract
Salmonella is a common foodborne infection. Many serovars belonging to Salmonella enterica subsp. enterica are present in the gut of various animal species. They can cause infection in human infants via breast milk or cross-contamination with powdered milk. In the present study, Salmonella BO was isolated from human milk in accordance with ISO 6579-1:2017 standards and sequenced using whole-genome sequencing (WGS), followed by serosequencing and genotyping. The results also allowed its pathogenicity to be predicted. The WGS results were compared with the bacterial phenotype. The isolated strain was found to be Salmonella enterica subsp. enterica serovar Typhimurium 4:i:1,2_69M (S. Typhimurium 69M); it showed a very close similarity to S. enterica subsp. enterica serovar Typhimurium LT2. Bioinformatics sequence analysis detected eleven SPIs (SPI-1, SPI-2, SPI-3, SPI-4, SPI-5, SPI-9, SPI-12, SPI-13, SPI-14, C63PI, CS54_island). Significant changes in gene sequences were noted, causing frameshift mutations in yeiG, rfbP, fumA, yeaL, ybeU (insertion) and lpfD, avrA, ratB, yacH (deletion). The sequences of several proteins were significantly different from those coded in the reference genome; their three-dimensional structure was predicted and compared with reference proteins. Our findings indicate the presence of a number of antimicrobial resistance genes that do not directly imply an antibiotic resistance phenotype.
Collapse
|
12
|
Popp PF, Lozano-Cruz T, Dürr F, Londaitsbehere A, Hartig J, de la Mata FJ, Gómez R, Mascher T, Revilla-Guarinos A. The Novel Synthetic Antibiotic BDTL049 Based on a Dendritic System Induces Lipid Domain Formation while Escaping the Cell Envelope Stress Resistance Determinants. Pharmaceutics 2023; 15:297. [PMID: 36678925 PMCID: PMC9866484 DOI: 10.3390/pharmaceutics15010297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
The threat of antimicrobial-resistant bacteria is ever increasing and over the past-decades development of novel therapeutic counter measurements have virtually come to a halt. This circumstance calls for interdisciplinary approaches to design, evaluate and validate the mode of action of novel antibacterial compounds. Hereby, carbosilane dendritic systems that exhibit antimicrobial properties have the potential to serve as synthetic and rationally designed molecules for therapeutic use. The bow-tie type topology of BDTL049 was recently investigated against the Gram-positive model organism Bacillus subtilis, revealing strong bactericidal properties. In this study, we follow up on open questions concerning the usability of BDTL049. For this, we synthesized a fluorescent-labeled version of BDTL049 that maintained all antimicrobial features to unravel the interaction of the compound and bacterial membrane. Subsequently, we highlight the bacterial sensitivity against BDTL049 by performing a mutational study of known resistance determinants. Finally, we address the cytotoxicity of the compound in human cells, unexpectedly revealing a high sensitivity of the eukaryotic cells upon BDTL049 exposure. The insights presented here further elaborate on the unique features of BDTL049 as a promising candidate as an antimicrobial agent while not precluding that further rounds of rational designing are needed to decrease cytotoxicity to ultimately pave the way for synthetic antibiotics toward clinical applicability.
Collapse
Affiliation(s)
- Philipp F. Popp
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Tania Lozano-Cruz
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, 28805 Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), 28805 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28805 Madrid, Spain
| | - Franziska Dürr
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Addis Londaitsbehere
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, 28805 Madrid, Spain
| | - Johanna Hartig
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Francisco Javier de la Mata
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, 28805 Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), 28805 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28805 Madrid, Spain
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, 28805 Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), 28805 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28805 Madrid, Spain
| | - Thorsten Mascher
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Ainhoa Revilla-Guarinos
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| |
Collapse
|
13
|
Revilla-Guarinos A, Popp PF, Dürr F, Lozano-Cruz T, Hartig J, de la Mata FJ, Gómez R, Mascher T. Synthesis and mechanism-of-action of a novel synthetic antibiotic based on a dendritic system with bow-tie topology. Front Microbiol 2022; 13:912536. [PMID: 36090105 PMCID: PMC9459136 DOI: 10.3389/fmicb.2022.912536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/26/2022] [Indexed: 12/05/2022] Open
Abstract
Over the course of the last decades, the continuous exposure of bacteria to antibiotics-at least in parts due to misprescription, misuse, and misdosing-has led to the widespread development of antimicrobial resistances. This development poses a threat to the available medication in losing their effectiveness in treating bacterial infections. On the drug development side, only minor advances have been made to bring forward novel therapeutics. In addition to increasing the efforts and approaches of tapping the natural sources of new antibiotics, synthetic approaches to developing novel antimicrobials are being pursued. In this study, BDTL049 was rationally designed using knowledge based on the properties of natural antibiotics. BDTL049 is a carbosilane dendritic system with bow-tie type topology, which has antimicrobial activity at concentrations comparable to clinically established natural antibiotics. In this report, we describe its mechanism of action on the Gram-positive model organism Bacillus subtilis. Exposure to BDTL049 resulted in a complex transcriptional response, which pointed toward disturbance of the cell envelope homeostasis accompanied by disruption of other central cellular processes of bacterial metabolism as the primary targets of BDTL049 treatment. By applying a combination of whole-cell biosensors, molecular staining, and voltage sensitive dyes, we demonstrate that the mode of action of BDTL049 comprises membrane depolarization concomitant with pore formation. As a result, this new molecule kills Gram-positive bacteria within minutes. Since BDTL049 attacks bacterial cells at different targets simultaneously, this might decrease the chances for the development of bacterial resistances, thereby making it a promising candidate for a future antimicrobial agent.
Collapse
Affiliation(s)
- Ainhoa Revilla-Guarinos
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Philipp F. Popp
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Franziska Dürr
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Tania Lozano-Cruz
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Johanna Hartig
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Francisco Javier de la Mata
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry “Andrés M. Del Río” (IQAR), University de Alcalá, Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Thorsten Mascher
- Department of General Microbiology, Institut Für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
14
|
Mendes SS, Marques J, Mesterházy E, Straetener J, Arts M, Pissarro T, Reginold J, Berscheid A, Bornikoel J, Kluj RM, Mayer C, Oesterhelt F, Friães S, Royo B, Schneider T, Brötz-Oesterhelt H, Romão CC, Saraiva LM. Synergetic Antimicrobial Activity and Mechanism of Clotrimazole-Linked CO-Releasing Molecules. ACS BIO & MED CHEM AU 2022; 2:419-436. [PMID: 35996473 PMCID: PMC9389576 DOI: 10.1021/acsbiomedchemau.2c00007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Several metal-based
carbon monoxide-releasing molecules (CORMs)
are active CO donors with established antibacterial activity. Among
them, CORM conjugates with azole antibiotics of type [Mn(CO)3(2,2′-bipyridyl)(azole)]+ display important synergies
against several microbes. We carried out a structure–activity
relationship study based upon the lead structure of [Mn(CO)3(Bpy)(Ctz)]+ by producing clotrimazole (Ctz) conjugates
with varying metal and ligands. We concluded that the nature of the
bidentate ligand strongly influences the bactericidal activity, with
the substitution of bipyridyl by small bicyclic ligands leading to
highly active clotrimazole conjugates. On the contrary, the metal
did not influence the activity. We found that conjugate [Re(CO)3(Bpy)(Ctz)]+ is more than the sum of its parts:
while precursor [Re(CO)3(Bpy)Br] has no antibacterial activity
and clotrimazole shows only moderate minimal inhibitory concentrations,
the potency of [Re(CO)3(Bpy)(Ctz)]+ is one order
of magnitude higher than that of clotrimazole, and the spectrum of
bacterial target species includes Gram-positive and Gram-negative
bacteria. The addition of [Re(CO)3(Bpy)(Ctz)]+ to Staphylococcus aureus causes a
general impact on the membrane topology, has inhibitory effects on
peptidoglycan biosynthesis, and affects energy functions. The mechanism
of action of this kind of CORM conjugates involves a sequence of events
initiated by membrane insertion, followed by membrane disorganization,
inhibition of peptidoglycan synthesis, CO release, and break down
of the membrane potential. These results suggest that conjugation
of CORMs to known antibiotics may produce useful structures with synergistic
effects that increase the conjugate’s activity relative to
that of the antibiotic alone.
Collapse
Affiliation(s)
- Sofia S Mendes
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Joana Marques
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Edit Mesterházy
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Jan Straetener
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Teresa Pissarro
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Jorgina Reginold
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Anne Berscheid
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Jan Bornikoel
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Robert M Kluj
- Institute of Microbiology and Infection Medicine, Dept. of Organismic Interactions, University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Christoph Mayer
- Institute of Microbiology and Infection Medicine, Dept. of Organismic Interactions, University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Filipp Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Sofia Friães
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Beatriz Royo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Carlos C Romão
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Lígia M Saraiva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| |
Collapse
|
15
|
Wang Z, Koirala B, Hernandez Y, Zimmerman M, Brady SF. Bioinformatic prospecting and synthesis of a bifunctional lipopeptide antibiotic that evades resistance. Science 2022; 376:991-996. [PMID: 35617397 PMCID: PMC10904332 DOI: 10.1126/science.abn4213] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Emerging resistance to currently used antibiotics is a global public health crisis. Because most of the biosynthetic capacity within the bacterial kingdom has remained silent in previous antibiotic discovery efforts, uncharacterized biosynthetic gene clusters found in bacterial genome-sequencing studies remain an appealing source of antibiotics with distinctive modes of action. Here, we report the discovery of a naturally inspired lipopeptide antibiotic called cilagicin, which we chemically synthesized on the basis of a detailed bioinformatic analysis of the cil biosynthetic gene cluster. Cilagicin's ability to sequester two distinct, indispensable undecaprenyl phosphates used in cell wall biosynthesis, together with the absence of detectable resistance in laboratory tests and among multidrug-resistant clinical isolates, makes it an appealing candidate for combating antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Zongqiang Wang
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY 10065, USA
| | - Bimal Koirala
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY 10065, USA
| | - Yozen Hernandez
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY 10065, USA
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
16
|
Phyletic Distribution and Diversification of the Phage Shock Protein Stress Response System in Bacteria and Archaea. mSystems 2022; 7:e0134821. [PMID: 35604119 PMCID: PMC9239133 DOI: 10.1128/msystems.01348-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The PspA protein domain is found in all domains of life, highlighting its central role in Psp networks. To date, all insights into the core functions of Psp responses derive mainly from protein network blueprints representing only three bacterial phyla.
Collapse
|
17
|
Diagne AM, Pelletier A, Durmort C, Faure A, Kanonenberg K, Freton C, Page A, Delolme F, Vorac J, Vallet S, Bellard L, Vivès C, Fieschi F, Vernet T, Rousselle P, Guiral S, Grangeasse C, Jault JM, Orelle C. Identification of a two-component regulatory system involved in antimicrobial peptide resistance in Streptococcus pneumoniae. PLoS Pathog 2022; 18:e1010458. [PMID: 35395062 PMCID: PMC9020739 DOI: 10.1371/journal.ppat.1010458] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/20/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Two-component regulatory systems (TCS) are among the most widespread mechanisms that bacteria use to sense and respond to environmental changes. In the human pathogen Streptococcus pneumoniae, a total of 13 TCS have been identified and many of them have been linked to pathogenicity. Notably, TCS01 strongly contributes to pneumococcal virulence in several infection models. However, it remains one of the least studied TCS in pneumococci and its functional role is still unclear. In this study, we demonstrate that TCS01 cooperates with a BceAB-type ABC transporter to sense and induce resistance to structurally-unrelated antimicrobial peptides of bacterial origin that all target undecaprenyl-pyrophosphate or lipid II, which are essential precursors of cell wall biosynthesis. Even though tcs01 and bceAB genes do not locate in the same gene cluster, disruption of either of them equally sensitized the bacterium to the same set of antimicrobial peptides. We show that the key function of TCS01 is to upregulate the expression of the transporter, while the latter appears the main actor in resistance. Electrophoretic mobility shift assays further demonstrated that the response regulator of TCS01 binds to the promoter region of the bceAB genes, implying a direct control of these genes. The BceAB transporter was overexpressed and purified from E. coli. After reconstitution in liposomes, it displayed substantial ATPase and GTPase activities that were stimulated by antimicrobial peptides to which it confers resistance to, revealing new functional features of a BceAB-type transporter. Altogether, this inducible defense mechanism likely contributes to the survival of the opportunistic microorganism in the human host, in which competition among commensal microorganisms is a key determinant for effective host colonization and invasive path.
Collapse
Affiliation(s)
- Aissatou Maty Diagne
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Anaïs Pelletier
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Claire Durmort
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Agathe Faure
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Kerstin Kanonenberg
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Céline Freton
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Adeline Page
- Protein Science Facility, SFR BioSciences, CNRS, UMS3444, INSERM US8, University of Lyon, Lyon, France
| | - Frédéric Delolme
- Protein Science Facility, SFR BioSciences, CNRS, UMS3444, INSERM US8, University of Lyon, Lyon, France
| | - Jaroslav Vorac
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Sylvain Vallet
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Laure Bellard
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Corinne Vivès
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Franck Fieschi
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Thierry Vernet
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR 5305 CNRS/University of Lyon, Lyon, France
| | - Sébastien Guiral
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Christophe Grangeasse
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Jean-Michel Jault
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Cédric Orelle
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| |
Collapse
|
18
|
Gottstein J, Zaschke-Kriesche J, Unsleber S, Voitsekhovskaia I, Kulik A, Behrmann LV, Overbeck N, Stühler K, Stegmann E, Smits SHJ. New insights into the resistance mechanism for the BceAB-type transporter SaNsrFP. Sci Rep 2022; 12:4232. [PMID: 35273305 PMCID: PMC8913810 DOI: 10.1038/s41598-022-08095-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
Treatment of bacterial infections is one of the major challenges of our time due to the evolved resistance mechanisms of pathogens against antibiotics. To circumvent this problem, it is necessary to understand the mode of action of the drug and the mechanism of resistance of the pathogen. One of the most potent antibiotic targets is peptidoglycan (PGN) biosynthesis, as this is an exclusively occurring and critical feature of bacteria. Lipid II is an essential PGN precursor synthesized in the cytosol and flipped into the outer leaflet of the membrane prior to its incorporation into nascent PGN. Antimicrobial peptides (AMPs), such as nisin and colistin, targeting PGN synthesis are considered promising weapons against multidrug-resistant bacteria. However, human pathogenic bacteria that were also resistant to these compounds evolved by the expression of an ATP-binding cassette transporter of the bacitracin efflux (BceAB) type localized in the membrane. In the human pathogen Streptococcus agalactiae, the BceAB transporter SaNsrFP is known to confer resistance to the antimicrobial peptide nisin. The exact mechanism of action for SaNsrFP is poorly understood. For a detailed characterization of the resistance mechanism, we heterologously expressed SaNsrFP in Lactococcus lactis. We demonstrated that SaNsrFP conferred resistance not only to nisin but also to a structurally diverse group of antimicrobial PGN-targeting compounds such as ramoplanin, lysobactin, or bacitracin/(Zn)-bacitracin. Growth experiments revealed that SaNsrFP-producing cells exhibited normal behavior when treated with nisin and/or bacitracin, in contrast to the nonproducing cells, for which growth was significantly reduced. We further detected the accumulation of PGN precursors in the cytoplasm after treating the cells with bacitracin. This did not appear when SaNsrFP was produced. Whole-cell proteomic protein experiments verified that the presence of SaNsrFP in L. lactis resulted in higher production of several proteins associated with cell wall modification. These included, for example, the N-acetylmuramic acid-6-phosphate etherase MurQ and UDP-glucose 4-epimerase. Analysis of components of the cell wall of SaNsrFP-producing cells implied that the transporter is involved in cell wall modification. Since we used an ATP-deficient mutant of the transporter as a comparison, we can show that SaNsrFP and its inactive mutant do not show the same phenotype, albeit expressed at similar levels, which demonstrates the ATP dependency of the mediated resistance processes. Taken together, our data agree to a target protection mechanism and imply a direct involvement of SaNsrFP in resistance by shielding the membrane-localized target of these antimicrobial peptides, resulting in modification of the cell wall.
Collapse
Affiliation(s)
- Julia Gottstein
- Institute of Biochemistry, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Julia Zaschke-Kriesche
- Institute of Biochemistry, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Sandra Unsleber
- Interfaculty Institute of Microbiology and Infection Medicin, Eberhard Karls University, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Irina Voitsekhovskaia
- Interfaculty Institute of Microbiology and Infection Medicin, Eberhard Karls University, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Andreas Kulik
- Interfaculty Institute of Microbiology and Infection Medicin, Eberhard Karls University, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Lara V Behrmann
- Institute of Biochemistry, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Nina Overbeck
- Molecular Proteomics Laboratory, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Evi Stegmann
- Interfaculty Institute of Microbiology and Infection Medicin, Eberhard Karls University, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Sander H J Smits
- Institute of Biochemistry, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Duesseldorf, Germany.
| |
Collapse
|
19
|
Hao W, Cui W, Cheng Z, Han L, Suo F, Liu Z, Zhou L, Zhou Z. Development of a base editor for protein evolution via in situ mutation in vivo. Nucleic Acids Res 2021; 49:9594-9605. [PMID: 34390349 PMCID: PMC8450078 DOI: 10.1093/nar/gkab673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/16/2022] Open
Abstract
Protein evolution has significantly enhanced the development of life science. However, it is difficult to achieve in vitro evolution of some special proteins because of difficulties with heterologous expression, purification, and function detection. To achieve protein evolution via in situ mutation in vivo, we developed a base editor by fusing nCas with a cytidine deaminase in Bacillus subtilis through genome integration. The base editor introduced a cytidine-to-thymidine mutation of approximately 100% across a 5 nt editable window, which was much higher than those of other base editors. The editable window was expanded to 8 nt by extending the length of sgRNA, and conversion efficiency could be regulated by changing culture conditions, which was suitable for constructing a mutant protein library efficiently in vivo. As proof-of-concept, the Sec-translocase complex and bacitracin-resistance-related protein BceB were successfully evolved in vivo using the base editor. A Sec mutant with 3.6-fold translocation efficiency and the BceB mutants with different sensitivity to bacitracin were obtained. As the construction of the base editor does not rely on any additional or host-dependent factors, such base editors (BEs) may be readily constructed and applicable to a wide range of bacteria for protein evolution via in situ mutation.
Collapse
Affiliation(s)
- Wenliang Hao
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Wenjing Cui
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Zhongyi Cheng
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Laichuang Han
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Feiya Suo
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Zhongmei Liu
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Li Zhou
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Zhemin Zhou
- The Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| |
Collapse
|
20
|
Popp PF, Friebel L, Benjdia A, Guillot A, Berteau O, Mascher T. The Epipeptide Biosynthesis Locus epeXEPAB Is Widely Distributed in Firmicutes and Triggers Intrinsic Cell Envelope Stress. Microb Physiol 2021; 31:306-318. [PMID: 34120110 DOI: 10.1159/000516750] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/22/2021] [Indexed: 11/19/2022]
Abstract
The epeXEPAB (formerly yydFGHIJ) locus of Bacillus subtilis encodes a minimalistic biosynthetic pathway for a linear antimicrobial epipeptide, EpeX, which is ribosomally produced and post-translationally processed by the action of the radical-SAM epimerase, EpeE, and a membrane-anchored signal 2 peptide peptidase, EpeP. The ABC transporter EpeAB provides intrinsic immunity against self-produced EpeX, without conferring resistance against extrinsically added EpeX. EpeX specifically targets, and severely perturbs the integrity of the cytoplasmic membrane, which leads to the induction of the Lia-dependent envelope stress response. Here, we provide new insights into the distribution, expression, and regulation of the minimalistic epeXEPAB locus of B. subtilis, as well as the biosynthesis and biological efficiency of the produced epipeptide EpeX*. A comprehensive comparative genomics study demonstrates that the epe-locus is restricted to but widely distributed within the phylum Firmicutes. The gene products of epeXEP are necessary and sufficient for the production of the mature antimicrobial peptide EpeX*. In B. subtilis, the epeXEPAB locus is transcribed from three different promoters, one upstream of epeX (PepeX) and two within epeP (PepeA1 and PepeA2). While the latter two are mostly constitutive, PepeX shows a growth phase-dependent induction at the onset of stationary phase. We demonstrate that this regulation is the result of the antagonistic action of two global regulators: The transition state regulator AbrB keeps the epe locus shut off during exponential growth by direct binding. This tight repression is relieved by the master regulator of sporulation, Spo0A, which counteracts the AbrB-dependent repression of epeXEPAB expression during the transition to stationary phase. The net result of these three -promoters is an expression pattern that ensures EpeAB-dependent autoimmunity prior to EpeX* production. In the absence of EpeAB, the general envelope stress response proteins LiaIH can compensate for the loss of specific autoimmunity by providing sufficient protection against the membrane-perturbating action of EpeX*. Hence, the transcriptional regulation of epe expression and the resulting intrinsic induction of the two corresponding resistance functions, encoded by epeAB and liaIH, are well balanced to provide a need-based immunity against mature EpeX*.
Collapse
Affiliation(s)
- Philipp F Popp
- Institute of Microbiology, Technische Universität (TU) Dresden, Dresden, Germany.,Institute for Biology - Bacterial Physiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lena Friebel
- Institute of Microbiology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Alhosna Benjdia
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, Jouy-en-Josas, France
| | - Alain Guillot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, Jouy-en-Josas, France
| | - Olivier Berteau
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, Jouy-en-Josas, France
| | - Thorsten Mascher
- Institute of Microbiology, Technische Universität (TU) Dresden, Dresden, Germany
| |
Collapse
|
21
|
Wirtz DA, Ludwig KC, Arts M, Marx CE, Krannich S, Barac P, Kehraus S, Josten M, Henrichfreise B, Müller A, König GM, Peoples AJ, Nitti A, Spoering AL, Ling LL, Lewis K, Crüsemann M, Schneider T. Biosynthesis and Mechanism of Action of the Cell Wall Targeting Antibiotic Hypeptin. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Daniel A. Wirtz
- Institute for Pharmaceutical Biology University of Bonn Nussallee 6 53115 Bonn Germany
| | - Kevin C. Ludwig
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
- DZIF German Center for Infectious Research, partner site Bonn-Cologne Germany
| | - Melina Arts
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
| | - Carina E. Marx
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
| | - Sebastian Krannich
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
| | - Paul Barac
- Institute for Pharmaceutical Biology University of Bonn Nussallee 6 53115 Bonn Germany
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology University of Bonn Nussallee 6 53115 Bonn Germany
| | - Michaele Josten
- DZIF German Center for Infectious Research, partner site Bonn-Cologne Germany
- Institute for Medical Microbiology, Immunology and Parasitology University Hospital Bonn Venusberg Campus 1 53127 Bonn Germany
| | - Beate Henrichfreise
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
| | - Anna Müller
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
| | - Gabriele M. König
- Institute for Pharmaceutical Biology University of Bonn Nussallee 6 53115 Bonn Germany
| | | | | | | | | | - Kim Lewis
- Department of Biology Antimicrobial Discovery Center Northeastern University Boston MA 02115 USA
| | - Max Crüsemann
- Institute for Pharmaceutical Biology University of Bonn Nussallee 6 53115 Bonn Germany
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology University of Bonn University Clinic Bonn Meckenheimer Allee 168 53115 Bonn Germany
| |
Collapse
|
22
|
Wirtz DA, Ludwig KC, Arts M, Marx CE, Krannich S, Barac P, Kehraus S, Josten M, Henrichfreise B, Müller A, König GM, Peoples AJ, Nitti A, Spoering AL, Ling LL, Lewis K, Crüsemann M, Schneider T. Biosynthesis and Mechanism of Action of the Cell Wall Targeting Antibiotic Hypeptin. Angew Chem Int Ed Engl 2021; 60:13579-13586. [PMID: 33768646 PMCID: PMC8252469 DOI: 10.1002/anie.202102224] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Hypeptin is a cyclodepsipeptide antibiotic produced by Lysobacter sp. K5869, isolated from an environmental sample by the iChip technology, dedicated to the cultivation of previously uncultured microorganisms. Hypeptin shares structural features with teixobactin and exhibits potent activity against a broad spectrum of gram‐positive pathogens. Using comprehensive in vivo and in vitro analyses, we show that hypeptin blocks bacterial cell wall biosynthesis by binding to multiple undecaprenyl pyrophosphate‐containing biosynthesis intermediates, forming a stoichiometric 2:1 complex. Resistance to hypeptin did not readily develop in vitro. Analysis of the hypeptin biosynthetic gene cluster (BGC) supported a model for the synthesis of the octapeptide. Within the BGC, two hydroxylases were identified and characterized, responsible for the stereoselective β‐hydroxylation of four building blocks when bound to peptidyl carrier proteins. In vitro hydroxylation assays corroborate the biosynthetic hypothesis and lead to the proposal of a refined structure for hypeptin.
Collapse
Affiliation(s)
- Daniel A Wirtz
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Kevin C Ludwig
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany.,DZIF, German Center for Infectious Research, partner site Bonn-Cologne, Germany
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| | - Carina E Marx
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| | - Sebastian Krannich
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| | - Paul Barac
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Michaele Josten
- DZIF, German Center for Infectious Research, partner site Bonn-Cologne, Germany.,Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Beate Henrichfreise
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| | - Anna Müller
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | | | - Anthony Nitti
- NovoBiotic Pharmaceuticals, Cambridge, MA, 02138, USA
| | | | - Losee L Ling
- NovoBiotic Pharmaceuticals, Cambridge, MA, 02138, USA
| | - Kim Lewis
- Department of Biology, Antimicrobial Discovery Center, Northeastern University, Boston, MA, 02115, USA
| | - Max Crüsemann
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| |
Collapse
|
23
|
A native conjugative plasmid confers potential selective advantages to plant growth-promoting Bacillus velezensis strain GH1-13. Commun Biol 2021; 4:582. [PMID: 33990691 PMCID: PMC8121941 DOI: 10.1038/s42003-021-02107-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/13/2021] [Indexed: 02/04/2023] Open
Abstract
The conjugative plasmid (pBV71) possibly confers a selective advantage to Bacillus velezensis strain GH1-13, although a selective marker gene is yet to be identified. Here we show that few non-mucoid wild-type GH1-13 cells are spontaneously converted to mucoid variants with or without the loss of pBV71. Mucoid phenotypes, which contain or lack the plasmid, become sensitive to bacitracin, gramicidin, selenite, and tellurite. Using the differences in antibiotic resistance and phenotype, we isolated a reverse complement (COM) and a transconjugant of strain FZB42 with the native pBV71. Transformed COM and FZB42p cells were similar to the wild-type strain GH1-13 with high antibiotic resistance and slow growth rates on lactose compared to those of mucoid phenotypes. RT-PCR analysis revealed that the expression of plasmid-encoded orphan aspartate phosphatase (pRapD) was coordinated with a new quorum-sensing (QS) cassette of RapF2-PhrF2 present in the chromosome of strain GH1-13, but not in strain FZB42. Multi-omics analysis on wild-type and plasmid-cured cells of strain GH1-13 suggested that the conjugative plasmid expression has a crucial role in induction of early envelope stress response that promotes cell morphogenesis, biofilm formation, catabolite repression, and biosynthesis of extracellular-matrix components and antibiotics for protection of host cell during exponential phase.
Collapse
|
24
|
Li Y, Jin G, Liu L, Kuang H, Xiao J, Xu C. A portable fluorescent microsphere-based lateral flow immunosensor for the simultaneous detection of colistin and bacitracin in milk. Analyst 2021; 145:7884-7892. [PMID: 33016277 DOI: 10.1039/d0an01463j] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The polypeptide antibiotics colistin (COL) and bacitracin (Baci) are extensively used as veterinary drugs and feedstock additives in the livestock industry, which inevitably causes residues in animal-origin food, which can accelerate human tolerance to antibiotics. In this study, a portable lateral flow immunoassay (LFIA) for the simultaneous determination of COL and Baci residues in milk was developed. The replacement of gold nanoparticles used in the traditional LFIA with fluorescent microspheres (FMs) to label monoclonal antibodies (mAbs) allowed qualitative and quantitative analyses within a few minutes. Based on the principle of competitive binding to FM-labelled mAbs between analytes in samples and fixed antigens on the membrane, the assay provided qualitative cut-off values of 100 and 50 ng mL-1 for Baci and COL in milk samples. Furthermore, a strip reader-based semi-quantitative detection system could detect lower limits of 7.85 and 1.89 ng mL-1 for Baci and COL, respectively. In conclusion, the proposed multiplex LFIA immunosensor provides an auxiliary analytical tool for the rapid and simultaneous screening of COL and Baci in large cohorts of samples.
Collapse
Affiliation(s)
- Yue Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
25
|
Willdigg JR, Helmann JD. Mini Review: Bacterial Membrane Composition and Its Modulation in Response to Stress. Front Mol Biosci 2021; 8:634438. [PMID: 34046426 PMCID: PMC8144471 DOI: 10.3389/fmolb.2021.634438] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/13/2021] [Indexed: 11/13/2022] Open
Abstract
Antibiotics and other agents that perturb the synthesis or integrity of the bacterial cell envelope trigger compensatory stress responses. Focusing on Bacillus subtilis as a model system, this mini-review summarizes current views of membrane structure and insights into how cell envelope stress responses remodel and protect the membrane. Altering the composition and properties of the membrane and its associated proteome can protect cells against detergents, antimicrobial peptides, and pore-forming compounds while also, indirectly, contributing to resistance against compounds that affect cell wall synthesis. Many of these regulatory responses are broadly conserved, even where the details of regulation may differ, and can be important in the emergence of antibiotic resistance in clinical settings.
Collapse
Affiliation(s)
| | - John D. Helmann
- Department of Microbiology, Cornell University, Ithaca, NY, United States
| |
Collapse
|
26
|
Rismondo J, Schulz LM. Not Just Transporters: Alternative Functions of ABC Transporters in Bacillus subtilis and Listeria monocytogenes. Microorganisms 2021; 9:microorganisms9010163. [PMID: 33450852 PMCID: PMC7828314 DOI: 10.3390/microorganisms9010163] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/24/2022] Open
Abstract
ATP-binding cassette (ABC) transporters are usually involved in the translocation of their cognate substrates, which is driven by ATP hydrolysis. Typically, these transporters are required for the import or export of a wide range of substrates such as sugars, ions and complex organic molecules. ABC exporters can also be involved in the export of toxic compounds such as antibiotics. However, recent studies revealed alternative detoxification mechanisms of ABC transporters. For instance, the ABC transporter BceAB of Bacillus subtilis seems to confer resistance to bacitracin via target protection. In addition, several transporters with functions other than substrate export or import have been identified in the past. Here, we provide an overview of recent findings on ABC transporters of the Gram-positive organisms B. subtilis and Listeria monocytogenes with transport or regulatory functions affecting antibiotic resistance, cell wall biosynthesis, cell division and sporulation.
Collapse
|
27
|
Willdigg JR, Helmann JD. Mini Review: Bacterial Membrane Composition and Its Modulation in Response to Stress. Front Mol Biosci 2021. [PMID: 34046426 DOI: 10.3389/fmolb.2021.634438/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023] Open
Abstract
Antibiotics and other agents that perturb the synthesis or integrity of the bacterial cell envelope trigger compensatory stress responses. Focusing on Bacillus subtilis as a model system, this mini-review summarizes current views of membrane structure and insights into how cell envelope stress responses remodel and protect the membrane. Altering the composition and properties of the membrane and its associated proteome can protect cells against detergents, antimicrobial peptides, and pore-forming compounds while also, indirectly, contributing to resistance against compounds that affect cell wall synthesis. Many of these regulatory responses are broadly conserved, even where the details of regulation may differ, and can be important in the emergence of antibiotic resistance in clinical settings.
Collapse
Affiliation(s)
- Jessica R Willdigg
- Department of Microbiology, Cornell University, Ithaca, NY, United States
| | - John D Helmann
- Department of Microbiology, Cornell University, Ithaca, NY, United States
| |
Collapse
|
28
|
The Cell Envelope Stress Response of Bacillus subtilis towards Laspartomycin C. Antibiotics (Basel) 2020; 9:antibiotics9110729. [PMID: 33114184 PMCID: PMC7690785 DOI: 10.3390/antibiotics9110729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 11/17/2022] Open
Abstract
Cell wall antibiotics are important tools in our fight against Gram-positive pathogens, but many strains become increasingly resistant against existing drugs. Laspartomycin C is a novel antibiotic that targets undecaprenyl phosphate (UP), a key intermediate in the lipid II cycle of cell wall biosynthesis. While laspartomycin C has been thoroughly examined biochemically, detailed knowledge about potential resistance mechanisms in bacteria is lacking. Here, we use reporter strains to monitor the activity of central resistance modules in the Bacillus subtilis cell envelope stress response network during laspartomycin C attack and determine the impact on the resistance of these modules using knock-out strains. In contrast to the closely related UP-binding antibiotic friulimicin B, which only activates ECF σ factor-controlled stress response modules, we find that laspartomycin C additionally triggers activation of stress response systems reacting to membrane perturbation and blockage of other lipid II cycle intermediates. Interestingly, none of the studied resistance genes conferred any kind of protection against laspartomycin C. While this appears promising for therapeutic use of laspartomycin C, it raises concerns that existing cell envelope stress response networks may already be poised for spontaneous development of resistance during prolonged or repeated exposure to this new antibiotic.
Collapse
|
29
|
Wu Y, Xie J, Li J, Zhao J, Qiao S, Li Y, Zeng J. Shared bicycle microbial community: a potential antibiotic-resistant bacteria warehouse. Folia Microbiol (Praha) 2020; 66:49-58. [PMID: 32888177 DOI: 10.1007/s12223-020-00820-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/30/2020] [Indexed: 02/05/2023]
Abstract
Shared bicycle is an emerging form of public transportation in China and around the world. However, the bacterial community and drug-resistant microbiome on these bicycles have not been reported. Samples from 10 shared bicycles were observed by scanning electron microscopy (SEM). Nine samples collected from 90 shared bicycles in three different kinds of location (hospital, metro station, shopping mall) were used for full-length 16S rDNA gene analysis to figure out the bacterial composition of the shared bicycle. Samples from 32 shared bicycles were used to investigate culturable drug-resistant bacteria of the shared bicycle bacterial community. It was found that in the shared bicycle bacterial community, Bacillus was the most abundant bacteria, as determined by both SEM observation and full-length 16S rDNA gene analysis. For the analysis of drug-resistant bacteria, Bacillus showed the strongest drug resist ability. Moreover, the resistances to bacitracin and sulfamethoxazole were the most common among all types of bacteria. Our study provides an important reference for the prevention of the potential spread of drug-resistant bacteria through shared bicycles.
Collapse
Affiliation(s)
- Yuqi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiahui Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junyi Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shihao Qiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
30
|
Revilla-Guarinos A, Dürr F, Popp PF, Döring M, Mascher T. Amphotericin B Specifically Induces the Two-Component System LnrJK: Development of a Novel Whole-Cell Biosensor for the Detection of Amphotericin-Like Polyenes. Front Microbiol 2020; 11:2022. [PMID: 32973732 PMCID: PMC7472640 DOI: 10.3389/fmicb.2020.02022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/30/2020] [Indexed: 11/13/2022] Open
Abstract
The rise of drug-resistant fungal pathogens urges for the development of new tools for the discovery of novel antifungal compounds. Polyene antibiotics are potent agents against fungal infections in humans and animals. They inhibit the growth of fungal cells by binding to sterols in the cytoplasmic membrane that subsequently causes pore formation and eventually results in cell death. Many polyenes are produced by Streptomycetes and released into the soil environment, where they can then target fungal hyphae. While not antibacterial, these compounds could nevertheless be also perceived by bacteria sharing the same habitat and serve as signaling molecules. We therefore addressed the question of how polyenes such as amphotericin B are perceived by the soil bacterium, Bacillus subtilis. Global transcriptional profiling identified a very narrow and specific response, primarily resulting in strong upregulation of the lnrLMN operon, encoding an ABC transporter previously associated with linearmycin resistance. Its strong and specific induction prompted a detailed analysis of the lnrL promoter element and its regulation. We demonstrate that the amphotericin response strictly depends on the two-component system LnrJK and that the target of LnrK-dependent gene regulation, the lnrLMN operon, negatively affects LnrJK-dependent signal transduction. Based on this knowledge, we developed a novel whole-cell biosensor, based on a P lnrL -lux fusion reporter construct in a lnrLMN deletion mutant background. This highly sensitive and dynamic biosensor is ready to be applied for the discovery or characterization of novel amphotericin-like polyenes, hopefully helping to increase the repertoire of antimycotic and antiparasitic polyenes available to treat human and animal infections.
Collapse
Affiliation(s)
| | | | | | | | - Thorsten Mascher
- Department of General Microbiology, Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
31
|
Hernandez-Valdes JA, Zhou L, de Vries MP, Kuipers OP. Impact of spatial proximity on territoriality among human skin bacteria. NPJ Biofilms Microbiomes 2020; 6:30. [PMID: 32764612 PMCID: PMC7413532 DOI: 10.1038/s41522-020-00140-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/14/2020] [Indexed: 12/30/2022] Open
Abstract
Bacteria display social behavior and establish cooperative or competitive interactions in the niches they occupy. The human skin is a densely populated environment where many bacterial species live. Thus, bacterial inhabitants are expected to find a balance in these interactions, which eventually defines their spatial distribution and the composition of our skin microbiota. Unraveling the physiological basis of the interactions between bacterial species in organized environments requires reductionist analyses using functionally relevant species. Here, we study the interaction between two members of our skin microbiota, Bacillus subtilis and Staphylococcus epidermidis. We show that B. subtilis actively responds to the presence of S. epidermidis in its proximity by two strategies: antimicrobial production and development of a subpopulation with migratory response. The initial response of B. subtilis is production of chlorotetain, which degrades the S. epidermidis at the colony level. Next, a subpopulation of B. subtilis motile cells emerges. Remarkably this subpopulation slides towards the remaining S. epidermidis colony and engulfs it. A slow response back from S. epidermidis cells give origin to resistant cells that prevent both attacks from B. subtilis. We hypothesized that this niche conquering and back-down response from B. subtilis and S. epidermidis, respectively, which resembles other conflicts in nature as the ones observed in animals, may play a role in defining the presence of certain bacterial species in the specific microenvironments that these bacteria occupy on our skin.
Collapse
Affiliation(s)
- Jhonatan A Hernandez-Valdes
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Lu Zhou
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Marcel P de Vries
- Department of Biomedical Engineering Antonius Deusinglaan 1, University Medical Center Groningen, Groningen University, 9713 AW, Groningen, Netherlands
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
32
|
Wingen LM, Rausch M, Schneider T, Menche D. Synthesis of Tetramic Acid Fragments Derived from Vancoresmycin Showing Inhibitory Effects towards S. aureus. ChemMedChem 2020; 15:1390-1393. [PMID: 32497343 PMCID: PMC7496136 DOI: 10.1002/cmdc.202000241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Indexed: 11/28/2022]
Abstract
An efficient route to various vancoresmycin‐type tetramic acids has been developed. The modular route is based on an effective Fries‐type rearrangement to introduce various appending acetyl residues. The minimum inhibitory concentration (MIC) values of the new tetramic acids against Staphylococcus aureus and Escherichia coli were determined, revealing that three of the new compounds exhibit antimicrobial activity against S. aureus. These bioactive compounds were structurally most closely related to the authentic vancoresmycin building block. Additionally, the compounds induced a lial‐lux bioreporter, which responds to cell wall stress induced by antibiotics that interfere with the lipid II biosynthesis cycle. These data suggest the tetramic acid moiety to be a part of the vancoresmycin pharmacophore.
Collapse
Affiliation(s)
- Lukas Martin Wingen
- Kekulé-Institut für Organische Chemie und Biochemie, Rheinische Friedrich-Wilhelms-Universität Bonn, Gerhard-Domagk-Str. 1, 53121, Bonn, Germany
| | - Marvin Rausch
- Institut für Pharmazeutische Mikrobiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany.,German Center for Infection Research (DZIF), Partner site, Bonn-Cologne, Bonn, Germany
| | - Tanja Schneider
- Institut für Pharmazeutische Mikrobiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Meckenheimer Allee 168, 53115, Bonn, Germany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und Biochemie, Rheinische Friedrich-Wilhelms-Universität Bonn, Gerhard-Domagk-Str. 1, 53121, Bonn, Germany
| |
Collapse
|
33
|
A critical review of antibiotic resistance in probiotic bacteria. Food Res Int 2020; 136:109571. [PMID: 32846610 DOI: 10.1016/j.foodres.2020.109571] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022]
Abstract
Probiotics are defined as live microorganisms that, when administered in adequate amounts, confer a health benefit upon the host. At present, probiotics are gaining popularity worldwide and are widely used in food and medicine. Consumption of probiotics is increasing with further in-depth research on the relationship between intestinal flora and host health. Most people pay more attention to the function of probiotics but ignore their potential risks, such as infection and antibiotic resistance transfer to pathogenic microbes. Physiological functions, effects and mechanisms of action of probiotics were covered in this review, as well as the antibiotic resistance phenotypes, mechanisms and genes found in probiotics. Typical cases of antibiotic resistance of probiotics were also highlighted, as well as the potential risks (including pathogenicity, infectivity and excessive immune response) and corresponding strategies (dosage, formulation, and administration route). This timely study provides an avenue for further research, development and application of probiotics.
Collapse
|
34
|
Revilla-Guarinos A, Zhang Q, Loderer C, Alcántara C, Müller A, Rahnamaeian M, Vilcinskas A, Gebhard S, Zúñiga M, Mascher T. ABC Transporter DerAB of Lactobacillus casei Mediates Resistance against Insect-Derived Defensins. Appl Environ Microbiol 2020; 86:e00818-20. [PMID: 32414796 PMCID: PMC7357469 DOI: 10.1128/aem.00818-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/09/2020] [Indexed: 01/28/2023] Open
Abstract
Bce-like systems mediate resistance against antimicrobial peptides in Firmicutes bacteria. Lactobacillus casei BL23 encodes an "orphan" ABC transporter that, based on homology to BceAB-like systems, was proposed to contribute to antimicrobial peptide resistance. A mutant lacking the permease subunit was tested for sensitivity against a collection of peptides derived from bacteria, fungi, insects, and humans. Our results show that the transporter specifically conferred resistance against insect-derived cysteine-stabilized αβ defensins, and it was therefore renamed DerAB for defensin resistance ABC transporter. Surprisingly, cells lacking DerAB showed a marked increase in resistance against the lantibiotic nisin. This could be explained by significantly increased expression of the antimicrobial peptide resistance determinants regulated by the Bce-like systems PsdRSAB (formerly module 09) and ApsRSAB (formerly module 12). Bacterial two-hybrid studies in Escherichia coli showed that DerB could interact with proteins of the sensory complex in the Psd resistance system. We therefore propose that interaction of DerAB with this complex in the cell creates signaling interference and reduces the cell's potential to mount an effective nisin resistance response. In the absence of DerB, this negative interference is relieved, leading to the observed hyperactivation of the Psd module and thus increased resistance to nisin. Our results unravel the function of a previously uncharacterized Bce-like orphan resistance transporter with pleiotropic biological effects on the cell.IMPORTANCE Antimicrobial peptides (AMPs) play an important role in suppressing the growth of microorganisms. They can be produced by bacteria themselves-to inhibit competitors-but are also widely distributed in higher eukaryotes, including insects and mammals, where they form an important component of innate immunity. In low-GC-content Gram-positive bacteria, BceAB-like transporters play a crucial role in AMP resistance but have so far been primarily associated with interbacterial competition. Here, we show that the orphan transporter DerAB from the lactic acid bacterium Lactobacillus casei is crucial for high-level resistance against insect-derived AMPs. It therefore represents an important mechanism for interkingdom defense. Furthermore, our results support a signaling interference from DerAB on the PsdRSAB module that might prevent the activation of a full nisin response. The Bce modules from L. casei BL23 illustrate a biological paradox in which the intrinsic nisin detoxification potential only arises in the absence of a defensin-specific ABC transporter.
Collapse
Affiliation(s)
| | - Qian Zhang
- Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Christoph Loderer
- Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Cristina Alcántara
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Ariane Müller
- Institut für Zoologie, Technische Universität Dresden, Dresden, Germany
| | - Mohammad Rahnamaeian
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Department of Bioresources, Giessen, Germany
| | - Andreas Vilcinskas
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Department of Bioresources, Giessen, Germany
- Institute for Insect Biotechnology, Justus Liebig University Giessen, Giessen, Germany
| | - Susanne Gebhard
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, United Kingdom
| | - Manuel Zúñiga
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Thorsten Mascher
- Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
35
|
Hassan A, Ali S, Farooq MA, Tahir HM, Awan MU, Mughal TA. Optimization of enhanced microbial production of zinc bacitracin by submerged fermentation technology. J Basic Microbiol 2020; 60:585-599. [PMID: 32364268 DOI: 10.1002/jobm.201900694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/01/2020] [Accepted: 04/20/2020] [Indexed: 11/10/2022]
Abstract
Bacitracin is one of the most important antibiotics used in different biomedical fields. It helps to achieve sizeable amount of foreign exchange due to its use in the poultry feed. The cheap agricultural wastes are readily available for the preparation of fermentation media used for bacitracin production. The microorganisms could be mutated with different chemicals and UV radiation to improve bacitracin production. Thus, the current study was focused on the synthesis of low-cost and effective bacitracin by mutant strains of Bacillus licheniformis, employing the submerged fermentation technique. The bacteria were exposed to the UV irradiation for various time periods ranging from 5 to 40 min. These mutants were named as BLAA-5-BLAA-40. Mutant strain BLAA-25 produced maximum bacitracin, with significantly high activity (142.81 IU/mg) against Klebsiella pneumoniae but less activity against Escherichia coli (115.19 IU/mg). Several fermentation conditions were investigated to optimize bacitracin production. The highest bacitracin yield was obtained by an inoculum size of 10%, fermentation period 48 hr, pH 7, T = 37°C, using soybean meal as a substrate. Among all substrates, cucumber peel was the substrate showing the highest minimum inhibitory concentration (2.3 mg/ml and 2.7 mg/ml against K. pneumoniae and E. coli respectively). A comparison between commercial and experimentally produced Zn bacitracin showed that commercial bacitracin has a low activity (63.2 IU/mg) as compared with experimental bacitracin. Hence, the agro wastes and mutation could be used to increase the synthesis of Zn bacitracin in B. licheniformis.
Collapse
Affiliation(s)
- Ali Hassan
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Muhammad A Farooq
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Hafiz M Tahir
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Muhammad U Awan
- Department of Botany, Government College University, Lahore, Pakistan
| | - Tafail A Mughal
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| |
Collapse
|
36
|
Ca 2+-Daptomycin targets cell wall biosynthesis by forming a tripartite complex with undecaprenyl-coupled intermediates and membrane lipids. Nat Commun 2020; 11:1455. [PMID: 32193379 PMCID: PMC7081307 DOI: 10.1038/s41467-020-15257-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/25/2020] [Indexed: 11/08/2022] Open
Abstract
The lipopeptide daptomycin is used as an antibiotic to treat severe infections with gram-positive pathogens, such as methicillin resistant Staphylococcus aureus (MRSA) and drug-resistant enterococci. Its precise mechanism of action is incompletely understood, and a specific molecular target has not been identified. Here we show that Ca2+-daptomycin specifically interacts with undecaprenyl-coupled cell envelope precursors in the presence of the anionic phospholipid phosphatidylglycerol, forming a tripartite complex. We use microbiological and biochemical assays, in combination with fluorescence and optical sectioning microscopy of intact staphylococcal cells and model membrane systems. Binding primarily occurs at the staphylococcal septum and interrupts cell wall biosynthesis. This is followed by delocalisation of components of the peptidoglycan biosynthesis machinery and massive membrane rearrangements, which may account for the pleiotropic cellular events previously reported. The identification of carrier-bound cell wall precursors as specific targets explains the specificity of daptomycin for bacterial cells. Our work reconciles apparently inconsistent previous results, and supports a concise model for the mode of action of daptomycin.
Collapse
|
37
|
BceAB-Type Antibiotic Resistance Transporters Appear To Act by Target Protection of Cell Wall Synthesis. Antimicrob Agents Chemother 2020; 64:AAC.02241-19. [PMID: 31871088 PMCID: PMC7038271 DOI: 10.1128/aac.02241-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/18/2019] [Indexed: 11/25/2022] Open
Abstract
Resistance against cell wall-active antimicrobial peptides in bacteria is often mediated by transporters. In low-GC-content Gram-positive bacteria, a common type of such transporters is BceAB-like systems, which frequently provide high-level resistance against peptide antibiotics that target intermediates of the lipid II cycle of cell wall synthesis. How a transporter can offer protection from drugs that are active on the cell surface, however, has presented researchers with a conundrum. Resistance against cell wall-active antimicrobial peptides in bacteria is often mediated by transporters. In low-GC-content Gram-positive bacteria, a common type of such transporters is BceAB-like systems, which frequently provide high-level resistance against peptide antibiotics that target intermediates of the lipid II cycle of cell wall synthesis. How a transporter can offer protection from drugs that are active on the cell surface, however, has presented researchers with a conundrum. Multiple theories have been discussed, ranging from removal of the peptides from the membrane and internalization of the drug for degradation to removal of the cellular target rather than the drug itself. To resolve this much-debated question, we here investigated the mode of action of the transporter BceAB of Bacillus subtilis. We show that it does not inactivate or import its substrate antibiotic bacitracin. Moreover, we present evidence that the critical factor driving transport activity is not the drug itself but instead the concentration of drug-target complexes in the cell. Our results, together with previously reported findings, lead us to propose that BceAB-type transporters act by transiently freeing lipid II cycle intermediates from the inhibitory grip of antimicrobial peptides and thus provide resistance through target protection of cell wall synthesis. Target protection has so far only been reported for resistance against antibiotics with intracellular targets, such as the ribosome. However, this mechanism offers a plausible explanation for the use of transporters as resistance determinants against cell wall-active antibiotics in Gram-positive bacteria where cell wall synthesis lacks the additional protection of an outer membrane.
Collapse
|
38
|
Popp PF, Benjdia A, Strahl H, Berteau O, Mascher T. The Epipeptide YydF Intrinsically Triggers the Cell Envelope Stress Response of Bacillus subtilis and Causes Severe Membrane Perturbations. Front Microbiol 2020; 11:151. [PMID: 32117169 PMCID: PMC7026026 DOI: 10.3389/fmicb.2020.00151] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/22/2020] [Indexed: 01/05/2023] Open
Abstract
The Gram-positive model organism and soil bacterium Bacillus subtilis naturally produces a variety of antimicrobial peptides (AMPs), including the ribosomally synthesized and post-translationally modified AMP YydF, which is encoded in the yydFGHIJ locus. The yydF gene encodes the pre-pro-peptide, which is, in a unique manner, initially modified at two amino acid positions by the radical SAM epimerase YydG. Subsequently, the membrane-anchored putative protease YydH is thought to cleave and release the mature AMP, YydF, to the environment. The AMP YydF, with two discreet epimerizations among 17 residues as sole post-translational modification, defines a novel class of ribosomally synthesized and post-translationally modified peptides (RiPPs) called epipeptides, for which the mode-of-action (MOA) is unknown. The predicted ABC transporter encoded by yydIJ was previously postulated as an autoimmunity determinant of B. subtilis against its own AMP. Here, we demonstrate that extrinsically added YydF* kills B. subtilis cells by dissipating membrane potential via membrane permeabilization. This severe membrane perturbation is accompanied by a rapid reduction of membrane fluidity, substantiated by lipid domain formation. The epipeptide triggers a narrow and highly specific cellular response. The strong induction of liaIH expression, a marker for cell envelope stress in B. subtilis, further supports the MOA described above. A subsequent mutational study demonstrates that LiaIH—and not YydIJ—represents the most efficient resistance determinant against YydF* action. Unexpectedly, none of the observed cellular effects upon YydF* treatment alone are able to trigger liaIH expression, indicating that only the unique combination of membrane permeabilization and membrane rigidification caused by the epipetide, leads to the observed cell envelope stress response.
Collapse
Affiliation(s)
- Philipp F Popp
- Institute of Microbiology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Alhosna Benjdia
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | - Henrik Strahl
- Center for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Olivier Berteau
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, Jouy-en-Josas, France
| | - Thorsten Mascher
- Institute of Microbiology, Technische Universität (TU) Dresden, Dresden, Germany
| |
Collapse
|
39
|
From Modules to Networks: a Systems-Level Analysis of the Bacitracin Stress Response in Bacillus subtilis. mSystems 2020; 5:5/1/e00687-19. [PMID: 32019833 PMCID: PMC7002115 DOI: 10.1128/msystems.00687-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Antibiotic resistance poses a major threat to global health, and systematic studies to understand the underlying resistance mechanisms are urgently needed. Although significant progress has been made in deciphering the mechanistic basis of individual resistance determinants, many bacterial species rely on the induction of a whole battery of resistance modules, and the complex regulatory networks controlling these modules in response to antibiotic stress are often poorly understood. In this work we combined experiments and theoretical modeling to decipher the resistance network of Bacillus subtilis against bacitracin, which inhibits cell wall biosynthesis in Gram-positive bacteria. We found a high level of cross-regulation between the two major resistance modules in response to bacitracin stress and quantified their effects on bacterial resistance. To rationalize our experimental data, we expanded a previously established computational model for the lipid II cycle through incorporating the quantitative action of the resistance modules. This led us to a systems-level description of the bacitracin stress response network that captures the complex interplay between resistance modules and the essential lipid II cycle of cell wall biosynthesis and accurately predicts the minimal inhibitory bacitracin concentration in all the studied mutants. With this, our study highlights how bacterial resistance emerges from an interlaced network of redundant homeostasis and stress response modules. Bacterial resistance against antibiotics often involves multiple mechanisms that are interconnected to ensure robust protection. So far, the knowledge about underlying regulatory features of those resistance networks is sparse, since they can hardly be determined by experimentation alone. Here, we present the first computational approach to elucidate the interplay between multiple resistance modules against a single antibiotic and how regulatory network structure allows the cell to respond to and compensate for perturbations of resistance. Based on the response of Bacillus subtilis toward the cell wall synthesis-inhibiting antibiotic bacitracin, we developed a mathematical model that comprehensively describes the protective effect of two well-studied resistance modules (BceAB and BcrC) on the progression of the lipid II cycle. By integrating experimental measurements of expression levels, the model accurately predicts the efficacy of bacitracin against the B. subtilis wild type as well as mutant strains lacking one or both of the resistance modules. Our study reveals that bacitracin-induced changes in the properties of the lipid II cycle itself control the interplay between the two resistance modules. In particular, variations in the concentrations of UPP, the lipid II cycle intermediate that is targeted by bacitracin, connect the effect of the BceAB transporter and the homeostatic response via BcrC to an overall resistance response. We propose that monitoring changes in pathway properties caused by a stressor allows the cell to fine-tune deployment of multiple resistance systems and may serve as a cost-beneficial strategy to control the overall response toward this stressor. IMPORTANCE Antibiotic resistance poses a major threat to global health, and systematic studies to understand the underlying resistance mechanisms are urgently needed. Although significant progress has been made in deciphering the mechanistic basis of individual resistance determinants, many bacterial species rely on the induction of a whole battery of resistance modules, and the complex regulatory networks controlling these modules in response to antibiotic stress are often poorly understood. In this work we combined experiments and theoretical modeling to decipher the resistance network of Bacillus subtilis against bacitracin, which inhibits cell wall biosynthesis in Gram-positive bacteria. We found a high level of cross-regulation between the two major resistance modules in response to bacitracin stress and quantified their effects on bacterial resistance. To rationalize our experimental data, we expanded a previously established computational model for the lipid II cycle through incorporating the quantitative action of the resistance modules. This led us to a systems-level description of the bacitracin stress response network that captures the complex interplay between resistance modules and the essential lipid II cycle of cell wall biosynthesis and accurately predicts the minimal inhibitory bacitracin concentration in all the studied mutants. With this, our study highlights how bacterial resistance emerges from an interlaced network of redundant homeostasis and stress response modules.
Collapse
|
40
|
Sun Z, Popp PF, Loderer C, Revilla-Guarinos A. Genetically Engineered Bacterial Biohybrid Microswimmers for Sensing Applications. SENSORS (BASEL, SWITZERLAND) 2019; 20:E180. [PMID: 31905650 PMCID: PMC6982730 DOI: 10.3390/s20010180] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/17/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
Bacterial biohybrid microswimmers aim at exploiting the inherent motion capabilities of bacteria (carriers) to transport objects (cargoes) at the microscale. One of the most desired properties of microswimmers is their ability to communicate with their immediate environment by processing the information and producing a useful response. Indeed, bacteria are naturally equipped with such communication skills. Hereby, two-component systems (TCSs) represent the key signal transducing machinery and enable bacteria to sense and respond to a variety of stimuli. We engineered a natural microswimmer based on the Gram-positive model bacterium Bacillus subtilis for the development of biohybrids with sensing abilities. B. subtilis naturally adhered to silica particles, giving rise to different motile biohybrids systems with variable ratios of carrier(s)-to-cargo(es). Genetically engineered TCS pathways allowed us to couple the binding to the inert particles with signaling the presence of antibiotics in their surroundings. Activation of the antibiotic-induced TCSs resulted in fluorescent bacterial carriers as a response readout. We demonstrate that the genetically engineered TCS-mediated signaling capabilities of B. subtilis allow for the custom design of bacterial hybrid microswimmers able to sense and signal the presence of target molecules in the environment. The generally recognized as safe (GRAS) status of B. subtilis makes it a promising candidate for human-related applications of these novel biohybrids.
Collapse
Affiliation(s)
- Zhiyong Sun
- Department of Molecular Biotechnology, Institute für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Philipp F. Popp
- Department of General Microbiology, Institute für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Christoph Loderer
- Department of Molecular Biotechnology, Institute für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| | - Ainhoa Revilla-Guarinos
- Department of General Microbiology, Institute für Mikrobiologie, Technische Universität Dresden, 01217 Dresden, Germany
| |
Collapse
|
41
|
Jung K, Brameyer S, Fabiani F, Gasperotti A, Hoyer E. Phenotypic Heterogeneity Generated by Histidine Kinase-Based Signaling Networks. J Mol Biol 2019; 431:4547-4558. [DOI: 10.1016/j.jmb.2019.03.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 01/16/2023]
|
42
|
Ma J, Liu J, Zhang Y, Wang D, Liu R, Liu G, Yao H, Pan Z. Bacitracin resistance and enhanced virulence of Streptococcus suis via a novel efflux pump. BMC Vet Res 2019; 15:377. [PMID: 31660968 PMCID: PMC6819616 DOI: 10.1186/s12917-019-2115-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023] Open
Abstract
Background Streptococcus suis is a prominent pathogen causing septicemia and meningitis in swine and humans. Bacitracin is used widely as a growth promoter in animal feed and to control the spread of necrotic enteritis in most developing countries. This study aimed to characterize a novel membrane transporter module Sst comprising SstE, SstF, and SstG for bacitracin resistance. Results Comparative genomics and protein homology analysis found a potential efflux pump SstFEG encoded upstream of well-known bacitracin-resistance genes bceAB and bceRS. A four-fold decrease in bacitracin susceptibility was observed in sstFEG deletion mutant comparing with S. suis wildtype strain CZ130302. Further studies indicated that the bacitracin tolerance mediated by SstFEG is not only independent of the BceAB transporter, but also regulated by the two-component system BceSR. Given that SstFEG are harbored by almost all virulent strains, but not in the avirulent strains, we managed to explore its potential role in bacterial pathogencity. Indeed, our results showed that SstFEG is involved in S. suis colonization and virulence in animal infection model by its potential competitive survival advantage against host bactericidal effect. Conclusion To our knowledge, this is the first study to functionally characterize the bacitracin efflux pump in S. suis to provide evidence regarding the important roles of the novel ABC transporter system SstFEG with respect to drug resistance and virulence.
Collapse
Affiliation(s)
- Jiale Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Jin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yue Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Dan Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Runxia Liu
- South Dakota State University, Brookings, SD, 57007, USA
| | - Guangjin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Huochun Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China
| | - Zihao Pan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China. .,OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China.
| |
Collapse
|
43
|
Environment Shapes the Accessible Daptomycin Resistance Mechanisms in Enterococcus faecium. Antimicrob Agents Chemother 2019; 63:AAC.00790-19. [PMID: 31332078 DOI: 10.1128/aac.00790-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
Daptomycin binds to bacterial cell membranes and disrupts essential cell envelope processes, leading to cell death. Bacteria respond to daptomycin by altering their cell envelopes to either decrease antibiotic binding to the membrane or by diverting binding away from septal targets. In Enterococcus faecalis, daptomycin resistance is typically coordinated by the three-component cell envelope stress response system, LiaFSR. Here, studying a clinical strain of multidrug-resistant Enterococcus faecium containing alleles associated with activation of the LiaFSR signaling pathway, we found that specific environments selected for different evolutionary trajectories, leading to high-level daptomycin resistance. Planktonic environments favored pathways that increased cell surface charge via yvcRS upregulation of dltABCD and mprF, causing a reduction in daptomycin binding. Alternatively, environments favoring complex structured communities, including biofilms, evolved both diversion and repulsion strategies via divIVA and oatA mutations, respectively. Both environments subsequently converged on cardiolipin synthase (cls) mutations, suggesting the importance of membrane modification across strategies. Our findings indicate that E. faecium can evolve diverse evolutionary trajectories to daptomycin resistance that are shaped by the environment to produce a combination of resistance strategies. The accessibility of multiple and different biochemical pathways simultaneously suggests that the outcome of daptomycin exposure results in a polymorphic population of resistant phenotypes, making E. faecium a recalcitrant nosocomial pathogen.
Collapse
|
44
|
Minimal exposure of lipid II cycle intermediates triggers cell wall antibiotic resistance. Nat Commun 2019; 10:2733. [PMID: 31227716 PMCID: PMC6588590 DOI: 10.1038/s41467-019-10673-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/23/2019] [Indexed: 01/08/2023] Open
Abstract
Cell wall antibiotics are crucial for combatting the emerging wave of resistant bacteria. Yet, our understanding of antibiotic action is limited, as many strains devoid of all resistance determinants display far higher antibiotic tolerance in vivo than suggested by the antibiotic-target binding affinity in vitro. To resolve this conflict, here we develop a comprehensive theory for the bacterial cell wall biosynthetic pathway and study its perturbation by antibiotics. We find that the closed-loop architecture of the lipid II cycle of wall biosynthesis features a highly asymmetric distribution of pathway intermediates, and show that antibiotic tolerance scales inversely with the abundance of the targeted pathway intermediate. We formalize this principle of minimal target exposure as intrinsic resistance mechanism and predict how cooperative drug-target interactions can mitigate resistance. The theory accurately predicts the in vivo efficacy for various cell wall antibiotics in different Gram-positive bacteria and contributes to a systems-level understanding of antibiotic action.
Collapse
|
45
|
Tan S, Ludwig KC, Müller A, Schneider T, Nodwell JR. The Lasso Peptide Siamycin-I Targets Lipid II at the Gram-Positive Cell Surface. ACS Chem Biol 2019; 14:966-974. [PMID: 31026131 DOI: 10.1021/acschembio.9b00157] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ribosomally synthesized post-translationally modified peptides (RiPPs) are a diverse class of biologically active molecules produced by many environmental bacteria. While thousands of these compounds have been identified, mostly through genome mining, a relatively small number has been investigated at the molecular level. One less understood class of RiPPs is the lasso peptides. These are 20-25 amino acid residue compounds bearing an N-terminal macrocyclic ring and a C-terminal tail that is threaded through the ring. We have carried out a detailed investigation on the mechanism of action of the siamycin-I lasso peptide. We demonstrate that siamycin-I interacts with lipid II, the central building block of the major cell wall component peptidoglycan, which is readily accessible on the outside of the cell. This interaction compromises cell wall biosynthesis in a manner that activates the liaI stress response. Additionally, resistance to siamycin-I can be brought about by mutations in the essential WalKR two-component system that causes thickening of the cell wall. Siamycin-I is the first lasso peptide that has been shown to inhibit cell wall biosynthesis.
Collapse
Affiliation(s)
- Stephanie Tan
- Department of Biochemistry, MaRS Discovery District, University of Toronto, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| | - Kevin C. Ludwig
- Institute for Pharmaceutical Microbiology, University of Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Anna Müller
- Institute for Pharmaceutical Microbiology, University of Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Justin R. Nodwell
- Department of Biochemistry, MaRS Discovery District, University of Toronto, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| |
Collapse
|
46
|
Miller WR, Tran TT, Diaz L, Rios R, Khan A, Reyes J, Prater AG, Panesso D, Shamoo Y, Arias CA. LiaR-independent pathways to daptomycin resistance in Enterococcus faecalis reveal a multilayer defense against cell envelope antibiotics. Mol Microbiol 2019; 111:811-824. [PMID: 30582877 PMCID: PMC6417935 DOI: 10.1111/mmi.14193] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2018] [Indexed: 12/17/2022]
Abstract
The lipopeptide antibiotic daptomycin (DAP) is a key drug against serious enterococcal infections, but the emergence of resistance in the clinical setting is a major concern. The LiaFSR system plays a prominent role in the development of DAP resistance (DAP-R) in enterococci, and blocking this stress response system has been proposed as a novel therapeutic strategy. In this work, we identify LiaR-independent pathways in Enterococcus faecalis that regulate cell membrane adaptation in response to antibiotics. We adapted E. faecalis OG1RF (a laboratory strain) and S613TM (a clinical strain) lacking liaR to increasing concentrations of DAP, leading to the development of DAP-R and elevated MICs to bacitracin and ceftriaxone. Whole genome sequencing identified changes in the YxdJK two-component regulatory system and a putative fatty acid kinase (dak) in both DAP-R strains. Deletion of the gene encoding the YxdJ response regulator in both the DAP-R mutant and wild-type OG1RF decreased MICs to DAP, even when a functional LiaFSR system was present. Mutations in dak were associated with slower growth, decreased membrane fluidity and alterations of cell morphology. These findings suggest that overlapping stress response pathways can provide protection against antimicrobial peptides in E. faecalis at a significant cost in bacterial fitness.
Collapse
Affiliation(s)
- William R. Miller
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Division of Infectious Diseases, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
| | - Truc T. Tran
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Division of Infectious Diseases, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
| | - Lorena Diaz
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Molecular Genetics and Antimicrobial Resistance Unit, International Center for Microbial Genomics, Universidad El Bosque, Bogotá, Colombia
| | - Rafael Rios
- Molecular Genetics and Antimicrobial Resistance Unit, International Center for Microbial Genomics, Universidad El Bosque, Bogotá, Colombia
| | - Ayesha Khan
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
| | - Jinnethe Reyes
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Molecular Genetics and Antimicrobial Resistance Unit, International Center for Microbial Genomics, Universidad El Bosque, Bogotá, Colombia
| | - Amy G. Prater
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Diana Panesso
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Division of Infectious Diseases, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Molecular Genetics and Antimicrobial Resistance Unit, International Center for Microbial Genomics, Universidad El Bosque, Bogotá, Colombia
| | - Yousif Shamoo
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Cesar A. Arias
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Division of Infectious Diseases, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, McGovern School of Medicine, Houston, TX, USA
- Center for Infectious Diseases, University of Texas Health Science Center, School of Public Health, Houston, TX, USA
- Molecular Genetics and Antimicrobial Resistance Unit, International Center for Microbial Genomics, Universidad El Bosque, Bogotá, Colombia
| |
Collapse
|
47
|
Loss of Bacitracin Resistance Due to a Large Genomic Deletion among Bacillus anthracis Strains. mSystems 2018; 3:mSystems00182-18. [PMID: 30417107 PMCID: PMC6208641 DOI: 10.1128/msystems.00182-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/04/2018] [Indexed: 12/25/2022] Open
Abstract
Anthrax is caused by Bacillus anthracis, an endospore-forming soil bacterium. The genetic diversity of B. anthracis is known to be low compared with that of Bacillus species. In this study, we performed whole-genome sequencing of Zambian isolates of B. anthracis to understand the genetic diversity between closely related strains. Comparison of genomic sequences revealed that closely related strains were separated into three groups based on single nucleotide polymorphisms distributed throughout the genome. A large genomic deletion was detected in the region containing a bacitracin resistance gene cluster flanked by rRNA operons, resulting in the loss of bacitracin resistance. The structure of the deleted region, which was also conserved among species of the Bacillus cereus group, has the potential for both deletion and amplification and thus might be enabling the species to flexibly control the level of bacitracin resistance for adaptive evolution. Bacillus anthracis is a Gram-positive endospore-forming bacterial species that causes anthrax in both humans and animals. In Zambia, anthrax cases are frequently reported in both livestock and wildlife, with occasional transmission to humans, causing serious public health problems in the country. To understand the genetic diversity of B. anthracis strains in Zambia, we sequenced and compared the genomic DNA of B. anthracis strains isolated across the country. Single nucleotide polymorphisms clustered these strains into three groups. Genome sequence comparisons revealed a large deletion in strains belonging to one of the groups, possibly due to unequal crossing over between a pair of rRNA operons. The deleted genomic region included genes conferring resistance to bacitracin, and the strains with the deletion were confirmed with loss of bacitracin resistance. Similar deletions between rRNA operons were also observed in a few B. anthracis strains phylogenetically distant from Zambian strains. The structure of bacitracin resistance genes flanked by rRNA operons was conserved only in members of the Bacillus cereus group. The diversity and genomic characteristics of B. anthracis strains determined in this study would help in the development of genetic markers and treatment of anthrax in Zambia. IMPORTANCE Anthrax is caused by Bacillus anthracis, an endospore-forming soil bacterium. The genetic diversity of B. anthracis is known to be low compared with that of Bacillus species. In this study, we performed whole-genome sequencing of Zambian isolates of B. anthracis to understand the genetic diversity between closely related strains. Comparison of genomic sequences revealed that closely related strains were separated into three groups based on single nucleotide polymorphisms distributed throughout the genome. A large genomic deletion was detected in the region containing a bacitracin resistance gene cluster flanked by rRNA operons, resulting in the loss of bacitracin resistance. The structure of the deleted region, which was also conserved among species of the Bacillus cereus group, has the potential for both deletion and amplification and thus might be enabling the species to flexibly control the level of bacitracin resistance for adaptive evolution.
Collapse
|
48
|
Patel V, Wu Q, Chandrangsu P, Helmann JD. A metabolic checkpoint protein GlmR is important for diverting carbon into peptidoglycan biosynthesis in Bacillus subtilis. PLoS Genet 2018; 14:e1007689. [PMID: 30248093 PMCID: PMC6171935 DOI: 10.1371/journal.pgen.1007689] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 10/04/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022] Open
Abstract
The Bacillus subtilis GlmR (formerly YvcK) protein is essential for growth on gluconeogenic carbon sources. Mutants lacking GlmR display a variety of phenotypes suggestive of impaired cell wall synthesis including antibiotic sensitivity, aberrant cell morphology and lysis. To define the role of GlmR, we selected suppressor mutations that ameliorate the sensitivity of a glmR null mutant to the beta-lactam antibiotic cefuroxime or restore growth on gluconeogenic carbon sources. Several of the resulting suppressors increase the expression of the GlmS and GlmM proteins that catalyze the first two committed steps in the diversion of carbon from central carbon metabolism into peptidoglycan biosynthesis. Chemical complementation studies indicate that the absence of GlmR can be overcome by provision of cells with N-acetylglucosamine (GlcNAc), even under conditions where GlcNAc cannot re-enter central metabolism and serve as a carbon source for growth. Our results indicate that GlmR facilitates the diversion of carbon from the central metabolite fructose-6-phosphate, which is limiting in cells growing on gluconeogenic carbon sources, into peptidoglycan biosynthesis. Our data suggest that GlmR stimulates GlmS activity, and we propose that this activation is antagonized by the known GlmR ligand and peptidoglycan intermediate UDP-GlcNAc. Thus, GlmR presides over a new mechanism for the regulation of carbon partitioning between central metabolism and peptidoglycan biosynthesis.
Collapse
Affiliation(s)
- Vaidehi Patel
- Cornell University, Department of Microbiology, Ithaca, NY, United States of America
| | - Qun Wu
- Cornell University, Department of Microbiology, Ithaca, NY, United States of America
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Pete Chandrangsu
- Cornell University, Department of Microbiology, Ithaca, NY, United States of America
| | - John D. Helmann
- Cornell University, Department of Microbiology, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
49
|
Vitamin D Compounds Are Bactericidal against Streptococcus mutans and Target the Bacitracin-Associated Efflux System. Antimicrob Agents Chemother 2017; 62:AAC.01675-17. [PMID: 29061743 DOI: 10.1128/aac.01675-17] [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: 08/14/2017] [Accepted: 10/07/2017] [Indexed: 12/14/2022] Open
Abstract
Vitamin D analogs were identified as compounds that induced lysis of planktonic cultures of Streptococcus mutans in a high-throughput screen of FDA-approved drugs. Previous studies have demonstrated that certain derivatives of vitamin D possess lytic activity against other bacteria, though the mechanism has not yet been established. Through the use of a combinatorial approach, the vitamin D derivative doxercalciferol was shown to act synergistically with bacitracin, a polypeptide-type drug that is known to interfere with cell wall synthesis, suggesting that doxercalciferol may act in a bacitracin-related pathway. Innate resistance to bacitracin is attributed to efflux by a conserved ABC-type transporter, which in S. mutans is encoded by the mbrABCD operon. S. mutans possesses two characterized mechanisms of resistance to bacitracin, the ABC transporter, S. mutans bacitracin resistance (Mbr) cassette, consisting of MbrABCD, and the rhamnose-glucose polysaccharide (Rgp) system, RgpABCDEFGHI. Loss of function of the transporter in ΔmbrA and ΔmbrD mutants exacerbated the effect of the combination of doxercalciferol and bacitracin. Despite conservation of a transporter homologous to mbrABCD, the combination of doxercalciferol and bacitracin appeared to be synergistic only in streptococcal species. We conclude that vitamin D derivatives possess lytic activity against S. mutans and act through a mechanism dependent on the bacitracin resistance mechanism of MbrABCD.
Collapse
|
50
|
Radeck J, Lautenschläger N, Mascher T. The Essential UPP Phosphatase Pair BcrC and UppP Connects Cell Wall Homeostasis during Growth and Sporulation with Cell Envelope Stress Response in Bacillus subtilis. Front Microbiol 2017; 8:2403. [PMID: 29259598 PMCID: PMC5723303 DOI: 10.3389/fmicb.2017.02403] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/20/2017] [Indexed: 12/03/2022] Open
Abstract
The bacterial cell wall separates the cell from its surrounding and protects it from environmental stressors. Its integrity is maintained by a highly regulated process of cell wall biosynthesis. The membrane-located lipid II cycle provides cell wall building blocks that are assembled inside the cytoplasm to the outside for incorporation. Its carrier molecule, undecaprenyl phosphate (UP), is then recycled by dephosphorylation from undecaprenyl pyrophosphate (UPP). In Bacillus subtilis, this indispensable reaction is catalyzed by the UPP phosphatases BcrC and UppP. Here, we study the physiological function of both phosphatases with respect to morphology, cell wall homeostasis and the resulting cell envelope stress response (CESR). We demonstrate that uppP and bcrC represent a synthetic lethal gene pair, which encodes an essential physiological function. Accordingly, cell growth and morphology were severely impaired during exponential growth if the overall UPP phosphatase level was limiting. UppP, but not BcrC, was crucial for normal sporulation. Expression of bcrC, but not uppP, was upregulated in the presence of cell envelope stress conditions caused by bacitracin if UPP phosphatase levels were limited. This homeostatic feedback renders BcrC more important during growth than UppP, particularly in defense against cell envelope stress.
Collapse
Affiliation(s)
- Jara Radeck
- Institute of Microbiology, Technische Universität Dresden, Dresden, Germany
| | | | - Thorsten Mascher
- Institute of Microbiology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|