1
|
Pinkett HW. The Evolution of ABC Importers. J Mol Biol 2025; 437:169082. [PMID: 40089147 PMCID: PMC12042770 DOI: 10.1016/j.jmb.2025.169082] [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: 01/14/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
I am an Associate Professor in the Department of Molecular Biosciences at Northwestern University. My research program investigates the structure, function, and regulation of membrane proteins, with a particular emphasis on ATP-binding cassette (ABC) importers. ABC transporters are a highly conserved superfamily of transmembrane proteins found across all organisms. These proteins utilize the energy from ATP binding and hydrolysis to transport of a broad array of substrates- including metabolites, lipids, peptides and drugs- across cellular membranes. In this perspective, I discuss how structural and biophysical characterization of ABC importers have significantly advanced our understanding of the mechanisms underlying their transport function. I also highlight the challenges in developing a unified mechanistic model and propose that the remarkable diversity of ABC transporters may necessitate multiple transport mechanisms for a complete picture of how these critical proteins function.
Collapse
Affiliation(s)
- Heather W Pinkett
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
2
|
Kulikovsky A, Yagmurov E, Grigoreva A, Popov A, Severinov K, Nair SK, Lippens G, Serebryakova M, Borukhov S, Dubiley S. Bacillus subtilis Utilizes Decarboxylated S-Adenosylmethionine for the Biosynthesis of Tandem Aminopropylated Microcin C, a Potent Inhibitor of Bacterial Aspartyl-tRNA Synthetase. J Am Chem Soc 2025; 147:11998-12011. [PMID: 40162528 DOI: 10.1021/jacs.4c18468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The biosynthetic pathways of natural products involve unusual biochemical reactions catalyzed by unique enzymes. Aminopropylation, although apparently simple, is an extremely rare modification outside polyamine biosynthesis. The canonical pathway used in the biosynthesis of peptide-adenylate antibiotic microcin C of E. coli (Eco-McC) entails alkylation by the S-adenosyl-methionine-derived 3-amino-3-carboxypropyl group of the adenylate moiety and subsequent decarboxylation to yield the bioactive aminopropylated compound. Here, we report the structure and biosynthesis of a new member of the microcin C family of antibiotics, Bsu-McC, produced by Bacillus subtilis MG27, which employs an alternative aminopropylation pathway. Like Eco-McC, Bsu-McC consists of a peptide moiety that facilitates prodrug import into susceptible bacteria and a warhead, a nonhydrolyzable modified isoasparaginyl-adenylate, which, when released into the cytoplasm, binds aspartyl-tRNA synthetase (AspRS) inhibiting translation. In contrast to the Eco-McC, whose warhead carries a single aminopropyl group attached to the phosphate moiety of isoasparaginyl-adenylate, the warhead of Bsu-McC is decorated with a tandem of two aminopropyl groups. Our in silico docking of the Bsu-McC warhead to the AspRS-tRNA complex suggests that two aminopropyl groups form extended interactions with the enzyme and tRNA, stabilizing the enzyme-inhibitor complex. We show that tandem aminopropylation results in a 32-fold increase in the biological activity of peptidyl-adenylate. We also show that B. subtilis adopted an alternative pathway for aminopropylation in which two homologous 3-aminopropyltransferases utilize decarboxylated S-adenosylmethionine as a substrate. Additionally, Bsu-McC biosynthesis alters the social behavior of the B. subtilis producer strain, resulting in a sharp decrease in their ability to form biofilms.
Collapse
Affiliation(s)
- Alexey Kulikovsky
- Institute of Gene Biology, Russian Academy of Science institution, Moscow 119334, Russia
| | - Eldar Yagmurov
- Institute of Gene Biology, Russian Academy of Science institution, Moscow 119334, Russia
| | - Anastasiia Grigoreva
- Institute of Gene Biology, Russian Academy of Science institution, Moscow 119334, Russia
| | - Aleksandr Popov
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa 230-0045, Japan
| | - Konstantin Severinov
- Institute of Gene Biology, Russian Academy of Science institution, Moscow 119334, Russia
| | - Satish K Nair
- Department of Biochemistry, University of Illinois, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois 61801, United States
- Center for Bio-physics and Quantitative Biology, University of Illinois, Urbana, Illinois 61801, United States
| | - Guy Lippens
- Toulouse Biotechnology Institute, Toulouse 31400, France
| | - Marina Serebryakova
- A.N. Belozersky Institute of Physicochemical Biology MSU, Moscow 119992, Russia
| | - Sergei Borukhov
- Department of Molecular Biology, Virtua Health College of Medicine and Life Sciences, Rowan University School of Osteopathic Medicine institution, Stratford, New Jersey 08084-1501, United States
| | | |
Collapse
|
3
|
Smith NT, Boukherissa A, Antaya K, Howe GW, Mergaert P, Rodríguez de la Vega RC, Shykoff JA, Alunni B, diCenzo GC. Taxonomic distribution of SbmA/BacA and BacA-like antimicrobial peptide transporters suggests independent recruitment and convergent evolution in host-microbe interactions. Microb Genom 2025; 11:001380. [PMID: 40238647 PMCID: PMC12003926 DOI: 10.1099/mgen.0.001380] [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: 10/15/2024] [Accepted: 02/13/2025] [Indexed: 04/18/2025] Open
Abstract
Antimicrobial peptides (AMPs) are often produced by eukaryotes to control bacterial populations in both pathogenic and mutualistic symbioses. Several pathogens and nitrogen-fixing legume symbionts depend on transporters called SbmA (or BacA) or BclA (BacA-like) to survive exposure to AMPs. However, how broadly these transporters are distributed amongst bacteria, and their evolutionary history, is poorly understood. We used computational approaches, including phylogenetic and sequence similarity analyses, to examine the distribution of SbmA/BacA and BclA proteins across 1,255 species spanning the domain Bacteria, leading to the identification of 71 and 177 SbmA/BacA and BclA proteins, respectively. In vitro sensitivity assays using legume AMPs and several BclA proteins confirmed that AMP transport is a common feature of BclA homologues. Our analyses indicated that SbmA/BacA homologues are encoded only by species in the phylum Pseudomonadota and are primarily found in just two orders: Hyphomicrobiales and Enterobacterales. BclA homologues are somewhat more broadly distributed and were found in clusters across four phyla. These included several orders of the phyla Pseudomonadota and Cyanobacteriota, the order Mycobacteriales (phylum Actinomycetota) and the class Negativicutes (phylum Bacillota). Many of the clades enriched for species encoding SbmA/BacA or BclA homologues are rich in species that interact with eukaryotic hosts in mutualistic or pathogenic interactions. These observations suggest that SbmA/BacA and BclA proteins have been repeatedly co-opted to facilitate associations with eukaryotic hosts by allowing bacteria to cope with host-encoded AMPs.
Collapse
Affiliation(s)
- Nicholas T. Smith
- Department of Biology, Queen’s University, Kingston, ON, K7L 3N6, Canada
- Department of Chemistry, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Amira Boukherissa
- Institute for Integrative Biology of the Cell, CNRS, CEA, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
- Écologie Systématique et Évolution, Université Paris-Saclay, CNRS, AgroParisTech, 91198, Gif-sur-Yvette, France
| | - Kiera Antaya
- Department of Biology, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Graeme W. Howe
- Department of Chemistry, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Peter Mergaert
- Institute for Integrative Biology of the Cell, CNRS, CEA, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
| | | | - Jacqui A. Shykoff
- Écologie Systématique et Évolution, Université Paris-Saclay, CNRS, AgroParisTech, 91198, Gif-sur-Yvette, France
| | - Benoît Alunni
- Institute for Integrative Biology of the Cell, CNRS, CEA, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin for Plant Sciences (IJPB), 78000, Versailles, France
| | - George C. diCenzo
- Department of Biology, Queen’s University, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
4
|
Knoke LR, Muskietorz M, Kühn L, Leichert LI. The ABC transporter Opp imports reduced glutathione, while Gsi imports glutathione disulfide in Escherichia coli. Redox Biol 2025; 79:103453. [PMID: 39689618 PMCID: PMC11719327 DOI: 10.1016/j.redox.2024.103453] [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: 10/08/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/19/2024] Open
Abstract
Glutathione is the major thiol-based antioxidant in a wide variety of biological systems, ranging from bacteria to eukaryotes. As a redox couple, consisting of reduced glutathione (GSH) and its oxidized form, glutathione disulfide (GSSG), it is crucial for the maintenance of the cellular redox balance. Glutathione transport out of and into cellular compartments and the extracellular space is a determinant of the thiol-disulfide redox state of the organelles and bodily fluids in question, but is currently not well understood. Here we use the genetically-encoded, glutathione-measuring redox probe Grx1-roGFP2 to comprehensively elucidate the import of extracellular glutathione into the cytoplasm of the model organism Escherichia coli. The elimination of only two ATP-Binding Cassette (ABC) transporter systems, Gsi and Opp, completely abrogates glutathione import into E. coli's cytoplasm, both in its reduced and oxidized form. The lack of only one of them, Gsi, completely prevents import of GSSG, while the lack of the other, Opp, substantially retards the uptake of reduced glutathione (GSH).
Collapse
Affiliation(s)
- Lisa R Knoke
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry - Microbial Biochemistry, Bochum, Germany
| | - Maik Muskietorz
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry - Microbial Biochemistry, Bochum, Germany
| | - Lena Kühn
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry - Microbial Biochemistry, Bochum, Germany
| | - Lars I Leichert
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry - Microbial Biochemistry, Bochum, Germany.
| |
Collapse
|
5
|
Luo S, Li XR, Gong XT, Kulikovsky A, Qu F, Beis K, Severinov K, Dubiley S, Feng X, Dong SH, Nair SK. Trojan horse peptide conjugates remodel the activity spectrum of clinical antibiotics. Proc Natl Acad Sci U S A 2025; 122:e2319483121. [PMID: 39739799 PMCID: PMC11725936 DOI: 10.1073/pnas.2319483121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/16/2024] [Indexed: 01/02/2025] Open
Abstract
Infections caused by gram-negative pathogens continue to be a major risk to human health because of the innate antibiotic resistance endowed by their unique cell membrane architecture. Nature has developed an elegant solution to target gram-negative strains, namely by conjugating toxic antibiotic warheads to a suitable carrier to facilitate the active import of the drug to a specific target organism. Microcin C7 (McC) is a Trojan horse peptide-conjugated antibiotic that specifically targets enterobacteria by exploiting active import through oligopeptide transport systems. Here, we characterize the molecular mechanism of McC recognition by YejA, the solute binding protein of the Escherichia coli oligopeptide transporter. Structure-guided mutational and functional analysis elucidates the determinants of substrate recognition. We demonstrate that the peptide carrier can serve as a passport for the entry of molecules that are otherwise not taken into E. coli cells. We show that peptide conjugation can remodel the antibiotic spectrum of clinically relevant parent compounds. Bioinformatics analysis reveals a broad distribution of YejA-like transporters in only the Proteobacteria, underscoring the potential for the development of Trojan horse antibiotics that are actively imported into such gram-negative bacteria.
Collapse
Affiliation(s)
- Shangwen Luo
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou730000, People’s Republic of China
| | - Xin-Rong Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou730000, People’s Republic of China
| | - Xiao-Tong Gong
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou730000, People’s Republic of China
| | - Alexey Kulikovsky
- Institute of Gene Biology, Russian Academy of Sciences, Moscow119334, Russia
| | - Feng Qu
- Department of Life Sciences, Imperial College London, LondonSW7 2AZ, United Kingdom
- Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot, OxfordshireOX11 OFA, United Kingdom
| | - Konstantinos Beis
- Department of Life Sciences, Imperial College London, LondonSW7 2AZ, United Kingdom
- Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot, OxfordshireOX11 OFA, United Kingdom
| | - Konstantin Severinov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow119334, Russia
- Waksman Institute for Microbiology, Rutgers University, Piscataway, NJ08901
| | - Svetlana Dubiley
- Institute of Gene Biology, Russian Academy of Sciences, Moscow119334, Russia
| | - Xinxin Feng
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan410082, People’s Republic of China
- School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan410082, People’s Republic of China
| | - Shi-Hui Dong
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou730000, People’s Republic of China
| | - Satish K. Nair
- Department of Biochemistry, University of Illinois, Urbana, IL61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois, Urbana, IL61801
| |
Collapse
|
6
|
Farhana A, Alsrhani A, Ejaz H, Alruwaili M, Alameen AAM, Manni E, Rasheed Z, Khan YS. Substrate-Induced Structural Dynamics and Evolutionary Linkage of Siderophore-Iron ABC Transporters of Mycobacterium tuberculosis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1891. [PMID: 39597076 PMCID: PMC11596928 DOI: 10.3390/medicina60111891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
Background and Objective: ATP-binding cassette (ABC) transporters are prominent drug targets due to their highly efficient trafficking capabilities and their significant physiological and clinical roles. Gaining insight into their biophysical and biomechanistic properties is crucial to maximize their pharmacological potential. Materials and Methods: In this study, we present the biochemical and biophysical characterization, and phylogenetic analysis of the domains of Mycobacterium tuberculosis (M. tuberculosis) ABC transporters: the exporter Rv1348 (IrtA) and the importer system Rv1349-Rv2895c (IrtB-Rv2895c), both involved in siderophore-mediated iron uptake. Results: Our findings reveal that the substrate-binding domain (SBD) of IrtA functions as an active monomer, while Rv2895c, which facilitates the uptake of siderophore-bound iron, exists in a dynamic equilibrium between dimeric and monomeric forms. Furthermore, ATP binding induces the dimerization of the ATPase domains in both IrtA (ATPase I) and IrtB (ATPaseII), but only the ATPase domain of IrtA (ATPase I) is active independently. We also analyzed the stability of substrate binding to the domains of the two transporters across varying temperature and pH ranges, revealing significant shifts in their activity under different conditions. Our study highlights the conformational changes that accompany substrate interaction with the transporter domains, providing insights into the fundamental mechanism required for the translocation of siderophore to the extracytoplasmic milieu by IrtB and, subsequently, import of their ferrated forms by the IrtB-Rv2895c complex. Phylogenetic analyses based on ATPase domains reveal that IrtA shares features with both archaeal and eukaryotic transporters, while IrtB is unique to mycobacterial species. Conclusions: Together, these findings provide valuable insights, which could accelerate the development of intervention strategies for this critical pathway pivotal in the progression of M. tuberculosis infection.
Collapse
Affiliation(s)
- Aisha Farhana
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia; (A.A.); (H.E.); (M.A.); (A.A.M.A.); (E.M.)
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia; (A.A.); (H.E.); (M.A.); (A.A.M.A.); (E.M.)
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia; (A.A.); (H.E.); (M.A.); (A.A.M.A.); (E.M.)
| | - Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia; (A.A.); (H.E.); (M.A.); (A.A.M.A.); (E.M.)
| | - Ayman A. M. Alameen
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia; (A.A.); (H.E.); (M.A.); (A.A.M.A.); (E.M.)
| | - Emad Manni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia; (A.A.); (H.E.); (M.A.); (A.A.M.A.); (E.M.)
| | - Zafar Rasheed
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Qassim, Saudi Arabia;
| | - Yusuf Saleem Khan
- Department of Anatomy, College of Medicine, University of Hail, Hail 55476, Hail, Saudi Arabia
| |
Collapse
|
7
|
Wang M, Wu M, Han M, Niu X, Fan A, Zhu S, Tong Y. Mining the Biosynthetic Landscape of Lactic Acid Bacteria Unearths a New Family of RiPPs Assembled by a Novel Type of ThiF-like Adenylyltransferases. ACS OMEGA 2024; 9:30891-30903. [PMID: 39035879 PMCID: PMC11256085 DOI: 10.1021/acsomega.4c03760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
Ribosomally synthesized and post-translationally modified peptides (RiPPs) are chemically diverse natural products of ribosomal origin. These peptides, which frequently act as signals or antimicrobials, are biosynthesized by conserved enzymatic machinery, making genome mining a powerful strategy for unearthing previously uncharacterized members of their class. Herein, we investigate the untapped biosynthetic potential of Lactobacillales (i.e., lactic acid bacteria), an order of Gram-positive bacteria closely associated with human life, including pathogenic species and industrially relevant fermenters of dairy products. Through genome mining methods, we systematically explored the distribution and diversity of ThiF-like adenylyltransferase-utilizing RiPP systems in lactic acid bacteria and identified a number of unprecedented biosynthetic gene clusters. In one of these clusters, we found a previously undescribed group of macrocyclic imide biosynthetic pathways containing multiple transporters that may be involved in a potential quorum sensing (QS) system. Through in vitro assays, we determined that one such adenylyltransferase specifically catalyzes the intracyclization of its precursor peptide through macrocyclic imide formation. Incubating the enzyme with various primary amines revealed that it could effectively amidate the C-terminus of the precursor peptide. This new transformation adds to the growing list of Nature's peptide macrocyclization strategies and expands the impressive catalytic repertoire of the adenylyltransferase family. The diverse RiPP systems identified herein represent a vast, unexploited landscape for the discovery of a novel class of natural products and QS systems.
Collapse
Affiliation(s)
- Mengjiao Wang
- College
of Life Science and Technology, Beijing
University of Chemical Technology, Beijing 100029, People’s Republic of China
| | - Mengyue Wu
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, People’s
Republic of China
| | - Meng Han
- MOE
Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Xiaogang Niu
- Beijing
Nuclear Magnetic Resonance Center, College of Chemistry and Molecular
Engineering, Peking University, Beijing 100871, People’s Republic of China
| | - Aili Fan
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, People’s
Republic of China
| | - Shaozhou Zhu
- National
Institutes for Food and Drug Control, Beijing 102629, People’s Republic of China
| | - Yigang Tong
- College
of Life Science and Technology, Beijing
University of Chemical Technology, Beijing 100029, People’s Republic of China
| |
Collapse
|
8
|
Yang F, Yang F, Huang J, Yu H, Qiao S. Microcin C7 as a Potential Antibacterial-Immunomodulatory Agent in the Postantibiotic Era: Overview of Its Bioactivity Aspects and Applications. Int J Mol Sci 2024; 25:7213. [PMID: 39000321 PMCID: PMC11241378 DOI: 10.3390/ijms25137213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
In the postantibiotic era, the pathogenicity and resistance of pathogens have increased, leading to an increase in intestinal inflammatory disease. Bacterial infections remain the leading cause of animal mortality. With increasing resistance to antibiotics, there has been a significant decrease in resistance to both inflammation and disease in animals, thus decreasing production efficiency and increasing production costs. These side effects have serious consequences and have detracted from the development of China's pig industry. Microcin C7 (McC7) demonstrates potent antibacterial activity against a broad spectrum of pathogens, stable physicochemical properties, and low toxicity, reducing the likelihood of resistance development. Thus, McC7 has received increasing attention as a potential clinical antibacterial and immunomodulatory agent. McC7 has the potential to serve as a new generation of antibiotic substitutes; however, its commercial applications in the livestock and poultry industry have been limited. In this review, we summarize and discuss the biosynthesis, biochemical properties, structural characteristics, mechanism of action, and immune strategies of McC7. We also describe the ability of McC7 to improve intestinal health. Our aim in this study was to provide a theoretical basis for the application of McC7 as a new feed additive or new veterinary drug in the livestock and poultry breeding industry, thus providing a new strategy for alleviating resistance through feed and mitigating drug resistance. Furthermore, this review provides insight into the new functions and anti-infection mechanisms of bacteriocin peptides and proposes crucial ideas for the research, product development, and application of bacteriocin peptides in different fields, such as the food and medical industries.
Collapse
Affiliation(s)
- Fengjuan Yang
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Centre, China Agricultural University, Beijing 100193, China
- Beijing Biofeed Additives Key Laboratory, Beijing 100193, China
| | - Feiyun Yang
- Chongqing Academy of Animal Science, Rongchang, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Jinxiu Huang
- Chongqing Academy of Animal Science, Rongchang, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Centre, China Agricultural University, Beijing 100193, China
- Beijing Biofeed Additives Key Laboratory, Beijing 100193, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Centre, China Agricultural University, Beijing 100193, China
- Beijing Biofeed Additives Key Laboratory, Beijing 100193, China
| |
Collapse
|
9
|
Frazier CL, Deb D, Weeks AM. Engineered reactivity of a bacterial E1-like enzyme enables ATP-driven modification of protein C termini. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593989. [PMID: 38798401 PMCID: PMC11118369 DOI: 10.1101/2024.05.13.593989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
In biological systems, ATP provides an energetic driving force for peptide bond formation, but protein chemists lack tools that emulate this strategy. Inspired by the eukaryotic ubiquitination cascade, we developed an ATP-driven platform for C-terminal activation and peptide ligation based on E. coli MccB, a bacterial ancestor of ubiquitin-activating (E1) enzymes that natively catalyzes C-terminal phosphoramidate bond formation. We show that MccB can act on non-native substrates to generate an O-AMPylated electrophile that can react with exogenous nucleophiles to form diverse C-terminal functional groups including thioesters, a versatile class of biological intermediates that have been exploited for protein semisynthesis. To direct this activity towards specific proteins of interest, we developed the Thioesterification C-terminal Handle (TeCH)-tag, a sequence that enables high-yield, ATP-driven protein bioconjugation via a thioester intermediate. By mining the natural diversity of the MccB family, we developed two additional MccB/TeCH-tag pairs that are mutually orthogonal to each other and to the E. coli system, facilitating the synthesis of more complex bioconjugates. Our method mimics the chemical logic of peptide bond synthesis that is widespread in biology for high-yield in vitro manipulation of protein structure with molecular precision.
Collapse
Affiliation(s)
- Clara L. Frazier
- Department of Biochemistry, University of Wisconsin – Madison, Madison, WI, USA 53706
| | - Debashrito Deb
- Department of Biochemistry, University of Wisconsin – Madison, Madison, WI, USA 53706
| | - Amy M. Weeks
- Department of Biochemistry, University of Wisconsin – Madison, Madison, WI, USA 53706
- Department of Chemistry, University of Wisconsin – Madison, Madison, Wisconsin 53706
| |
Collapse
|
10
|
Ackroyd BK, Dodson EJ, Mehboob J, Dowle AA, Thomas GH, Wilkinson AJ. Structure and ligand binding in the putative anti-microbial peptide transporter protein, YejA. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001430. [PMID: 38334478 PMCID: PMC10924461 DOI: 10.1099/mic.0.001430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
YejABEF is an ATP-binding cassette transporter that is implicated in the sensitivity of Escherichia coli to anti-microbial peptides, the best-characterized example being microcin C, a peptide-nucleotide antibiotic that targets aspartyl-tRNA synthetase. Here the structure of the extracellular solute binding protein, YejA, has been determined, revealing an oligopeptide-binding protein fold enclosing a ligand-binding pocket larger than those of other peptide-binding proteins of known structure. Prominent electron density in this cavity defines an undecapeptide sequence LGEPRYAFNFN, an observation that is confirmed by mass spectrometry. In the structure, the peptide interactions with the protein are mediated by main chain hydrogen bonds with the exception of Arg5 whose guanidinium side chain makes a set of defining polar interactions with four YejA residues. More detailed characterization of purified recombinant YejA, by a combination of ESI and MALDI-mass spectrometry as well as thermal shift assays, reveals a set of YejA complexes containing overlapping peptides 10-19 residues in length. All contain the sequence LGEPRYAFN. Curiously, these peptides correspond to residues 8-26 of the mature YejA protein, which belong to a unique N-terminal extension that distinguishes YejA from other cluster C oligopeptide binding proteins of known structure. This 35-residue extension is well-ordered and packs across the surface of the protein. The undecapeptide ligand occupies only a fraction of the enclosed pocket volume suggesting the possibility that much larger peptides or peptide conjugates could be accommodated, though thermal shift assays of YejA binding to antimicrobial peptides and peptides unrelated to LGEPRYAFNFN have not provided evidence of binding. While the physiological significance of this 'auto-binding' is not clear, the experimental data suggest that it is not an artefact of the crystallization process and that it may have a function in the sensing of periplasmic or membrane stress.
Collapse
Affiliation(s)
- Bryony K. Ackroyd
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
- Department of Biology and York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Eleanor J. Dodson
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
| | - Javeria Mehboob
- Department of Biology and York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Adam A. Dowle
- Bioscience Technology Facility, Department of Biology, University of York, York YO10 5DD, UK
| | - Gavin H. Thomas
- Department of Biology and York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Anthony J. Wilkinson
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
| |
Collapse
|
11
|
Yu H, Lin J, Wang M, Ying S, Yuan S, Guo Y, Xie Y, Yao W. Molecular and proteomic response of Pseudomonas fluorescens biofilm cultured on lettuce (Lactuca sativa L.) after ultrasound treatment at different intensity levels. Food Microbiol 2024; 117:104387. [PMID: 37919011 DOI: 10.1016/j.fm.2023.104387] [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: 08/07/2023] [Accepted: 09/17/2023] [Indexed: 11/04/2023]
Abstract
Ultrasonic treatment is widely used for surface cleaning of vegetables in the processing of agricultural products. In the present study, the molecular and proteomic response of Pseudomonas fluorescens biofilm cultured on lettuce was investigated after ultrasound treatment at different intensity levels. The results show that the biofilm was efficiently removed after ultrasound treatment with intensity higher than 21.06 W/cm2. However, at an intensity of less than 18.42 W/cm2, P. fluorescens was stimulated by ultrasound leading to promoted bacterial growth, extracellular protease activity, extracellular polysaccharide secretion (EPS), and synthesis of acyl-homoserine lactones (AHLs) as quorum-sensing signaling molecules. The expression of biofilm-related genes, stress response, and dual quorum sensing system was upregulated during post-treatment ultrasound. Proteomic analysis showed that ultrasound activated proteins in the flagellar system, which led to changes in bacterial tendency; meanwhile, a large number of proteins in the dual-component system began to be regulated. ABC transporters accelerated the membrane transport of substances inside and outside the cell membrane and equalized the permeability conditions of the cell membrane. In addition, the expression of proteins related to DNA repair was upregulated, suggesting that bacteria repair damaged DNA after ultrasound exposure.
Collapse
Affiliation(s)
- Hang Yu
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China.
| | - Jiang Lin
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China
| | - Mengru Wang
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China
| | - Su Ying
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China
| | - Shaofeng Yuan
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China
| | - Yahui Guo
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China
| | - Yunfei Xie
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China
| | - Weirong Yao
- State Key Laboratory of Food Science and Resource, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No.1800 Lihu Avenue, Wuxi, Jiangsu Province, 214122, China.
| |
Collapse
|
12
|
Yang G, Shang L, Liu L, Li Z, Zeng X, Ding X, Huang J, Qiao S, Yu H. Engineering and Purification of Microcin C7 Variants Resistant to Trypsin and Analysis of Their Biological Activity. Antibiotics (Basel) 2023; 12:1346. [PMID: 37760643 PMCID: PMC10525924 DOI: 10.3390/antibiotics12091346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Microcin C7 (McC) as a viable form of antimicrobial has gained substantial attention due to its distinctive antimicrobial activity, by targeting aspartyl tRNA synthetase. McC can be a potential solution against pathogenic microbial infections in the postantibiotic era. However, considering that degradation by digestive enzymes can disrupt the function of this peptide in the gastrointestinal tract, in this study, we attempt to design McC variants to overcome several barriers that may affect its stability and biological activity. The mccA gene encoding the McC peptide precursor was mutated and 12 new McC variants with trypsin resistance were found. The Yej+rimL- strain was used as an indicator to determine the minimum inhibitory concentrations (MICs). The results showed that three variants, including R2A, R2T and R2Q, among 12 variants formed by the replacement of the second arginine of the McC peptide with different amino acids, were resistant to trypsin and had an outstanding antimicrobial ability, with MIC values of 12.5, 25, and 25 μg/mL, respectively. Taken together, our findings show that the engineering of the site-directed mutagenesis of McC significantly enhances McC trypsin resistance and maintains a great antimicrobial activity.
Collapse
Affiliation(s)
- Guangxin Yang
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture Rural Affairs Feed Industry Centre, China Agricultural University, Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China (L.S.); (L.L.); (S.Q.)
| | - Lijun Shang
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture Rural Affairs Feed Industry Centre, China Agricultural University, Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China (L.S.); (L.L.); (S.Q.)
| | - Lu Liu
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture Rural Affairs Feed Industry Centre, China Agricultural University, Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China (L.S.); (L.L.); (S.Q.)
- Luzhou Modern Agriculture Development Promotion Center, Luzhou 646000, China
| | - Zeqiang Li
- Shanghai Menon Animal Nutrition Technology Co., Ltd., Shanghai 201807, China;
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture Rural Affairs Feed Industry Centre, China Agricultural University, Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China (L.S.); (L.L.); (S.Q.)
| | - Xiuliang Ding
- Chongqing Academy of Animal Science, Rongchang, Chongqing 402460, China; (X.D.); (J.H.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Jinxiu Huang
- Chongqing Academy of Animal Science, Rongchang, Chongqing 402460, China; (X.D.); (J.H.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture Rural Affairs Feed Industry Centre, China Agricultural University, Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China (L.S.); (L.L.); (S.Q.)
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture Rural Affairs Feed Industry Centre, China Agricultural University, Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China (L.S.); (L.L.); (S.Q.)
| |
Collapse
|
13
|
Dual-Uptake Mode of the Antibiotic Phazolicin Prevents Resistance Acquisition by Gram-Negative Bacteria. mBio 2023; 14:e0021723. [PMID: 36802165 PMCID: PMC10128002 DOI: 10.1128/mbio.00217-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Phazolicin (PHZ) is a peptide antibiotic exhibiting narrow-spectrum activity against rhizobia closely related to its producer, Rhizobium sp. strain Pop5. Here, we show that the frequency of spontaneous PHZ-resistant mutants in Sinorhizobium meliloti is below the detection limit. We find that PHZ can enter S. meliloti cells through two distinct promiscuous peptide transporters, BacA and YejABEF, which belong to the SLiPT (SbmA-like peptide transporter) and ABC (ATP-binding cassette) transporter families, respectively. The dual-uptake mode explains the lack of observed resistance acquisition because the simultaneous inactivation of both transporters is necessary for resistance to PHZ. Since both BacA and YejABEF are essential for the development of functional symbiosis of S. meliloti with leguminous plants, the unlikely acquisition of PHZ resistance via the inactivation of these transporters is further disfavored. A whole-genome transposon sequencing screen did not reveal additional genes that can provide strong PHZ resistance when inactivated. However, it was found that the capsular polysaccharide KPS, the novel putative envelope polysaccharide PPP (PHZ-protecting polysaccharide), as well as the peptidoglycan layer jointly contribute to the sensitivity of S. meliloti to PHZ, most likely serving as barriers that reduce the amount of PHZ transported inside the cell. IMPORTANCE Many bacteria produce antimicrobial peptides to eliminate competitors and create an exclusive niche. These peptides act either by membrane disruption or by inhibiting essential intracellular processes. The Achilles' heel of the latter type of antimicrobials is their dependence on transporters to enter susceptible cells. Transporter inactivation results in resistance. Here, we show that a rhizobial ribosome-targeting peptide, phazolicin (PHZ), uses two different transporters, BacA and YejABEF, to enter the cells of a symbiotic bacterium, Sinorhizobium meliloti. This dual-entry mode dramatically reduces the probability of the appearance of PHZ-resistant mutants. Since these transporters are also crucial for S. meliloti symbiotic associations with host plants, their inactivation in natural settings is strongly disfavored, making PHZ an attractive lead for the development of biocontrol agents for agriculture.
Collapse
|
14
|
Peptidoglycan recycling mediated by an ABC transporter in the plant pathogen Agrobacterium tumefaciens. Nat Commun 2022; 13:7927. [PMID: 36566216 PMCID: PMC9790009 DOI: 10.1038/s41467-022-35607-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022] Open
Abstract
During growth and division, the bacterial cell wall peptidoglycan (PG) is remodelled, resulting in the liberation of PG muropeptides which are typically reinternalized and recycled. Bacteria belonging to the Rhizobiales and Rhodobacterales orders of the Alphaproteobacteria lack the muropeptide transporter AmpG, despite having other key PG recycling enzymes. Here, we show that an alternative transporter, YejBEF-YepA, takes over this role in the Rhizobiales phytopathogen Agrobacterium tumefaciens. Muropeptide import by YejBEF-YepA governs expression of the β-lactamase AmpC in A. tumefaciens, contributing to β-lactam resistance. However, we show that the absence of YejBEF-YepA causes severe cell wall defects that go far beyond lowered AmpC activity. Thus, contrary to previously established Gram-negative models, PG recycling is vital for cell wall integrity in A. tumefaciens. YepA is widespread in the Rhizobiales and Rhodobacterales, suggesting that YejBEF-YepA-mediated PG recycling could represent an important but overlooked aspect of cell wall biology in these bacteria.
Collapse
|
15
|
Couturier C, Groß S, von Tesmar A, Hoffmann J, Deckarm S, Fievet A, Dubarry N, Taillier T, Pöverlein C, Stump H, Kurz M, Toti L, Haag Richter S, Schummer D, Sizun P, Hoffmann M, Prasad Awal R, Zaburannyi N, Harmrolfs K, Wink J, Lessoud E, Vermat T, Cazals V, Silve S, Bauer A, Mourez M, Fraisse L, Leroi‐Geissler C, Rey A, Versluys S, Bacqué E, Müller R, Renard S. Structure Elucidation, Total Synthesis, Antibacterial In Vivo Efficacy and Biosynthesis Proposal of Myxobacterial Corramycin. Angew Chem Int Ed Engl 2022; 61:e202210747. [PMID: 36197755 PMCID: PMC10099666 DOI: 10.1002/anie.202210747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Indexed: 11/22/2022]
Abstract
Herein, we describe the myxobacterial natural product Corramycin isolated from Corallococcus coralloides. The linear peptide structure contains an unprecedented (2R,3S)-γ-N-methyl-β-hydroxy-histidine moiety. Corramycin exhibits anti-Gram-negative activity against Escherichia coli (E. coli) and is taken up via two transporter systems, SbmA and YejABEF. Furthermore, the Corramycin biosynthetic gene cluster (BGC) was identified and a biosynthesis model was proposed involving a 12-modular non-ribosomal peptide synthetase/polyketide synthase. Bioinformatic analysis of the BGC combined with the development of a total synthesis route allowed for the elucidation of the molecule's absolute configuration. Importantly, intravenous administration of 20 mg kg-1 of Corramycin in an E. coli mouse infection model resulted in 100 % survival of animals without toxic side effects. Corramycin is thus a promising starting point to develop a potent antibacterial drug against hospital-acquired infections.
Collapse
Affiliation(s)
| | - Sebastian Groß
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Alexander von Tesmar
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Judith Hoffmann
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Selina Deckarm
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | | | | | | | | | - Heike Stump
- Sanofi13, Quai Jules Guesde94400Vitry-sur-SeineFrance
| | - Michael Kurz
- Sanofi13, Quai Jules Guesde94400Vitry-sur-SeineFrance
| | - Luigi Toti
- Sanofi13, Quai Jules Guesde94400Vitry-sur-SeineFrance
| | | | - Dietmar Schummer
- Technische Hochschule MittelhessenWiesenstraße 1435390GießenGermany
| | | | - Michael Hoffmann
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Ram Prasad Awal
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Nestor Zaburannyi
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Kirsten Harmrolfs
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Joachim Wink
- Mikrobielle StammsammlungHelmholtz Centre for Infection Research (HZI)Inhoffenstraße 738124BraunschweigGermany
| | - Emilie Lessoud
- Evotec1541, avenue Marcel Mérieux69280Marcy L'EtoileFrance
| | - Thierry Vermat
- Evotec1541, avenue Marcel Mérieux69280Marcy L'EtoileFrance
| | | | - Sandra Silve
- Evotec1541, avenue Marcel Mérieux69280Marcy L'EtoileFrance
| | - Armin Bauer
- Sanofi13, Quai Jules Guesde94400Vitry-sur-SeineFrance
| | - Michael Mourez
- Ecole d'Ingénieurs de Purpan75 voie du ToecBP57611, 31076ToulouseFrance
| | - Laurent Fraisse
- Drug for Neglected Diseases InitiativeChemin Camille-Vidart 151202GenevaSwitzerland
| | | | - Astrid Rey
- Charles River Laboratories327 impasse du domaine rozier69210Saint-Germain-NuellesFrance
| | | | - Eric Bacqué
- Evotec1541, avenue Marcel Mérieux69280Marcy L'EtoileFrance
| | - Rolf Müller
- Microbial Natural ProductsHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland UniversityCampus Building E8.166123SaarbrückenGermany
| | | |
Collapse
|
16
|
Ongpipattanakul C, Desormeaux EK, DiCaprio A, van der Donk WA, Mitchell DA, Nair SK. Mechanism of Action of Ribosomally Synthesized and Post-Translationally Modified Peptides. Chem Rev 2022; 122:14722-14814. [PMID: 36049139 PMCID: PMC9897510 DOI: 10.1021/acs.chemrev.2c00210] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ribosomally synthesized and post-translationally modified peptides (RiPPs) are a natural product class that has undergone significant expansion due to the rapid growth in genome sequencing data and recognition that they are made by biosynthetic pathways that share many characteristic features. Their mode of actions cover a wide range of biological processes and include binding to membranes, receptors, enzymes, lipids, RNA, and metals as well as use as cofactors and signaling molecules. This review covers the currently known modes of action (MOA) of RiPPs. In turn, the mechanisms by which these molecules interact with their natural targets provide a rich set of molecular paradigms that can be used for the design or evolution of new or improved activities given the relative ease of engineering RiPPs. In this review, coverage is limited to RiPPs originating from bacteria.
Collapse
Affiliation(s)
- Chayanid Ongpipattanakul
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Emily K. Desormeaux
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Adam DiCaprio
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Wilfred A. van der Donk
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| | - Douglas A. Mitchell
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Microbiology, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| | - Satish K. Nair
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| |
Collapse
|
17
|
Akhtar AA, Turner DP. The role of bacterial ATP-binding cassette (ABC) transporters in pathogenesis and virulence: Therapeutic and vaccine potential. Microb Pathog 2022; 171:105734. [PMID: 36007845 DOI: 10.1016/j.micpath.2022.105734] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
The ATP-binding cassette (ABC) transporter superfamily is found in all domains of life, facilitating critical biological processes through the translocation of a wide variety of substrates from, ions to proteins, across cellular membranes in an ATP-coupled process. The role of ABC transporters in eukaryotes has been well established: the facilitation of genetic diseases and multi-drug resistance (MDR) in cancer patients. In contrast, the role of ABC transporters in prokaryotes has been ambiguous due to their diverse functions and the sheer number of organisms in which they reside. This review examines the role of bacterial ABC transporters in pathogenesis and virulence, and their potential for therapeutic and vaccine application. We demonstrate how ABC transporters play a vital role in the virulence and pathogenesis of several pathogenic bacteria through the import of essential molecules, such as metal ions, amino acids, peptides, vitamins and osmoprotectants, as well as, the export of virulent determinants involved in glycoconjugate biosynthesis and Type I secretion. Furthermore, ABC exporters facilitate the persistence of pathogenic bacteria through the export of toxic xenobiotic substances, thus, contributing to the development of antimicrobial resistance. We also show that ABC transporters display considerable potential for therapeutic application through immunisation and resistance reversal. In conclusion, bacterial ABC transporters play an immense role in virulence and pathogenesis and display desirable traits for clinical use, therefore, potentially aiding in the battle against MDR.
Collapse
Affiliation(s)
- Armaan A Akhtar
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom.
| | - David Pj Turner
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
18
|
S51 Family Peptidases Provide Resistance to Peptidyl-Nucleotide Antibiotic McC. mBio 2022; 13:e0080522. [PMID: 35467414 PMCID: PMC9239234 DOI: 10.1128/mbio.00805-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Microcin C (McC)-like compounds are natural Trojan horse peptide-nucleotide antibiotics produced by diverse bacteria. The ribosomally synthesized peptide parts of these antibiotics are responsible for their facilitated transport into susceptible cells. Once inside the cell, the peptide part is degraded, releasing the toxic payload, an isoaspartyl-nucleotide that inhibits aspartyl-tRNA synthetase, an enzyme essential for protein synthesis. Bacteria that produce microcin C-like compounds have evolved multiple ways to avoid self-intoxication. Here, we describe a new strategy through the action of S51 family peptidases, which we name MccG. MccG cleaves the toxic isoaspartyl-nucleotide, rendering it inactive. While some MccG homologs are encoded by gene clusters responsible for biosynthesis of McC-like compounds, most are encoded by standalone genes whose products may provide a basal level of resistance to peptide-nucleotide antibiotics in phylogenetically distant bacteria.
Collapse
|
19
|
Parker JK, Davies BW. Microcins reveal natural mechanisms of bacterial manipulation to inform therapeutic development. MICROBIOLOGY (READING, ENGLAND) 2022; 168:001175. [PMID: 35438625 PMCID: PMC10233263 DOI: 10.1099/mic.0.001175] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/17/2022] [Indexed: 12/20/2022]
Abstract
Microcins are an understudied and poorly characterized class of antimicrobial peptides. Despite the existence of only 15 examples, all identified from the Enterobacteriaceae, microcins display diversity in sequence, structure, target cell uptake, cytotoxic mechanism of action and target specificity. Collectively, these features describe some of the unique means nature has contrived for molecules to cross the 'impermeable' barrier of the Gram-negative bacterial outer membrane and inflict cytotoxic effects. Microcins appear to be widely dispersed among different species and in different environments, where they function in regulating microbial communities in diverse ways, including through competition. Growing evidence suggests that microcins may be adapted for therapeutic uses such as antimicrobial drugs, microbiome modulators or facilitators of peptide uptake into cells. Advancing our biological, ecological and biochemical understanding of the roles of microcins in bacterial interactions, and learning how to regulate and modify microcin activity, is essential to enable such therapeutic applications.
Collapse
Affiliation(s)
| | - Bryan William Davies
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- John Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
20
|
Bauman KD, Butler KS, Moore BS, Chekan JR. Genome mining methods to discover bioactive natural products. Nat Prod Rep 2021; 38:2100-2129. [PMID: 34734626 PMCID: PMC8597713 DOI: 10.1039/d1np00032b] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Indexed: 12/22/2022]
Abstract
Covering: 2016 to 2021With genetic information available for hundreds of thousands of organisms in publicly accessible databases, scientists have an unprecedented opportunity to meticulously survey the diversity and inner workings of life. The natural product research community has harnessed this breadth of sequence information to mine microbes, plants, and animals for biosynthetic enzymes capable of producing bioactive compounds. Several orthogonal genome mining strategies have been developed in recent years to target specific chemical features or biological properties of bioactive molecules using biosynthetic, resistance, or transporter proteins. These "biosynthetic hooks" allow researchers to query for biosynthetic gene clusters with a high probability of encoding previously undiscovered, bioactive compounds. This review highlights recent case studies that feature orthogonal approaches that exploit genomic information to specifically discover bioactive natural products and their gene clusters.
Collapse
Affiliation(s)
- Katherine D Bauman
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Keelie S Butler
- Department of Chemistry and Biochemistry, University of North Carolina Greensboro, Greensboro, NC, 27402, USA.
| | - Bradley S Moore
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jonathan R Chekan
- Department of Chemistry and Biochemistry, University of North Carolina Greensboro, Greensboro, NC, 27402, USA.
| |
Collapse
|
21
|
Fathizadeh H, Pakdel F, Saffari M, Esmaeili DD, Momen-Heravi M, Dao S, Ganbarov K, Kafil HS. Bacteriocins: Recent advances in application as an antimicrobial alternative. Curr Pharm Biotechnol 2021; 23:1028-1040. [PMID: 34493194 DOI: 10.2174/1389201022666210907121254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 11/22/2022]
Abstract
Due to the emergence and development of antibiotic resistance in the treatment of bacterial infections, efforts to discover new antimicrobial agents have increased. One of these antimicrobial agents is a compound produced by a large number of bacteria called bacteriocin. Bacteriocins are small ribosomal polypeptides that can exert their antibacterial effects against bacteria close to their producer strain or even non-closely strains. Adequate knowledge of the structure and functional mechanisms of bacteriocins and their spectrum of activity, as well as knowledge of the mechanisms of possible resistance to these compounds will lead to further development of their use as an alternative to antibiotics. Furthermore, most bacteria that live in the gastrointestinal tract (GIT) have the ability to produce bacteriocins, which spread throughout the GIT. Despite antimicrobial studies in vitro, our knowledge of bacteriocins in the GIT and the migration of these bacteriocins from the epithelial barrier is low. Hence, in this study, we reviewed general information about bacteriocins, such as classification, mechanism of action and resistance, emphasizing their presence, stability, and spectrum of activity in the GIT.
Collapse
Affiliation(s)
- Hadis Fathizadeh
- Department of Microbiology and immunology, Kashan University of Medical Sciences, Kashan. Iran
| | - Farzaneh Pakdel
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Mahmood Saffari
- Department of Microbiology and immunology, Kashan University of Medical Sciences, Kashan. Iran
| | - Davoud Davoud Esmaeili
- Department of Microbiology and Applied Microbiology Research Center, Systems biology and poisonings institute, Baqiyatallah University of Medical sciences, Tehran. Iran
| | - Mansooreh Momen-Heravi
- Infectious Diseases Research Center, Kashan University of Medical Sciences, Kashan. Iran
| | - Sounkalo Dao
- Faculté de Médecine, de Pharmacie et d'Odonto-Stomatologie (FMPOS), University of Bamako, Bamako. Mali
| | | | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, IR. Iran
| |
Collapse
|
22
|
Chan DCK, Burrows LL. Thiocillin and micrococcin exploit the ferrioxamine receptor of Pseudomonas aeruginosa for uptake. J Antimicrob Chemother 2021; 76:2029-2039. [PMID: 33907816 DOI: 10.1093/jac/dkab124] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Thiopeptides are a class of antibiotics that are active against Gram-positive bacteria and inhibit translation. They were considered inactive against Gram-negative bacteria due to their inability to cross the outer membrane. However, we discovered previously that a member of this class, thiostrepton (TS), has activity against Pseudomonas aeruginosa and Acinetobacter baumannii under iron-limiting conditions. TS hijacks the pyoverdine siderophore receptors of P. aeruginosa to cross the outer membrane and synergizes with iron chelators. OBJECTIVES To test other thiopeptides for antimicrobial activity against P. aeruginosa and determine their mechanism of uptake, action and spectrum of activity. METHODS Eight thiopeptides were screened in chequerboard assays against a mutant of P. aeruginosa PA14 lacking both pyoverdine receptors. Thiopeptides that retain activity against a pyoverdine receptor-null mutant may use alternative siderophore receptors for entry. Susceptibility testing against siderophore receptor mutants was used to determine thiopeptide mechanism of uptake. RESULTS The thiopeptides thiocillin (TC) and micrococcin (MC) use the ferrioxamine siderophore receptor (FoxA) for uptake and inhibit the growth of P. aeruginosa at low micromolar concentrations. The activity of TC required the TonB-ExbBD system used to energize siderophore uptake. TC acted through its canonical mechanism of action of translation inhibition. CONCLUSIONS Multiple thiopeptides have antimicrobial activity against P. aeruginosa, countering the historical assumption that they cannot cross the outer membrane. These results demonstrate the potential for thiopeptides to act as antipseudomonal antibiotics.
Collapse
Affiliation(s)
- Derek C K Chan
- Department of Biochemistry and Biomedical Sciences, McMaster Children's Hospital, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Lori L Burrows
- Department of Biochemistry and Biomedical Sciences, McMaster Children's Hospital, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
23
|
Yin CM, Pan XY, Cao XT, Li T, Zhang YH, Lan JF. A crayfish ALF inhibits the proliferation of microbiota by binding to RPS4 and MscL of E. coli. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 121:104106. [PMID: 33878364 DOI: 10.1016/j.dci.2021.104106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 06/12/2023]
Abstract
Antimicrobial peptides (AMPs), most of which are small proteins, are necessary for innate immunity against pathogens. Anti-lipopolysaccharide factor (ALF) with a conserved lipopolysaccharide binding domain (LBD) can bind to lipopolysaccharide (LPS) and neutralize LPS activity. The antibacterial mechanism of ALF, especially its role in bacteria, needs to be further investigated. In this study, the antibacterial role of an anti-lipopolysaccharide factor (PcALF5) derived from Procambarus clarkii was analyzed. PcALF5 could inhibit the replication of the microbiota in vitro and enhance the bacterial clearance ability in crayfish in vivo. Far-western blot assay results indicated that PcALF5 bound to two proteins of E. coli (approximately 25 kDa and 15 kDa). Mass spectrometry (MS), far-western blot assay, and pull-down results showed that 30S ribosomal protein S4 (RPS4, 25 kD) interacted with PcALF5. Further studies revealed that another E. coli protein binding to PcALF5 could be the large mechanosensitive channel (MscL), which is reported to participate in the transport of peptides and antibiotics. Additional assays showed that PcALF5 inhibited protein synthesis and promoted the transcription of ribosomal component genes in E. coli. Overall, these results indicate that PcALF5 could transfer into E. coli by binding to MscL and inhibit protein synthesis by interacting with RPS4. This study reveals the mechanism underlying ALF involvement in the antibacterial immune response and provides a new reference for the research on antibacterial drugs.
Collapse
Affiliation(s)
- Cheng-Ming Yin
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiao-Yi Pan
- Agriculture Ministry Key Laboratory of Healthy Freshwater Aquaculture, Key Laboratory of Freshwater Aquaculture Genetic and Breeding of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou, 313001, China
| | - Xiao-Tong Cao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, 271018, China
| | - Tong Li
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ying-Hao Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiang-Feng Lan
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, 271018, China.
| |
Collapse
|
24
|
Sinorhizobium meliloti Functions Required for Resistance to Antimicrobial NCR Peptides and Bacteroid Differentiation. mBio 2021; 12:e0089521. [PMID: 34311575 PMCID: PMC8406287 DOI: 10.1128/mbio.00895-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Legumes of the Medicago genus have a symbiotic relationship with the bacterium Sinorhizobium meliloti and develop root nodules housing large numbers of intracellular symbionts. Members of the nodule-specific cysteine-rich peptide (NCR) family induce the endosymbionts into a terminal differentiated state. Individual cationic NCRs are antimicrobial peptides that have the capacity to kill the symbiont, but the nodule cell environment prevents killing. Moreover, the bacterial broad-specificity peptide uptake transporter BacA and exopolysaccharides contribute to protect the endosymbionts against the toxic activity of NCRs. Here, we show that other S. meliloti functions participate in the protection of the endosymbionts; these include an additional broad-specificity peptide uptake transporter encoded by the yejABEF genes and lipopolysaccharide modifications mediated by lpsB and lpxXL, as well as rpoH1, encoding a stress sigma factor. Strains with mutations in these genes show a strain-specific increased sensitivity profile against a panel of NCRs and form nodules in which bacteroid differentiation is affected. The lpsB mutant nodule bacteria do not differentiate, the lpxXL and rpoH1 mutants form some seemingly fully differentiated bacteroids, although most of the nodule bacteria are undifferentiated, while the yejABEF mutants form hypertrophied but nitrogen-fixing bacteroids. The nodule bacteria of all the mutants have a strongly enhanced membrane permeability, which is dependent on the transport of NCRs to the endosymbionts. Our results suggest that S. meliloti relies on a suite of functions, including peptide transporters, the bacterial envelope structures, and stress response regulators, to resist the aggressive assault of NCR peptides in the nodule cells. IMPORTANCE The nitrogen-fixing symbiosis of legumes with rhizobium bacteria has a predominant ecological role in the nitrogen cycle and has the potential to provide the nitrogen required for plant growth in agriculture. The host plants allow the rhizobia to colonize specific symbiotic organs, the nodules, in large numbers in order to produce sufficient reduced nitrogen for the plants' needs. Some legumes, including Medicago spp., produce massively antimicrobial peptides to keep this large bacterial population in check. These peptides, known as NCRs, have the potential to kill the rhizobia, but in nodules, they rather inhibit the division of the bacteria, which maintain a high nitrogen-fixing activity. In this study, we show that the tempering of the antimicrobial activity of the NCR peptides in the Medicago symbiont Sinorhizobium meliloti is multifactorial and requires the YejABEF peptide transporter, the lipopolysaccharide outer membrane, and the stress response regulator RpoH1.
Collapse
|
25
|
Rapp J, Wagner B, Brilisauer K, Forchhammer K. In vivo Inhibition of the 3-Dehydroquinate Synthase by 7-Deoxysedoheptulose Depends on Promiscuous Uptake by Sugar Transporters in Cyanobacteria. Front Microbiol 2021; 12:692986. [PMID: 34248919 PMCID: PMC8261047 DOI: 10.3389/fmicb.2021.692986] [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: 04/09/2021] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
7-Deoxysedoheptulose (7dSh) is a bioactive deoxy-sugar actively excreted by the unicellular cyanobacterium Synechococcus elongatus PCC 7942 (S. elongatus) but also Streptomyces setonensis. In our previous publications we have shown that in S. elongatus, 7dSh is exclusively synthesized by promiscuous enzyme activity from an inhibitory by-product of radical SAM enzymes, without a specific gene cluster being involved. Additionally, we showed that 7dSh inhibits the growth of cyanobacteria, but also the growth of plants and fungi, presumably by inhibiting the 3-dehydroquinate synthase (DHQS), the second enzyme of the shikimate pathway, as the substrate of this enzyme strongly accumulates in cells treated with 7dSh. In this study, by using purified DHQS of Anabaena variabilis ATCC 29413 (A. variabilis) we biochemically confirmed that 7dSh is a competitive inhibitor of this enzyme. By analyzing the effect of 7dSh on a subset of cyanobacteria from all the five subsections, we identified different species whose growth was inhibited by 7dSh. We also found that in some of the susceptible cyanobacteria import of 7dSh is mediated by structurally different and promiscuous transporters: 7dSh can be taken up by the fructose ABC-transporter in A. variabilis and via the glucose permease in Synechocystis sp. PCC 6803 (Synechocystis sp.). In both cases, an effective uptake and thereby intracellular enrichment of 7dSh was essential for the inhibitory activity. Importantly, spontaneous mutations in the sugar transporters of A. variabilis and Synechocystis sp. not only disabled growth of the two strains on fructose and glucose, respectively, but also almost abolished their sensitivity to 7dSh. Although we have clearly shown in these examples that the effective uptake plays an essential role in the inhibitory effect of 7dSh, questions remain about how 7dSh resistance works in other (cyano)bacteria. Also, the involvement of a putative ribokinase in 7dSh resistance in the producer strain S. elongatus remained to be further investigated. Overall, these data establish 7dSh as the first allelochemical targeting the shikimate pathway in other cyanobacteria and plants and suggest a role of 7dSh in niche competition.
Collapse
Affiliation(s)
| | | | | | - Karl Forchhammer
- Interfaculty Institute of Microbiology and Infection Medicine, Organismic Interactions, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
26
|
Travin DY, Severinov K, Dubiley S. Natural Trojan horse inhibitors of aminoacyl-tRNA synthetases. RSC Chem Biol 2021; 2:468-485. [PMID: 34382000 PMCID: PMC8323819 DOI: 10.1039/d0cb00208a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
For most antimicrobial compounds with intracellular targets, getting inside the cell is the major obstacle limiting their activity. To pass this barrier some antibiotics mimic the compounds of specific interest for the microbe (siderophores, peptides, carbohydrates, etc.) and hijack the transport systems involved in their active uptake followed by the release of a toxic warhead inside the cell. In this review, we summarize the information about the structures, biosynthesis, and transport of natural inhibitors of aminoacyl-tRNA synthetases (albomycin, microcin C-related compounds, and agrocin 84) that rely on such "Trojan horse" strategy to enter the cell. In addition, we provide new data on the composition and distribution of biosynthetic gene clusters reminiscent of those coding for known Trojan horse aminoacyl-tRNA synthetases inhibitors. The products of these clusters are likely new antimicrobials that warrant further investigation.
Collapse
Affiliation(s)
- Dmitrii Y Travin
- Center of Life Sciences, Skolkovo Institute of Science and Technology Moscow Russia
- Institute of Gene Biology, Russian Academy of Sciences Moscow Russia
| | - Konstantin Severinov
- Center of Life Sciences, Skolkovo Institute of Science and Technology Moscow Russia
- Institute of Gene Biology, Russian Academy of Sciences Moscow Russia
- Waksman Institute for Microbiology, Rutgers, Piscataway New Jersey USA
| | - Svetlana Dubiley
- Center of Life Sciences, Skolkovo Institute of Science and Technology Moscow Russia
- Institute of Gene Biology, Russian Academy of Sciences Moscow Russia
| |
Collapse
|
27
|
Pang L, Weeks SD, Van Aerschot A. Aminoacyl-tRNA Synthetases as Valuable Targets for Antimicrobial Drug Discovery. Int J Mol Sci 2021; 22:1750. [PMID: 33578647 PMCID: PMC7916415 DOI: 10.3390/ijms22041750] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/20/2022] Open
Abstract
Aminoacyl-tRNA synthetases (aaRSs) catalyze the esterification of tRNA with a cognate amino acid and are essential enzymes in all three kingdoms of life. Due to their important role in the translation of the genetic code, aaRSs have been recognized as suitable targets for the development of small molecule anti-infectives. In this review, following a concise discussion of aaRS catalytic and proof-reading activities, the various inhibitory mechanisms of reported natural and synthetic aaRS inhibitors are discussed. Using the expanding repository of ligand-bound X-ray crystal structures, we classified these compounds based on their binding sites, focusing on their ability to compete with the association of one, or more of the canonical aaRS substrates. In parallel, we examined the determinants of species-selectivity and discuss potential resistance mechanisms of some of the inhibitor classes. Combined, this structural perspective highlights the opportunities for further exploration of the aaRS enzyme family as antimicrobial targets.
Collapse
Affiliation(s)
- Luping Pang
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49–box 1041, 3000 Leuven, Belgium;
- KU Leuven, Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, Herestraat 49–box 822, 3000 Leuven, Belgium
| | | | - Arthur Van Aerschot
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49–box 1041, 3000 Leuven, Belgium;
| |
Collapse
|
28
|
Telhig S, Ben Said L, Zirah S, Fliss I, Rebuffat S. Bacteriocins to Thwart Bacterial Resistance in Gram Negative Bacteria. Front Microbiol 2020; 11:586433. [PMID: 33240239 PMCID: PMC7680869 DOI: 10.3389/fmicb.2020.586433] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022] Open
Abstract
An overuse of antibiotics both in human and animal health and as growth promoters in farming practices has increased the prevalence of antibiotic resistance in bacteria. Antibiotic resistant and multi-resistant bacteria are now considered a major and increasing threat by national health agencies, making the need for novel strategies to fight bugs and super bugs a first priority. In particular, Gram-negative bacteria are responsible for a high proportion of nosocomial infections attributable for a large part to Enterobacteriaceae, such as pathogenic Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. To cope with their highly competitive environments, bacteria have evolved various adaptive strategies, among which the production of narrow spectrum antimicrobial peptides called bacteriocins and specifically microcins in Gram-negative bacteria. They are produced as precursor peptides that further undergo proteolytic cleavage and in many cases more or less complex posttranslational modifications, which contribute to improve their stability and efficiency. Many have a high stability in the gastrointestinal tract where they can target a single pathogen whilst only slightly perturbing the gut microbiota. Several microcins and antibiotics can bind to similar bacterial receptors and use similar pathways to cross the double-membrane of Gram-negative bacteria and reach their intracellular targets, which they also can share. Consequently, bacteria may use common mechanisms of resistance against microcins and antibiotics. This review describes both unmodified and modified microcins [lasso peptides, siderophore peptides, nucleotide peptides, linear azole(in)e-containing peptides], highlighting their potential as weapons to thwart bacterial resistance in Gram-negative pathogens and discusses the possibility of cross-resistance and co-resistance occurrence between antibiotics and microcins in Gram-negative bacteria.
Collapse
Affiliation(s)
- Soufiane Telhig
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
- Laboratory Molecules of Communication and Adaptation of Microorganisms, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Laila Ben Said
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Séverine Zirah
- Laboratory Molecules of Communication and Adaptation of Microorganisms, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Ismail Fliss
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Sylvie Rebuffat
- Laboratory Molecules of Communication and Adaptation of Microorganisms, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
29
|
Cui Y, Luo L, Wang X, Lu Y, Yi Y, Shan Y, Liu B, Zhou Y, Lü X. Mining, heterologous expression, purification, antibactericidal mechanism, and application of bacteriocins: A review. Compr Rev Food Sci Food Saf 2020; 20:863-899. [PMID: 33443793 DOI: 10.1111/1541-4337.12658] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/04/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Bacteriocins are generally considered as low-molecular-weight ribosomal peptides or proteins synthesized by G+ and G- bacteria that inhibit or kill other related or unrelated microorganisms. However, low yield is an important factor restricting the application of bacteriocins. This paper reviews mining methods, heterologous expression in different systems, the purification technologies applied to bacteriocins, and identification methods, as well as the antibacterial mechanism and applications in three different food systems. Bioinformatics improves the efficiency of bacteriocins mining. Bacteriocins can be heterologously expressed in different expression systems (e.g., Escherichia coli, Lactobacillus, and yeast). Ammonium sulfate precipitation, dialysis membrane, pH-mediated cell adsorption/desorption, solvent extraction, macroporous resin column, and chromatography are always used as purification methods for bacteriocins. The bacteriocins are identified through electrophoresis and mass spectrum. Cell envelope (e.g., cell permeabilization and pore formation) and inhibition of gene expression are common antibacterial mechanisms of bacteriocins. Bacteriocins can be added to protect meat products (e.g., beef and sausages), dairy products (e.g., cheese, milk, and yogurt), and vegetables and fruits (e.g., salad, apple juice, and soybean sprouts). The future research directions are also prospected.
Collapse
Affiliation(s)
- Yanlong Cui
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Lingli Luo
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xin Wang
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yingying Lu
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yanglei Yi
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yuanyuan Shan
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Bianfang Liu
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yuan Zhou
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xin Lü
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
30
|
Smits SHJ, Schmitt L, Beis K. Self-immunity to antibacterial peptides by ABC transporters. FEBS Lett 2020; 594:3920-3942. [PMID: 33040342 DOI: 10.1002/1873-3468.13953] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/22/2020] [Accepted: 10/05/2020] [Indexed: 01/17/2023]
Abstract
Bacteria produce under certain stress conditions bacteriocins and microcins that display antibacterial activity against closely related species for survival. Bacteriocins and microcins exert their antibacterial activity by either disrupting the membrane or inhibiting essential intracellular processes of the bacterial target. To this end, they can lyse bacterial membranes and cause subsequent loss of their integrity or nutrients, or hijack membrane receptors for internalisation. Both bacteriocins and microcins are ribosomally synthesised and several are posttranslationally modified, whereas others are not. Such peptides are also toxic to the producer bacteria, which utilise immunity proteins or/and dedicated ATP-binding cassette (ABC) transporters to achieve self-immunity and peptide export. In this review, we discuss the structure and mechanism of self-protection that is conferred by these ABC transporters.
Collapse
Affiliation(s)
- Sander H J Smits
- Institute of Biochemistry, Heinrich-Heine-University, Duesseldorf, Germany.,Center for Structural Studies, Heinrich-Heine-University, Duesseldorf, Germany
| | - Lutz Schmitt
- Institute of Biochemistry, Heinrich-Heine-University, Duesseldorf, Germany
| | - Konstantinos Beis
- Department of Life Sciences, Imperial College London, UK.,Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot, UK
| |
Collapse
|
31
|
Simons A, Alhanout K, Duval RE. Bacteriocins, Antimicrobial Peptides from Bacterial Origin: Overview of Their Biology and Their Impact against Multidrug-Resistant Bacteria. Microorganisms 2020; 8:E639. [PMID: 32349409 PMCID: PMC7285073 DOI: 10.3390/microorganisms8050639] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Currently, the emergence and ongoing dissemination of antimicrobial resistance among bacteria are critical health and economic issue, leading to increased rates of morbidity and mortality related to bacterial infections. Research and development for new antimicrobial agents is currently needed to overcome this problem. Among the different approaches studied, bacteriocins seem to be a promising possibility. These molecules are peptides naturally synthesized by ribosomes, produced by both Gram-positive bacteria (GPB) and Gram-negative bacteria (GNB), which will allow these bacteriocin producers to survive in highly competitive polymicrobial environment. Bacteriocins exhibit antimicrobial activity with variable spectrum depending on the peptide, which may target several bacteria. Already used in some areas such as agro-food, bacteriocins may be considered as interesting candidates for further development as antimicrobial agents used in health contexts, particularly considering the issue of antimicrobial resistance. The aim of this review is to present an updated global report on the biology of bacteriocins produced by GPB and GNB, as well as their antibacterial activity against relevant bacterial pathogens, and especially against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Alexis Simons
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France
- Institut Micalis, équipe Bactéries Pathogènes et Santé, Faculté de Pharmacie, Université Paris-Saclay—INRAE—AgroParisTech, 92296 Châtenay-Malabry, France
| | - Kamel Alhanout
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France
| | - Raphaël E. Duval
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France
- ABC Platform, Faculté de Pharmacie, F-54505 Vandœuvre-lès-Nancy, France
| |
Collapse
|
32
|
Travin DY, Bikmetov D, Severinov K. Translation-Targeting RiPPs and Where to Find Them. Front Genet 2020; 11:226. [PMID: 32296456 PMCID: PMC7136475 DOI: 10.3389/fgene.2020.00226] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/26/2020] [Indexed: 11/15/2022] Open
Abstract
Prokaryotic translation is among the major targets of diverse natural products with antibacterial activity including several classes of clinically relevant antibiotics. In this review, we summarize the information about the structure, biosynthesis, and modes of action of translation inhibiting ribosomally synthesized and post-translationally modified peptides (RiPPs). Azol(in)e-containing RiPPs are known to target translation, and several new compounds inhibiting the ribosome have been characterized recently. We performed a systematic search for biosynthetic gene clusters (BGCs) of azol(in)e-containing RiPPs. This search uncovered several groups of clusters that likely direct the synthesis of novel compounds, some of which may be targeting the ribosome.
Collapse
Affiliation(s)
- Dmitrii Y Travin
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry Bikmetov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Konstantin Severinov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Waksman Institute for Microbiology, Rutgers, Piscataway, NJ, United States
| |
Collapse
|
33
|
Soni DK, Dubey SK, Bhatnagar R. ATP-binding cassette (ABC) import systems of Mycobacterium tuberculosis: target for drug and vaccine development. Emerg Microbes Infect 2020; 9:207-220. [PMID: 31985348 PMCID: PMC7034087 DOI: 10.1080/22221751.2020.1714488] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nutrient procurement specifically from nutrient-limiting environment is essential for pathogenic bacteria to survive and/or persist within the host. Long-term survival or persistent infection is one of the main reasons for the overuse of antibiotics, and contributes to the development and spread of antibiotic resistance. Mycobacterium tuberculosis is known for long-term survival within the host, and develops multidrug resistance. Before and during infection, the pathogen encounters various harsh environmental conditions. To cope up with such nutrient-limiting conditions, it is crucial to uptake essential nutrients such as ions, sugars, amino acids, peptides, and metals, necessary for numerous vital biological activities. Among the various types of transporters, ATP-binding cassette (ABC) importers are essentially unique to bacteria, accessible as drug targets without penetrating the cytoplasmic membrane, and offer an ATP-dependent gateway into the cell by mimicking substrates of the importer and designing inhibitors against substrate-binding proteins, ABC importers endeavour for the development of successful drug candidates and antibiotics. Alternatively, the production of antibodies against substrate-binding proteins could lead to vaccine development. In this review, we will emphasize the role of M. tuberculosis ABC importers for survival and virulence within the host. Furthermore, we will elucidate their unique characteristics to discover emerging therapies to combat tuberculosis.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Suresh Kumar Dubey
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
34
|
Baquero F, Lanza VF, Baquero MR, Del Campo R, Bravo-Vázquez DA. Microcins in Enterobacteriaceae: Peptide Antimicrobials in the Eco-Active Intestinal Chemosphere. Front Microbiol 2019; 10:2261. [PMID: 31649628 PMCID: PMC6795089 DOI: 10.3389/fmicb.2019.02261] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022] Open
Abstract
Microcins are low-molecular-weight, ribosomally produced, highly stable, bacterial-inhibitory molecules involved in competitive, and amensalistic interactions between Enterobacteriaceae in the intestine. These interactions take place in a highly complex chemical landscape, the intestinal eco-active chemosphere, composed of chemical substances that positively or negatively influence bacterial growth, including those originated from nutrient uptake, and those produced by the action of the human or animal host and the intestinal microbiome. The contribution of bacteria results from their effect on the host generated molecules, on food and digested food, and organic substances from microbial origin, including from bacterial degradation. Here, we comprehensively review the main chemical substances present in the human intestinal chemosphere, particularly of those having inhibitory effects on microorganisms. With this background, and focusing on Enterobacteriaceae, the most relevant human pathogens from the intestinal microbiota, the microcin’s history and classification, mechanisms of action, and mechanisms involved in microcin’s immunity (in microcin producers) and resistance (non-producers) are reviewed. Products from the chemosphere likely modulate the ecological effects of microcin activity. Several cross-resistance mechanisms are shared by microcins, colicins, bacteriophages, and some conventional antibiotics, which are expected to produce cross-effects. Double-microcin-producing strains (such as microcins MccM and MccH47) have been successfully used for decades in the control of pathogenic gut organisms. Microcins are associated with successful gut colonization, facilitating translocation and invasion, leading to bacteremia, and urinary tract infections. In fact, Escherichia coli strains from the more invasive phylogroups (e.g., B2) are frequently microcinogenic. A publicly accessible APD3 database http://aps.unmc.edu/AP/ shows particular genes encoding microcins in 34.1% of E. coli strains (mostly MccV, MccM, MccH47, and MccI47), and much less in Shigella and Salmonella (<2%). Some 4.65% of Klebsiella pneumoniae are microcinogenic (mostly with MccE492), and even less in Enterobacter or Citrobacter (mostly MccS). The high frequency and variety of microcins in some Enterobacteriaceae indicate key ecological functions, a notion supported by their dominance in the intestinal microbiota of biosynthetic gene clusters involved in the synthesis of post-translationally modified peptide microcins.
Collapse
Affiliation(s)
- Fernando Baquero
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - Val F Lanza
- Bioinformatics Unit, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - Maria-Rosario Baquero
- Department of Microbiology, Alfonso X El Sabio University, Villanueva de la Cañada, Spain
| | - Rosa Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - Daniel A Bravo-Vázquez
- Department of Microbiology, Alfonso X El Sabio University, Villanueva de la Cañada, Spain
| |
Collapse
|
35
|
Niu G, Li Z, Huang P, Tan H. Engineering nucleoside antibiotics toward the development of novel antimicrobial agents. J Antibiot (Tokyo) 2019; 72:906-912. [DOI: 10.1038/s41429-019-0230-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/25/2019] [Accepted: 08/14/2019] [Indexed: 11/09/2022]
|
36
|
Jindal S, Yang L, Day PJ, Kell DB. Involvement of multiple influx and efflux transporters in the accumulation of cationic fluorescent dyes by Escherichia coli. BMC Microbiol 2019; 19:195. [PMID: 31438868 PMCID: PMC6704527 DOI: 10.1186/s12866-019-1561-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Background It is widely believed that most xenobiotics cross biomembranes by diffusing through the phospholipid bilayer, and that the use of protein transporters is an occasional adjunct. According to an alternative view, phospholipid bilayer transport is negligible, and several different transporters may be involved in the uptake of an individual molecular type. We recognise here that the availability of gene knockout collections allows one to assess the contributions of all potential transporters, and flow cytometry based on fluorescence provides a convenient high-throughput assay for xenobiotic uptake in individual cells. Results We used high-throughput flow cytometry to assess the ability of individual gene knockout strains of E coli to take up two membrane-permeable, cationic fluorescent dyes, namely the carbocyanine diS-C3(5) and the DNA dye SYBR Green. Individual strains showed a large range of distributions of uptake. The range of modal steady-state uptakes for the carbocyanine between the different strains was 36-fold. Knockouts of the ATP synthase α- and β-subunits greatly inhibited uptake, implying that most uptake was ATP-driven rather than being driven by a membrane potential. Dozens of transporters changed the steady-state uptake of the dye by more than 50% with respect to that of the wild type, in either direction (increased or decreased); knockouts of known influx and efflux transporters behaved as expected, giving credence to the general strategy. Many of the knockouts with the most reduced uptake were transporter genes of unknown function (‘y-genes’). Similarly, several overexpression variants in the ‘ASKA’ collection had the anticipated, opposite effects. Similar results were obtained with SYBR Green (the range being approximately 69-fold). Although it too contains a benzothiazole motif there was negligible correlation between its uptake and that of the carbocyanine when compared across the various strains (although the membrane potential is presumably the same in each case). Conclusions Overall, we conclude that the uptake of these dyes may be catalysed by a great many transporters of putatively broad and presently unknown specificity, and that the very large range between the ‘lowest’ and the ‘highest’ levels of uptake, even in knockouts of just single genes, implies strongly that phospholipid bilayer transport is indeed negligible. This work also casts serious doubt upon the use of such dyes as quantitative stains for representing either bioenergetic parameters or the amount of cellular DNA in unfixed cells (in vivo). By contrast, it opens up their potential use as transporter assay substrates in high-throughput screening. Electronic supplementary material The online version of this article (10.1186/s12866-019-1561-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Srijan Jindal
- Department of Chemistry, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK.,Manchester Institute of Biotechnology, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK.,Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Lei Yang
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Philip J Day
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK.,Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Douglas B Kell
- Department of Chemistry, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK. .,Manchester Institute of Biotechnology, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK. .,Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs, Lyngby, Denmark. .,Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| |
Collapse
|
37
|
Reiterative Synthesis by the Ribosome and Recognition of the N-Terminal Formyl Group by Biosynthetic Machinery Contribute to Evolutionary Conservation of the Length of Antibiotic Microcin C Peptide Precursor. mBio 2019; 10:mBio.00768-19. [PMID: 31040244 PMCID: PMC6495379 DOI: 10.1128/mbio.00768-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli microcin C (McC) is a representative member of peptide-nucleotide antibiotics produced by diverse microorganisms. The vast majority of biosynthetic gene clusters responsible for McC-like compound production encode 7-amino-acid-long precursor peptides, which are C-terminally modified by dedicated biosynthetic enzymes with a nucleotide moiety to produce a bioactive compound. In contrast, the sequences of McC-like compound precursor peptides are not conserved. Here, we studied the consequences of E. coli McC precursor peptide length increase on antibiotic production and activity. We show that increasing the precursor peptide length strongly decreases McC production by affecting multiple biosynthetic steps, suggesting that the McC biosynthesis system has evolved under significant functional constraints to maintain the precursor peptide length. Microcin C (McC) is a peptide adenylate antibiotic produced by Escherichiacoli cells bearing a plasmid-borne mcc gene cluster. Most MccA precursors, encoded by validated mcc operons from diverse bacteria, are 7 amino acids long, but the significance of this precursor length conservation has remained unclear. Here, we created derivatives of E. colimcc operons encoding longer precursors and studied their synthesis and bioactivities. We found that increasing the precursor length to 11 amino acids and beyond strongly decreased antibiotic production. We found this decrease to depend on several parameters. First, reiterative synthesis of the MccA peptide by the ribosome was decreased at longer mccA open reading frames, leading to less efficient competition with other messenger RNAs. Second, the presence of a formyl group at the N-terminal methionine of the heptameric peptide had a strong stimulatory effect on adenylation by the MccB enzyme. No such formyl group stimulation was observed for longer peptides. Finally, the presence of the N-terminal formyl on the heptapeptide adenylate stimulated bioactivity, most likely at the uptake stage. Together, these factors should contribute to optimal activity of McC-like compounds as 7-amino-acid peptide moieties and suggest convergent evolution of several steps of the antibiotic biosynthesis pathway and their adjustment to sensitive cell uptake machinery to create a potent drug.
Collapse
|
38
|
Mohanty B, Hanson-Manful P, Finn TJ, Chambers CR, McKellar JLO, Macindoe I, Helder S, Setiyaputra S, Zhong Y, Mackay JP, Patrick WM. The uncharacterized bacterial protein YejG has the same architecture as domain III of elongation factor G. Proteins 2019; 87:699-705. [PMID: 30958578 DOI: 10.1002/prot.25687] [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: 11/13/2018] [Revised: 02/23/2019] [Accepted: 04/04/2019] [Indexed: 11/08/2022]
Abstract
InterPro family IPR020489 comprises ~1000 uncharacterized bacterial proteins. Previously we showed that overexpressing the Escherichia coli representative of this family, EcYejG, conferred low-level resistance to aminoglycoside antibiotics. In an attempt to shed light on the biochemical function of EcYejG, we have solved its structure using multinuclear solution NMR spectroscopy. The structure most closely resembles that of domain III from elongation factor G (EF-G). EF-G catalyzes ribosomal translocation and mutations in EF-G have also been associated with aminoglycoside resistance. While we were unable to demonstrate a direct interaction between EcYejG and the ribosome, the protein might play a role in translation.
Collapse
Affiliation(s)
- Biswaranjan Mohanty
- Faculty of Pharmacy and Pharmaceutical Sciences, Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Paulina Hanson-Manful
- Institute of Natural and Mathematical Sciences, Massey University, Auckland, New Zealand
| | - Thomas J Finn
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | | | - Ingrid Macindoe
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephanie Helder
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Surya Setiyaputra
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Yichen Zhong
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Wayne M Patrick
- School of Biological Sciences, Victoria University, Wellington, New Zealand
| |
Collapse
|
39
|
Ghosh C, Sarkar P, Issa R, Haldar J. Alternatives to Conventional Antibiotics in the Era of Antimicrobial Resistance. Trends Microbiol 2019; 27:323-338. [PMID: 30683453 DOI: 10.1016/j.tim.2018.12.010] [Citation(s) in RCA: 413] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 11/30/2018] [Accepted: 12/19/2018] [Indexed: 02/07/2023]
Abstract
As more antibiotics are rendered ineffective by drug-resistant bacteria, focus must be shifted towards alternative therapies for treating infections. Although several alternatives already exist in nature, the challenge is to implement them in clinical use. Advancements within biotechnology, genetic engineering, and synthetic chemistry have opened up new avenues towards the search for therapies that can substitute for antibiotics. This review provides an introduction to the various promising approaches that have been adopted in this regard. Whilst the use of bacteriophages and antibodies has been partly implemented, other promising strategies, such as probiotics, lysins, and antimicrobial peptides, are in various stages of development. Propitious concepts such as genetically modified phages, antibacterial oligonucleotides, and CRISPR-Cas9 are also discussed.
Collapse
Affiliation(s)
- Chandradhish Ghosh
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, India
| | - Paramita Sarkar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, India
| | - Rahaf Issa
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, India.
| |
Collapse
|
40
|
Dong SH, Kulikovsky A, Zukher I, Estrada P, Dubiley S, Severinov K, Nair SK. Biosynthesis of the RiPP trojan horse nucleotide antibiotic microcin C is directed by the N-formyl of the peptide precursor. Chem Sci 2018; 10:2391-2395. [PMID: 30881667 PMCID: PMC6385645 DOI: 10.1039/c8sc03173h] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023] Open
Abstract
The N-formyl moiety of the peptide precursor directs the biosynthesis of the RiPP trojan horse nucleotide antibiotic McC.
Microcin C7 (McC) is a peptide antibiotic modified by a linkage of the terminal isoAsn amide to AMP via a phosphoramidate bond. Post-translational modification on this ribosomally produced heptapeptide precursor is carried out by MccB, which consumes two equivalents of ATP to generate the N–P linkage. We demonstrate that MccB only efficiently processes the precursor heptapeptide that retains the N-formylated initiator Met (fMet). Binding studies and kinetic measurements evidence the role of the N-formyl moiety. Structural data show that the N-formyl peptide binding results in an ordering of residues in the MccB “crossover loop”, which dictates specificity in homologous ubiquitin activating enzymes. The N-formyl peptide exhibits substrate inhibition, and cannot be displaced from MccB by the desformyl counterpart. Such substrate inhibition may be a strategy to avert unwanted McC buildup and avert toxicity in the cytoplasm of producing organisms.
Collapse
Affiliation(s)
- Shi-Hui Dong
- Department of Biochemistry , University of Illinois at Urbana-Champaign , Illinois , USA . .,Carl Woese Institute for Genomic Biology , University of Illinois at Urbana-Champaign , Illinois , USA
| | - Alexey Kulikovsky
- Department of Biochemistry , University of Illinois at Urbana-Champaign , Illinois , USA . .,Institute of Gene Biology , Russian Academy of Science , 34/5 Vavilo str. , 11934 Moscow , Russia.,Center for Life Sciences , Skolkov Institute of Science and Technology , 3 Nobel str. , 143026 Moscow , Russia
| | - Inna Zukher
- Institute of Gene Biology , Russian Academy of Science , 34/5 Vavilo str. , 11934 Moscow , Russia
| | - Paola Estrada
- Department of Biochemistry , University of Illinois at Urbana-Champaign , Illinois , USA .
| | - Svetlana Dubiley
- Institute of Gene Biology , Russian Academy of Science , 34/5 Vavilo str. , 11934 Moscow , Russia.,Center for Life Sciences , Skolkov Institute of Science and Technology , 3 Nobel str. , 143026 Moscow , Russia
| | - Konstantin Severinov
- Institute of Gene Biology , Russian Academy of Science , 34/5 Vavilo str. , 11934 Moscow , Russia.,Center for Life Sciences , Skolkov Institute of Science and Technology , 3 Nobel str. , 143026 Moscow , Russia.,Waksman Institute for Microbiology , 190 Frelinghuysen Road , Piscataway , New Jersey , USA .
| | - Satish K Nair
- Department of Biochemistry , University of Illinois at Urbana-Champaign , Illinois , USA . .,Carl Woese Institute for Genomic Biology , University of Illinois at Urbana-Champaign , Illinois , USA.,Center for Biophysics and Quantitative Biology , University of Illinois at Urbana-Champaign , Illinois , USA
| |
Collapse
|
41
|
Abstract
Copper-binding metallophores, or chalkophores, play a role in microbial copper homeostasis that is analogous to that of siderophores in iron homeostasis. The best-studied chalkophores are members of the methanobactin (Mbn) family-ribosomally produced, posttranslationally modified natural products first identified as copper chelators responsible for copper uptake in methane-oxidizing bacteria. To date, Mbns have been characterized exclusively in those species, but there is genomic evidence for their production in a much wider range of bacteria. This review addresses the current state of knowledge regarding the function, biosynthesis, transport, and regulation of Mbns. While the roles of several proteins in these processes are supported by substantial genetic and biochemical evidence, key aspects of Mbn manufacture, handling, and regulation remain unclear. In addition, other natural products that have been proposed to mediate copper uptake as well as metallophores that have biologically relevant roles involving copper binding, but not copper uptake, are discussed.
Collapse
Affiliation(s)
- Grace E Kenney
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA; ,
| | - Amy C Rosenzweig
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA; ,
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, USA
| |
Collapse
|
42
|
Tanaka KJ, Song S, Mason K, Pinkett HW. Selective substrate uptake: The role of ATP-binding cassette (ABC) importers in pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:868-877. [PMID: 28847505 PMCID: PMC5807212 DOI: 10.1016/j.bbamem.2017.08.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023]
Abstract
The uptake of nutrients, including metals, amino acids and peptides are required for many biological processes. Pathogenic bacteria scavenge these essential nutrients from microenvironments to survive within the host. Pathogens must utilize a myriad of mechanisms to acquire these essential nutrients from the host while mediating the effects of toxicity. Bacteria utilize several transport proteins, including ATP-binding cassette (ABC) transporters to import and expel substrates. ABC transporters, conserved across all organisms, are powered by the energy from ATP to move substrates across cellular membranes. In this review, we will focus on nutrient uptake, the role of ABC importers at the host-pathogen interface, and explore emerging therapies to combat pathogenesis. This article is part of a Special Issue entitled: Beyond the Structure-Function Horizon of Membrane Proteins edited by Ute Hellmich, Rupak Doshi and Benjamin McIlwain.
Collapse
Affiliation(s)
- Kari J Tanaka
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Saemee Song
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Kevin Mason
- The Research Institute at Nationwide Children's Hospital and The Ohio State University, College of Medicine, Department of Pediatrics, Center for Microbial Pathogenesis, Columbus, OH, USA
| | - Heather W Pinkett
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
43
|
Kenney GE, Rosenzweig AC. Methanobactins: Maintaining copper homeostasis in methanotrophs and beyond. J Biol Chem 2018; 293:4606-4615. [PMID: 29348173 PMCID: PMC5880147 DOI: 10.1074/jbc.tm117.000185] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Methanobactins (Mbns) are ribosomally produced, post-translationally modified natural products that bind copper with high affinity and specificity. Originally identified in methanotrophic bacteria, which have a high need for copper, operons encoding these compounds have also been found in many non-methanotrophic bacteria. The proteins responsible for Mbn biosynthesis include several novel enzymes. Mbn transport involves export through a multidrug efflux pump and re-internalization via a TonB-dependent transporter. Release of copper from Mbn and the molecular basis for copper regulation of Mbn production remain to be elucidated. Future work is likely to result in the identification of new enzymatic chemistry, opportunities for bioengineering and drug targeting of copper metabolism, and an expanded understanding of microbial metal homeostasis.
Collapse
Affiliation(s)
- Grace E Kenney
- Departments of Molecular Biosciences, Evanston, Illinois 60208
| | - Amy C Rosenzweig
- Departments of Molecular Biosciences, Evanston, Illinois 60208; Chemistry, Northwestern University, Evanston, Illinois 60208.
| |
Collapse
|
44
|
Palmer M, Steenkamp ET, Coetzee MPA, Blom J, Venter SN. Genome-Based Characterization of Biological Processes That Differentiate Closely Related Bacteria. Front Microbiol 2018; 9:113. [PMID: 29467735 PMCID: PMC5808187 DOI: 10.3389/fmicb.2018.00113] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/17/2018] [Indexed: 12/21/2022] Open
Abstract
Bacteriologists have strived toward attaining a natural classification system based on evolutionary relationships for nearly 100 years. In the early twentieth century it was accepted that a phylogeny-based system would be the most appropriate, but in the absence of molecular data, this approach proved exceedingly difficult. Subsequent technical advances and the increasing availability of genome sequencing have allowed for the generation of robust phylogenies at all taxonomic levels. In this study, we explored the possibility of linking biological characters to higher-level taxonomic groups in bacteria by making use of whole genome sequence information. For this purpose, we specifically targeted the genus Pantoea and its four main lineages. The shared gene sets were determined for Pantoea, the four lineages within the genus, as well as its sister-genus Tatumella. This was followed by functional characterization of the gene sets using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. In comparison to Tatumella, various traits involved in nutrient cycling were identified within Pantoea, providing evidence for increased efficacy in recycling of metabolites within the genus. Additionally, a number of traits associated with pathogenicity were identified within species often associated with opportunistic infections, with some support for adaptation toward overcoming host defenses. Some traits were also only conserved within specific lineages, potentially acquired in an ancestor to the lineage and subsequently maintained. It was also observed that the species isolated from the most diverse sources were generally the most versatile in their carbon metabolism. By investigating evolution, based on the more variable genomic regions, it may be possible to detect biologically relevant differences associated with the course of evolution and speciation.
Collapse
Affiliation(s)
- Marike Palmer
- Department of Microbiology and Plant Pathology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Emma T Steenkamp
- Department of Microbiology and Plant Pathology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Martin P A Coetzee
- Department of Genetic, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Jochen Blom
- Bioinformatics and Systems Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Stephanus N Venter
- Department of Microbiology and Plant Pathology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
45
|
Tsibulskaya D, Mokina O, Kulikovsky A, Piskunova J, Severinov K, Serebryakova M, Dubiley S. The Product of Yersinia pseudotuberculosis mcc Operon Is a Peptide-Cytidine Antibiotic Activated Inside Producing Cells by the TldD/E Protease. J Am Chem Soc 2017; 139:16178-16187. [PMID: 29045133 DOI: 10.1021/jacs.7b07118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microcin C is a heptapeptide-adenylate antibiotic produced by some strains of Escherichia coli. Its peptide part is responsible for facilitated transport inside sensitive cells where it is proteolyzed with release of a toxic warhead-a nonhydrolyzable aspartamidyl-adenylate, which inhibits aspartyl-tRNA synthetase. Recently, a microcin C homologue from Bacillus amyloliquefaciens containing a longer peptide part modified with carboxymethyl-cytosine instead of adenosine was described, but no biological activity of this compound was revealed. Here, we characterize modified peptide-cytidylate from Yersinia pseudotuberculosis. As reported for B. amyloliquefaciens homologue, the initially synthesized compound contains a long peptide that is biologically inactive. This compound is subjected to endoproteolytic processing inside producing cells by the evolutionary conserved TldD/E protease. As a result, an 11-amino acid long peptide with C-terminal modified cytosine residue is produced. This compound is exported outside the producing cell and is bioactive, inhibiting sensitive cells in the same way as E. coli microcin C. Proteolytic processing inside producing cells is a novel strategy of peptide-nucleotide antibiotics biosynthesis that may help control production levels and avoid toxicity to the producer.
Collapse
Affiliation(s)
- Darya Tsibulskaya
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology , 3 Nobel str., 143026 Moscow, Russia
| | - Olga Mokina
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology , 3 Nobel str., 143026 Moscow, Russia
| | - Alexey Kulikovsky
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology , 3 Nobel str., 143026 Moscow, Russia.,Department of Biochemistry, University of Illinois at Urbana-Champaign , 600 S. Mathews Ave., Urbana, Illinois 61801, United States
| | - Julia Piskunova
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology , 3 Nobel str., 143026 Moscow, Russia
| | - Konstantin Severinov
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology , 3 Nobel str., 143026 Moscow, Russia.,Waksman Institute for Microbiology , 190 Frelinghuysen Road, Piscataway, New Jersey 08854-8020, United States
| | - Marina Serebryakova
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University , Leninskie Gory 1, Bldg. 40, Moscow 119991, Russia
| | - Svetlana Dubiley
- Institute of Gene Biology , Russian Academy of Science, 34/5 Vavilov str., 119334 Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology , 3 Nobel str., 143026 Moscow, Russia
| |
Collapse
|
46
|
Purification and primary characterization of a novel bacteriocin, LiN333, from Lactobacillus casei, an isolate from a Chinese fermented food. Lebensm Wiss Technol 2017. [DOI: 10.1016/j.lwt.2017.04.056] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Metelev M, Osterman IA, Ghilarov D, Khabibullina NF, Yakimov A, Shabalin K, Utkina I, Travin DY, Komarova ES, Serebryakova M, Artamonova T, Khodorkovskii M, Konevega AL, Sergiev PV, Severinov K, Polikanov YS. Klebsazolicin inhibits 70S ribosome by obstructing the peptide exit tunnel. Nat Chem Biol 2017; 13:1129-1136. [PMID: 28846667 PMCID: PMC5701663 DOI: 10.1038/nchembio.2462] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/19/2017] [Indexed: 12/19/2022]
Abstract
While screening of small-molecular metabolites produced by most cultivatable microorganisms often results in rediscovery of known compounds, genome-mining programs allow to harness much greater chemical diversity and result in discovery of new molecular scaffolds. Here we report genome-guided identification of a new antibiotic klebsazolicin (KLB) from Klebsiella pneumoniae that inhibits growth of sensitive cells by targeting ribosome. A member of ribosomally-synthesized post-translationally modified peptides (RiPPs), KLB is characterized by the presence of unique N-terminal amidine ring essential for its activity. Biochemical in vitro studies indicate that KLB inhibits ribosome by interfering with translation elongation. Structural analysis of the ribosome-KLB complex reveals the compound bound in the peptide exit tunnel overlapping with the binding sites of macrolides or streptogramins-B. KLB adopts compact conformation and largely obstructs the tunnel. Engineered KLB fragments retain in vitro activity and can serve as a starting point for the development of new bioactive compounds.
Collapse
Affiliation(s)
- Mikhail Metelev
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia.,Institute of Antimicrobial Chemotherapy, Smolensk State Medical Academy, Smolensk, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Institute of Gene Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Ilya A Osterman
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry Ghilarov
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Institute of Gene Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Nelli F Khabibullina
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Alexander Yakimov
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia.,Petersburg Nuclear Physics Institute, NRC Kurchatov Institute, Gatchina, Russia
| | - Konstantin Shabalin
- Petersburg Nuclear Physics Institute, NRC Kurchatov Institute, Gatchina, Russia
| | - Irina Utkina
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Dmitry Y Travin
- Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina S Komarova
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Marina Serebryakova
- Institute of Gene Biology of the Russian Academy of Sciences, Moscow, Russia.,Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatyana Artamonova
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia
| | - Mikhail Khodorkovskii
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia
| | - Andrey L Konevega
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia.,Petersburg Nuclear Physics Institute, NRC Kurchatov Institute, Gatchina, Russia
| | - Petr V Sergiev
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin Severinov
- Research Center of Nanobiotechnologies, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia.,Institute of Gene Biology of the Russian Academy of Sciences, Moscow, Russia.,Waksman Institute for Microbiology, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
48
|
Ageitos J, Sánchez-Pérez A, Calo-Mata P, Villa T. Antimicrobial peptides (AMPs): Ancient compounds that represent novel weapons in the fight against bacteria. Biochem Pharmacol 2017; 133:117-138. [DOI: 10.1016/j.bcp.2016.09.018] [Citation(s) in RCA: 402] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/19/2016] [Indexed: 01/01/2023]
|
49
|
Waldman AJ, Ng TL, Wang P, Balskus EP. Heteroatom-Heteroatom Bond Formation in Natural Product Biosynthesis. Chem Rev 2017; 117:5784-5863. [PMID: 28375000 PMCID: PMC5534343 DOI: 10.1021/acs.chemrev.6b00621] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Natural products that contain functional groups with heteroatom-heteroatom linkages (X-X, where X = N, O, S, and P) are a small yet intriguing group of metabolites. The reactivity and diversity of these structural motifs has captured the interest of synthetic and biological chemists alike. Functional groups containing X-X bonds are found in all major classes of natural products and often impart significant biological activity. This review presents our current understanding of the biosynthetic logic and enzymatic chemistry involved in the construction of X-X bond containing functional groups within natural products. Elucidating and characterizing biosynthetic pathways that generate X-X bonds could both provide tools for biocatalysis and synthetic biology, as well as guide efforts to uncover new natural products containing these structural features.
Collapse
Affiliation(s)
- Abraham J. Waldman
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Tai L. Ng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Peng Wang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Emily P. Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| |
Collapse
|
50
|
Ran R, Zeng H, Zhao D, Liu R, Xu X. The Novel Property of Heptapeptide of Microcin C7 in Affecting the Cell Growth of Escherichia coli. Molecules 2017; 22:E432. [PMID: 28282893 PMCID: PMC6155343 DOI: 10.3390/molecules22030432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/06/2017] [Indexed: 12/01/2022] Open
Abstract
Microcin C7 (McC), widely distributed in enterobacteria, is a promising antibiotic against antibiotic resistance [...].
Collapse
Affiliation(s)
- Rensen Ran
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Huan Zeng
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Dong Zhao
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ruiyuan Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xia Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|