1
|
Rice AJ, Sword TT, Chengan K, Mitchell DA, Mouncey NJ, Moore SJ, Bailey CB. Cell-free synthetic biology for natural product biosynthesis and discovery. Chem Soc Rev 2025; 54:4314-4352. [PMID: 40104998 PMCID: PMC11920963 DOI: 10.1039/d4cs01198h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Indexed: 03/20/2025]
Abstract
Natural products have applications as biopharmaceuticals, agrochemicals, and other high-value chemicals. However, there are challenges in isolating natural products from their native producers (e.g. bacteria, fungi, plants). In many cases, synthetic chemistry or heterologous expression must be used to access these important molecules. The biosynthetic machinery to generate these compounds is found within biosynthetic gene clusters, primarily consisting of the enzymes that biosynthesise a range of natural product classes (including, but not limited to ribosomal and nonribosomal peptides, polyketides, and terpenoids). Cell-free synthetic biology has emerged in recent years as a bottom-up technology applied towards both prototyping pathways and producing molecules. Recently, it has been applied to natural products, both to characterise biosynthetic pathways and produce new metabolites. This review discusses the core biochemistry of cell-free synthetic biology applied to metabolite production and critiques its advantages and disadvantages compared to whole cell and/or chemical production routes. Specifically, we review the advances in cell-free biosynthesis of ribosomal peptides, analyse the rapid prototyping of natural product biosynthetic enzymes and pathways, highlight advances in novel antimicrobial discovery, and discuss the rising use of cell-free technologies in industrial biotechnology and synthetic biology.
Collapse
Affiliation(s)
- Andrew J Rice
- Department of Biochemistry, School of Medicine - Basic Sciences, Vanderbilt University Medical Research Building-IV, Nashville, Tennessee, 37232, USA
| | - Tien T Sword
- Department of Chemistry, University of Tennessee-Knoxville, Knoxville, TN, USA
| | | | - Douglas A Mitchell
- Department of Biochemistry, School of Medicine - Basic Sciences, Vanderbilt University Medical Research Building-IV, Nashville, Tennessee, 37232, USA
- Department of Chemistry, Vanderbilt University, Medical Research Building-IV, Nashville, Tennessee, 37232, USA
| | - Nigel J Mouncey
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, 94720, USA
| | - Simon J Moore
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - Constance B Bailey
- School of Chemistry, University of Sydney, Camperdown, NSW, 2001, Australia.
| |
Collapse
|
2
|
Owens SL, Ahmed SR, Lang RM, Stewart LE, Mori S. Natural Products That Contain Higher Homologated Amino Acids. Chembiochem 2024; 25:e202300822. [PMID: 38487927 PMCID: PMC11386549 DOI: 10.1002/cbic.202300822] [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: 12/04/2023] [Revised: 03/13/2024] [Indexed: 04/11/2024]
Abstract
This review focuses on discussing natural products (NPs) that contain higher homologated amino acids (homoAAs) in the structure as well as the proposed and characterized biosynthesis of these non-proteinogenic amino acids. Homologation of amino acids includes the insertion of a methylene group into its side chain. It is not a very common modification found in NP biosynthesis as approximately 450 homoAA-containing NPs have been isolated from four bacterial phyla (Cyanobacteria, Actinomycetota, Myxococcota, and Pseudomonadota), two fungal phyla (Ascomycota and Basidiomycota), and one animal phylum (Porifera), except for a few examples. Amino acids that are found to be homologated and incorporated in the NP structures include the following ten amino acids: alanine, arginine, cysteine, isoleucine, glutamic acid, leucine, phenylalanine, proline, serine, and tyrosine, where isoleucine, leucine, phenylalanine, and tyrosine share the comparable enzymatic pathway. Other amino acids have their individual homologation pathway (arginine, proline, and glutamic acid for bacteria), likely utilize the primary metabolic pathway (alanine and glutamic acid for fungi), or have not been reported (cysteine and serine). Despite its possible high potential in the drug discovery field, the biosynthesis of homologated amino acids has a large room to explore for future combinatorial biosynthesis and metabolic engineering purpose.
Collapse
Affiliation(s)
- Skyler L. Owens
- Department of Chemistry and Biochemistry, Augusta University, 1120 15th Street, Augusta, GA 30912
| | - Shopno R. Ahmed
- Department of Chemistry and Biochemistry, Augusta University, 1120 15th Street, Augusta, GA 30912
| | - Rebecca M. Lang
- Department of Chemistry and Biochemistry, Augusta University, 1120 15th Street, Augusta, GA 30912
| | - Laura E. Stewart
- Department of Chemistry and Biochemistry, Augusta University, 1120 15th Street, Augusta, GA 30912
| | - Shogo Mori
- Department of Chemistry and Biochemistry, Augusta University, 1120 15th Street, Augusta, GA 30912
| |
Collapse
|
3
|
Abstract
The ability to site-selectively modify equivalent functional groups in a molecule has the potential to streamline syntheses and increase product yields by lowering step counts. Enzymes catalyze site-selective transformations throughout primary and secondary metabolism, but leveraging this capability for non-native substrates and reactions requires a detailed understanding of the potential and limitations of enzyme catalysis and how these bounds can be extended by protein engineering. In this review, we discuss representative examples of site-selective enzyme catalysis involving functional group manipulation and C-H bond functionalization. We include illustrative examples of native catalysis, but our focus is on cases involving non-native substrates and reactions often using engineered enzymes. We then discuss the use of these enzymes for chemoenzymatic transformations and target-oriented synthesis and conclude with a survey of tools and techniques that could expand the scope of non-native site-selective enzyme catalysis.
Collapse
Affiliation(s)
- Dibyendu Mondal
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Harrison M Snodgrass
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Christian A Gomez
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jared C Lewis
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
4
|
Schuler SMM, Jürjens G, Marker A, Hemmann U, Rey A, Yvon S, Lagrevol M, Hamiti M, Nguyen F, Hirsch R, Pöverlein C, Vilcinskas A, Hammann P, Wilson DN, Mourez M, Coyne S, Bauer A. Full Profiling of GE81112A, an Underexplored Tetrapeptide Antibiotic with Activity against Gram-Negative Pathogens. Microbiol Spectr 2023; 11:e0224722. [PMID: 37140391 PMCID: PMC10269895 DOI: 10.1128/spectrum.02247-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 05/05/2023] Open
Abstract
After the first total synthesis combined with structure revision, we performed thorough in vitro and in vivo profiling of the underexplored tetrapeptide GE81112A. From the determination of the biological activity spectrum and physicochemical and early absorption-distribution-metabolism-excretion-toxicity (eADMET) properties, as well as in vivo data regarding tolerability and pharmacokinetics (PK) in mice and efficacy in an Escherichia coli-induced septicemia model, we were able to identify the critical and limiting parameters of the original hit compound. Thus, the generated data will serve as the basis for further compound optimization programs and developability assessments to identify candidates for preclinical/clinical development derived from GE81112A as the lead structure. IMPORTANCE The spread of antimicrobial resistance (AMR) is becoming a more and more important global threat to human health. With regard to current medical needs, penetration into the site of infection represents the major challenge in the treatment of infections caused by Gram-positive bacteria. Considering infections associated with Gram-negative bacteria, resistance is a major issue. Obviously, novel scaffolds for the design of new antibacterials in this arena are urgently needed to overcome this crisis. Such a novel potential lead structure is represented by the GE81112 compounds, which inhibit protein synthesis by interacting with the small 30S ribosomal subunit using a binding site distinct from that of other known ribosome-targeting antibiotics. Therefore, the tetrapeptide antibiotic GE81112A was chosen for further exploration as a potential lead for the development of antibiotics with a new mode of action against Gram-negative bacteria.
Collapse
Affiliation(s)
- Sören M. M. Schuler
- Branch Bioresources of the Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - Gerrit Jürjens
- Branch Bioresources of the Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | | | | | - Astrid Rey
- Sanofi R&D, Therapeutic Area Infectious Diseases, Marcy L’Etoile, France
| | - Stéphane Yvon
- Sanofi R&D, Therapeutic Area Infectious Diseases, Marcy L’Etoile, France
| | - Marjorie Lagrevol
- Sanofi R&D, Therapeutic Area Infectious Diseases, Marcy L’Etoile, France
| | - Mohamed Hamiti
- Sanofi R&D, Therapeutic Area Infectious Diseases, Marcy L’Etoile, France
| | - Fabian Nguyen
- Gene Center, Department for Biochemistry and Center for Protein Science Munich, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rolf Hirsch
- Branch Bioresources of the Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | | | - Andreas Vilcinskas
- Branch Bioresources of the Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
- Institute for Insect Biotechnology, Justus-Liebig University of Giessen, Giessen, Germany
| | | | - Daniel N. Wilson
- Gene Center, Department for Biochemistry and Center for Protein Science Munich, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Mourez
- Sanofi R&D, Therapeutic Area Infectious Diseases, Marcy L’Etoile, France
| | - Sebastien Coyne
- Sanofi R&D, Therapeutic Area Infectious Diseases, Marcy L’Etoile, France
| | - Armin Bauer
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| |
Collapse
|
5
|
Wang J, Wang Y, Wu Q, Zhang Y. Multidimensional engineering of Escherichia coli for efficient biosynthesis of cis-3-hydroxypipecolic acid. BIORESOURCE TECHNOLOGY 2023; 382:129173. [PMID: 37187331 DOI: 10.1016/j.biortech.2023.129173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
Cis-3-hydroxypipecolic acid (cis-3-HyPip) is the crucial part of many alkaloids and drugs. However, its bio-based industrial production remains challenging. Here, lysine cyclodeaminase from Streptomyces malaysiensis (SmLCD) and pipecolic acid hydroxylase from Streptomyces sp. L-49973 (StGetF) were screened to achieve the conversion of L-lysine to cis-3-HyPip. Considering the high-cost of cofactors, NAD(P)H oxidase from Lactobacillus sanfranciscensis (LsNox) was further overexpressed in chassis strain Escherichia coli W3110 ΔsucCD (α-ketoglutarate-producing strain) to construct the NAD+ regeneration system, thus realizing the bioconversion of cis-3-HyPip from low-cost substrate L-lysine without NAD+ and α-ketoglutarate addition. To further accelerate the transmission efficiency of cis-3-HyPip biosynthetic pathway, multiple-enzyme expression optimization and transporter dynamic regulation via promoter engineering were conducted. Through fermentation optimization, the final engineered strain HP-13 generated 78.4 g/L cis-3-HyPip with 78.9% conversion in a 5-L fermenter, representing the highest production level achieved so far. These strategies described herein show promising potentials for large-scale production of cis-3-HyPip.
Collapse
Affiliation(s)
- Jiaping Wang
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China
| | - Yaqiong Wang
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China
| | - Qin Wu
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China
| | - Yimin Zhang
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China.
| |
Collapse
|
6
|
Fayad S, Jafari A, Schuler SMM, Kurz M, Plettenburg O, Hammann PE, Bauer A, Jürjens G, Pöverlein C. Total Synthesis of GE81112A: An Orthoester-Based Approach. J Org Chem 2023; 88:5597-5608. [PMID: 37023463 PMCID: PMC10167690 DOI: 10.1021/acs.joc.3c00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
The GE81112 series, consisting of three naturally occurring tetrapeptides and synthetic derivatives, is evaluated as a potential lead structure for the development of a new antibacterial drug. Although the first total synthesis of GE81112A reported by our group provided sufficient amounts of material for an initial in depth biological profiling of the compound, improvements of the routes toward the key building blocks were needed for further upscaling and structure-activity relationship studies. The major challenges identified were poor stereoselectivity in the synthesis of the C-terminal β-hydroxy histidine intermediate and a concise access to all four isomers of the 3-hydroxy pipecolic acid. Herein, we report a second-generation synthesis of GE81112A, which is also applicable to access further representatives of this series. Based on Lajoie's ortho-ester-protected serine aldehydes as key building blocks, the described route provides both a satisfactory improvement in stereoselectivity of the β-hydroxy histidine intermediate synthesis and a stereoselective approach toward both orthogonally protected cis and trans-3-hydroxy pipecolic acid.
Collapse
Affiliation(s)
- Scherin Fayad
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
| | - Ardalan Jafari
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
| | - Sören M M Schuler
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Bioresources, 35392 Giessen, Germany
- Evotec International GmbH, 37079 Göttingen, Germany
| | - Michael Kurz
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Oliver Plettenburg
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
| | - Peter E Hammann
- Evotec International GmbH, 37079 Göttingen, Germany
- Sanofi-Aventis Deutschland GmbH, R&D, Infectious Diseases, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Armin Bauer
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Gerrit Jürjens
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Bioresources, 35392 Giessen, Germany
| | - Christoph Pöverlein
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
7
|
Tao H, Abe I. Oxidative modification of free-standing amino acids by Fe(II)/αKG-dependent oxygenases. ENGINEERING MICROBIOLOGY 2023; 3:100062. [PMID: 39628521 PMCID: PMC11611013 DOI: 10.1016/j.engmic.2022.100062] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/06/2024]
Abstract
Fe(II)/α-ketoglutarate (αKG)-dependent oxygenases catalyze the oxidative modification of various molecules, from DNA, RNA, and proteins to primary and secondary metabolites. They also catalyze a variety of biochemical reactions, including hydroxylation, halogenation, desaturation, epoxidation, cyclization, peroxidation, epimerization, and rearrangement. Given the versatile catalytic capability of such oxygenases, numerous studies have been conducted to characterize their functions and elucidate their structure-function relationships over the past few decades. Amino acids, particularly nonproteinogenic amino acids, are considered as important building blocks for chemical synthesis and components for natural product biosynthesis. In addition, the Fe(II)/αKG-dependent oxygenase superfamily includes important enzymes for generating amino acid derivatives, as they efficiently modify various free-standing amino acids. The recent discovery of new Fe(II)/αKG-dependent oxygenases and the repurposing of known enzymes in this superfamily have promoted the generation of useful amino acid derivatives. Therefore, this study will focus on the recent progress achieved from 2019 to 2022 to provide a clear view of the mechanism by which these enzymes have expanded the repertoire of free amino acid oxidative modifications.
Collapse
Affiliation(s)
- Hui Tao
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ikuro Abe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Han P, Chen Z, Liu Y, Ma A, Li S, Jia Y. An accurate strategy for pointing the key biocatalytic sites of bre2691A protein for modification of the brevilaterin from Brevibacillus laterosporus. Microb Cell Fact 2022; 21:196. [PMID: 36123650 PMCID: PMC9484153 DOI: 10.1186/s12934-022-01918-x] [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] [Received: 06/08/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Brevilaterin A-E, a novel class of multi-component cationic antimicrobial lipopeptides, were biosynthesized by a non-ribosomal peptides synthetase (NRPS) in Brevibacillus laterosporus. However, the antimicrobial abilities of different brevilaterin components varied greatly, and this multi-component form was impeding the scale production of the excellent component, and a little information about the brevilaterin biosynthesis mechanism was available to apply in brevilaterin design modification. In this study, we used an accurate strategy that revealed the reason for producing multi-component was the substrate selectivity of bre2691A protein being not enough specific and pinpointed the key design sites to make the specificity of bre2691A enhanced. RESULTS Bioinformatic analysis revealed that the biocatalytic site of bre2691A, which was an adenylation domain catalyzed and recognized methionine, leucine, valine and isoleucine and thus introduced them into brevilaterins and caused different components (brevilaterin A-E), was consisted of A1 ~ A10 residues named specificity-conferring code. Coupling molecular docking simulations with mutation studies identified A2 and A7 as critical residues, where determined substrate-specificity and impacted activity. The in virto activity assay showed that the A2 mutant (G193A) would lose activity against methionine and have no effect on the other three amino acids, the A7 mutant (G285C) would enhance the catalytic activity against four substrates, especially against leucine at almost a double activity. When the A2 and A7 residues were synchronously mutated, this mutant would be more focused on recognizing leucine. CONCLUSIONS An accurate strategy that combined with bioinformatics and site-directed mutation techniques revealed the pivotal site A2 and A7 positions of bre2691A protein that could be used to design and modify brevilaterins, thus further providing a reasonable direction of genetic engineering for Brevibacillus laterosporus. A deeper understanding of the function of crucial residues in the adenylation domain would make it get more accurate and highly efficient design and more fully utilized. Furthermore, it would contribute to biotechnological applications, namely for the large centralized synthesis of antimicrobial peptides, or for the optimization of their production.
Collapse
Affiliation(s)
- Panpan Han
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Zhou Chen
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Yangliu Liu
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Aijin Ma
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Siting Li
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Yingmin Jia
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
9
|
Hu S, Li Y, Zhang A, Li H, Chen K, Ouyang P. Designing of an Efficient Whole-Cell Biocatalyst System for Converting L-Lysine Into Cis-3-Hydroxypipecolic Acid. Front Microbiol 2022; 13:945184. [PMID: 35832817 PMCID: PMC9271919 DOI: 10.3389/fmicb.2022.945184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022] Open
Abstract
Cis-3-hydroxypipecolic acid (cis-3-HyPip), a key structural component of tetrapeptide antibiotic GE81112, which has attracted substantial attention for its broad antimicrobial properties and unique ability to inhibit bacterial translation initiation. In this study, a combined strategy to increase the productivity of cis-3-HyPip was investigated. First, combinatorial optimization of the ribosomal binding site (RBS) sequence was performed to tune the gene expression translation rates of the pathway enzymes. Next, in order to reduce the addition of the co-substrate α-ketoglutarate (2-OG), the major engineering strategy was to reconstitute the tricarboxylic acid (TCA) cycle of Escherichia coli to force the metabolic flux to go through GetF catalyzed reaction for 2-OG to succinate conversion, a series of engineered strains were constructed by the deletion of the relevant genes. In addition, the metabolic flux (gltA and icd) was improved and glucose concentrations were optimized to enhance the supply and catalytic efficiency of continuous 2-OG supply powered by glucose. Finally, under optimal conditions, the cis-3-HyPip titer of the best strain catalysis reached 33 mM, which was remarkably higher than previously reported.
Collapse
|
10
|
Neis N, Xie F, Krug D, Zhao H, Siebert A, Binz T, Fu C, Müller R, Kazmaier U. Stereoselective Syntheses of Deuterated Pipecolic Acids as Tools to Investigate the Stereoselectivity of the Hydroxylase GetF. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nadine Neis
- Saarland University: Universitat des Saarlandes Organic Chemistry GERMANY
| | - Feng Xie
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Daniel Krug
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Haowen Zhao
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Andreas Siebert
- Saarland University: Universitat des Saarlandes Organic Chemistry GERMANY
| | - Tina Binz
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Chengzhang Fu
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Rolf Müller
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Uli Kazmaier
- Saarland University Organic Chemistry Campus, Bldg. C4.2 66123 Saarbrücken GERMANY
| |
Collapse
|
11
|
Hu S, Yang P, Li Y, Zhang A, Chen K, Ouyang P. Biosynthesis of cis-3-hydroxypipecolic acid from L-lysine using an in vivo dual-enzyme cascade. Enzyme Microb Technol 2021; 154:109958. [PMID: 34891103 DOI: 10.1016/j.enzmictec.2021.109958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 11/13/2021] [Accepted: 11/30/2021] [Indexed: 11/03/2022]
Abstract
Cis-3-Hydroxypipecolic acid (cis-3-HyPip) is an important intermediate for the synthesis of GE81112 tetrapeptides, a small family of unusual nonribosomal peptide congeners with potent inhibitory activity against prokaryotic translation initiation. In this study, we constructed a microbial cell factory that can convert L-lysine into cis-3-hydroxypipecolic acid (cis-3-HyPip). Lysine cyclodeaminase SpLCD and Fe(II)/α-ketoglutarate (2-OG)-based oxygenase GetF were co-expressed in Escherichia coli. Plasmids with different copy numbers were used to balance the expression of these two enzymes, and the cell with the most appropriate balance of this kind for carrying plasmid pET-duet-getf-splcd was obtained. After determining the temperature (30 °C), pH (7.0), cell biomass, substrate concentration, Fe2+ concentration (10 mM), L-ascorbate concentration (10 mM), and TritonX-100 concentration (0.1% w/v) that were optimal for whole-cell catalysis, the yield of cis-3-HyPip reached as high as 25 mM (3.63 g/L).
Collapse
Affiliation(s)
- Shewei Hu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| | - Pengfan Yang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| | - Yangyang Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| | - Alei Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China.
| | - Kequan Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China.
| | - Pingkai Ouyang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| |
Collapse
|
12
|
Stout CN, Renata H. Reinvigorating the Chiral Pool: Chemoenzymatic Approaches to Complex Peptides and Terpenoids. Acc Chem Res 2021; 54:1143-1156. [PMID: 33543931 DOI: 10.1021/acs.accounts.0c00823] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biocatalytic transformations that leverage the selectivity and efficiency of enzymes represent powerful tools for the construction of complex natural products. Enabled by innovations in genome mining, bioinformatics, and enzyme engineering, synthetic chemists are now more than ever able to develop and employ enzymes to solve outstanding chemical problems, one of which is the reliable and facile generation of stereochemistry within natural product scaffolds. In recognition of this unmet need, our group has sought to advance novel chemoenzymatic strategies to both expand and reinvigorate the chiral pool. Broadly defined, the chiral pool comprises cheap, enantiopure feedstock chemicals that serve as popular foundations for asymmetric total synthesis. Among these building blocks, amino acids and enantiopure terpenes, whose core structures can be mapped onto several classes of structurally and pharmaceutically intriguing natural products, are of particular interest to the synthetic community.In this Account, we summarize recent efforts from our group in leveraging biocatalytic transformations to expand the chiral pool, as well as efforts toward the efficient application of these transformations in natural products total synthesis, the ultimate testing ground for any novel methodology. First, we describe several examples of enzymatic generation of noncanonical amino acids as means to simplify the synthesis of peptide natural products. By extracting amino acid hydroxylases from native biosynthetic pathways, we obtain efficient access to hydroxylated variants of proline, lysine, arginine, and their derivatives. The newly installed hydroxyl moiety then becomes a chemical handle that can facilitate additional complexity generation, thereby expanding the pool of amino acid-derived building blocks available for peptide synthesis. Next, we present our efforts in enzymatic C-H oxidations of diverse terpene scaffolds, in which traditional chemistry can be combined with strategic applications of biocatalysis to selectively and efficiently derivatize several commercial terpenoid skeletons. The synergistic logic of this approach enables a small handful of synthetic intermediates to provide access to a plethora of terpenoid natural product families. Taken together, these findings demonstrate the advantages of applying enzymes in total synthesis in conjunction with established methodologies, as well as toward the expansion of the chiral pool to enable facile incorporation of stereochemistry during synthetic campaigns.
Collapse
Affiliation(s)
- Carter N. Stout
- Department of Chemistry, Scripps Research, 110 Scripps Way, Jupiter, Florida 33458, United States
| | - Hans Renata
- Department of Chemistry, Scripps Research, 110 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
13
|
Zwick CR, Sosa MB, Renata H. Modular Chemoenzymatic Synthesis of GE81112 B1 and Related Analogues Enables Elucidation of Its Key Pharmacophores. J Am Chem Soc 2021; 143:1673-1679. [PMID: 33416325 DOI: 10.1021/jacs.0c13424] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The GE81112 complex has garnered much interest due to its broad antimicrobial properties and unique ability to inhibit bacterial translation initiation. Herein we report the use of a chemoenzymatic strategy to complete the first total synthesis of GE81112 B1. By pairing iron and α-ketoglutarate dependent hydroxylases found in GE81112 biosynthesis with traditional synthetic methodology, we were able to access the natural product in 11 steps (longest linear sequence). Following this strategy, 10 GE81112 B1 analogues were synthesized, allowing for identification of its key pharmacophores. A key feature of our medicinal chemistry effort is the incorporation of additional biocatalytic hydroxylations in modular analogue synthesis to rapidly enable exploration of relevant chemical space.
Collapse
Affiliation(s)
- Christian R Zwick
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Max B Sosa
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Hans Renata
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
14
|
Renata H. Exploration of Iron- and a-Ketoglutarate-Dependent Dioxygenases as Practical Biocatalysts in Natural Product Synthesis. Synlett 2021; 32:775-784. [PMID: 34413574 PMCID: PMC8372184 DOI: 10.1055/s-0040-1707320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Catalytic C─H oxidation is a powerful transformation with enormous promise to streamline access to complex molecules. In recent years, biocatalytic C─H oxidation strategies have received tremendous attention due to their potential to address unmet regio- and stereoselectivity challenges that are often encountered with the use of small-molecule-based catalysts. This Account provides an overview of recent contributions from our laboratory in this area, specifically in the use of iron- and α-ketoglutarate-dependent dioxygenases in the chemoenzymatic synthesis of complex natural products.
Collapse
Affiliation(s)
- Hans Renata
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| |
Collapse
|
15
|
Hara R, Kino K. Enzymatic reactions and microorganisms producing the various isomers of hydroxyproline. Appl Microbiol Biotechnol 2020; 104:4771-4779. [PMID: 32291491 DOI: 10.1007/s00253-020-10603-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023]
Abstract
Hydroxyproline is an industrially important compound with applications in the pharmaceutical, nutrition, and cosmetic industries. trans-4-Hydroxy-L-proline is recognized as the most abundant of the eight possible isomers (hydroxy group at C-3 or C-4, cis- or trans-configuration, and L- or D-form). However, little attention has been paid to the rare isomers, probably due to their limited availability. This mini-review provides an overview of recent advances in microbial and enzymatic processes to develop practical production strategies for various hydroxyprolines. Here, we introduce three screening strategies, namely, activity-, sequence-, and metabolite-based approaches, allowing identification of diverse proline-hydroxylating enzymes with different product specificities. All naturally occurring hydroxyproline isomers can be produced by using suitable hydroxylases in a highly regio- and stereo-selective manner. Furthermore, crystal structures of relevant hydroxylases provide much insight into their functional roles. Since hydroxylases acting on free L-proline belong to the 2-oxoglutarate-dependent dioxygenase superfamily, cellular metabolism of Escherichia coli coupled with a hydroxylase is a valuable source of 2-oxoglutarate, which is indispensable as a co-substrate in L-proline hydroxylation. Further, microbial hydroxyproline 2-epimerase may serve as a highly adaptable tool to convert L-hydroxyproline into D-hydroxyproline. KEY POINTS: • Proline hydroxylases serve as powerful tools for selectivel-proline hydroxylation. • Engineered Escherichia coli are a robust platform for hydroxyproline production. • Hydroxyproline epimerase convertsl-hydroxyproline intod-hydroxyproline.
Collapse
Affiliation(s)
- Ryotaro Hara
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan.,Laboratory of Industrial Microbiology, Graduate School of Agriculture, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Kuniki Kino
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan. .,Department of Applied Chemistry, Faculty of Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
| |
Collapse
|
16
|
Zwick CR, Sosa MB, Renata H. Characterization of a Citrulline 4-Hydroxylase from Nonribosomal Peptide GE81112 Biosynthesis and Engineering of Its Substrate Specificity for the Chemoenzymatic Synthesis of Enduracididine. Angew Chem Int Ed Engl 2019; 58:18854-18858. [PMID: 31610076 PMCID: PMC6917913 DOI: 10.1002/anie.201910659] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/06/2019] [Indexed: 12/15/2022]
Abstract
The GE81112 tetrapeptides are a small family of unusual nonribosomal peptide congeners with potent inhibitory activity against prokaryotic translation initiation. With the exception of the 3-hydroxy-l-pipecolic acid unit, little is known about the biosynthetic origins of the non-proteinogenic amino acid monomers of the natural product family. Here, we elucidate the biogenesis of the 4-hydroxy-l-citrulline unit and establish the role of an iron- and α-ketoglutarate-dependent enzyme (Fe/αKG) in the pathway. Homology modelling and sequence alignment analysis further facilitate the rational engineering of this enzyme to become a specific 4-arginine hydroxylase. We subsequently demonstrate the utility of this engineered enzyme in the synthesis of a dipeptide fragment of the antibiotic enduracidin. This work highlights the value of applying a bioinformatics-guided approach in the discovery of novel enzymes and engineering of new catalytic activity into existing ones.
Collapse
Affiliation(s)
- Christian R. Zwick
- Department of Chemistry The Scripps Research Institute 130 Scripps Way, Jupiter, FL 33458
| | - Max B. Sosa
- Department of Chemistry The Scripps Research Institute 130 Scripps Way, Jupiter, FL 33458
| | - Hans Renata
- Department of Chemistry The Scripps Research Institute 130 Scripps Way, Jupiter, FL 33458
| |
Collapse
|
17
|
Fisher B, Snodgrass HM, Jones KA, Andorfer MC, Lewis JC. Site-Selective C-H Halogenation Using Flavin-Dependent Halogenases Identified via Family-Wide Activity Profiling. ACS CENTRAL SCIENCE 2019; 5:1844-1856. [PMID: 31807686 PMCID: PMC6891866 DOI: 10.1021/acscentsci.9b00835] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Indexed: 05/19/2023]
Abstract
Enzymes are powerful catalysts for site-selective C-H bond functionalization. Identifying suitable enzymes for this task and for biocatalysis in general remains challenging, however, due to the fundamental difficulty of predicting catalytic activity from sequence information. In this study, family-wide activity profiling was used to obtain sequence-function information on flavin-dependent halogenases (FDHs). This broad survey provided a number of insights into FDH activity, including halide specificity and substrate preference, that were not apparent from the more focused studies reported to date. Regions of FDH sequence space that are most likely to contain enzymes suitable for halogenating small-molecule substrates were also identified. FDHs with novel substrate scope and complementary regioselectivity on large, three-dimensionally complex compounds were characterized and used for preparative-scale late-stage C-H functionalization. In many cases, these enzymes provide activities that required several rounds of directed evolution to accomplish in previous efforts, highlighting that this approach can achieve significant time savings for biocatalyst identification and provide advanced starting points for further evolution.
Collapse
Affiliation(s)
- Brian
F. Fisher
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Harrison M. Snodgrass
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Krysten A. Jones
- Department
of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | - Mary C. Andorfer
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jared C. Lewis
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
- E-mail:
| |
Collapse
|
18
|
Zwick CR, Sosa MB, Renata H. Characterization of a Citrulline 4‐Hydroxylase from Nonribosomal Peptide GE81112 Biosynthesis and Engineering of Its Substrate Specificity for the Chemoenzymatic Synthesis of Enduracididine. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201910659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Christian R. Zwick
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Max B. Sosa
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Hans Renata
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| |
Collapse
|
19
|
Structural basis of the nonribosomal codes for nonproteinogenic amino acid selective adenylation enzymes in the biosynthesis of natural products. ACTA ACUST UNITED AC 2019; 46:515-536. [DOI: 10.1007/s10295-018-2084-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/25/2018] [Indexed: 01/09/2023]
Abstract
Abstract
Nonproteinogenic amino acids are the unique building blocks of nonribosomal peptides (NRPs) and hybrid nonribosomal peptide–polyketides (NRP–PKs) and contribute to their diversity of chemical structures and biological activities. In the biosynthesis of NRPs and NRP–PKs, adenylation enzymes select and activate an amino acid substrate as an aminoacyl adenylate, which reacts with the thiol of the holo form of the carrier protein to afford an aminoacyl thioester as the electrophile for the condensation reaction. Therefore, the substrate specificity of adenylation enzymes is a key determinant of the structure of NRPs and NRP–PKs. Here, we focus on nonproteinogenic amino acid selective adenylation enzymes, because understanding their unique selection mechanisms will lead to accurate functional predictions and protein engineering toward the rational biosynthesis of designed molecules containing amino acids. Based on recent progress in the structural analysis of adenylation enzymes, we discuss the nonribosomal codes of nonproteinogenic amino acid selective adenylation enzymes.
Collapse
|
20
|
Khusro A, Aarti C, Barbabosa-Pliego A, Rivas-Cáceres RR, Cipriano-Salazar M. Venom as therapeutic weapon to combat dreadful diseases of 21 st century: A systematic review on cancer, TB, and HIV/AIDS. Microb Pathog 2018; 125:96-107. [PMID: 30195644 DOI: 10.1016/j.micpath.2018.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/26/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022]
Abstract
Cancer and infectious diseases are the preeminent causes of human morbidities and mortalities worldwide. At present, chemotherapy, radiotherapy, immunotherapy, and gene therapy are considered as predominant options in order to treat cancer. But these therapies provide inadequate consequences by affecting both the normal and tumor cells. On the other hand, tuberculosis (TB), and HIV (human immunodeficiency virus) infections are significant threats, causing over a million mortalities each year. The extensive applications of antibiotics have caused the microbes to acquire resistance to the existing antibiotics. With the emerging dilemma of drug resistant microbes, it has become imperative to identify novel therapeutic agents from natural sources as emphatic alternative approach. Over the past few decades, venoms derived from several reptiles, amphibians, and arthropods including snakes, scorpions, frogs, spiders, honey bees, wasps, beetles, caterpillars, ants, centipedes, and sponges have been identified as efficient therapeutics. Venoms constitute plethora of bioactive components, particularly peptides, enzymes, and other chemical entities, which exhibit a large array of anticancer and anti-pathogenic activities. This review highlights the panorama of bioactive components of animal venoms divulging the anticancer, anti-tubercular, and anti-HIV activities. In a nutshell, this context discloses the decisive role of animal venoms as alternative natural resources to combat these deadly diseases of 21st century, and propounding the plausible development of new therapeutic drugs in the present era.
Collapse
Affiliation(s)
- Ameer Khusro
- Research Department of Plant Biology and Biotechnology, Loyola College, Nungambakkam, Chennai, 600034, Tamil Nadu, India.
| | - Chirom Aarti
- Research Department of Plant Biology and Biotechnology, Loyola College, Nungambakkam, Chennai, 600034, Tamil Nadu, India
| | - Alberto Barbabosa-Pliego
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Raymundo Rene Rivas-Cáceres
- Universidad Autónoma de Ciudad Juárez, Ave. Plutarco Elías Calles No. 1210, FOVISSSTE Chamizal Cd, Juarez, C.P. 32310, Mexico
| | | |
Collapse
|
21
|
Jürjens G, Schuler SMM, Kurz M, Petit S, Couturier C, Jeannot F, Nguyen F, Wende RC, Hammann PE, Wilson DN, Bacqué E, Pöverlein C, Bauer A. Totalsynthese und Strukturkorrektur des antibiotisch wirksamen Tetrapeptids GE81112A. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201805901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Gerrit Jürjens
- Fraunhofer IME; Projektgruppe Bioressourcen; Gießen Deutschland
- Helmholtz Zentrum München; Institut für Medizinalchemie; Hannover Deutschland
| | | | - Michael Kurz
- R&D Integrated Drug Discovery; Sanofi-Aventis Deutschland GmbH; Industriepark Höchst 65926 Frankfurt am Main Deutschland
| | - Sylvain Petit
- Therapeutic Area Infectious Diseases; Sanofi R&D; Marcy L'Etoile Frankreich
- Derzeitige Adresse: UCB Pharma SA; Braine-L'Alleud Belgien
| | - Cédric Couturier
- Therapeutic Area Infectious Diseases; Sanofi R&D; Marcy L'Etoile Frankreich
| | - Frédéric Jeannot
- Therapeutic Area Infectious Diseases; Sanofi R&D; Marcy L'Etoile Frankreich
| | - Fabian Nguyen
- Gene Center; Department for Biochemistry and Center for Protein Science Munich (CiPSM); Ludwig-Maximilians-Universität München; Deutschland
- Derzeitige Adresse: Institut für Biochemie und Molekularbiologie; Universität Hamburg; Deutschland
| | | | - Peter E. Hammann
- R&D Therapeutic Area Infectious Diseases; Sanofi-Aventis Deutschland GmbH; Industriepark Höchst 65926 Frankfurt am Main Deutschland
| | - Daniel N. Wilson
- Gene Center; Department for Biochemistry and Center for Protein Science Munich (CiPSM); Ludwig-Maximilians-Universität München; Deutschland
- Derzeitige Adresse: Institut für Biochemie und Molekularbiologie; Universität Hamburg; Deutschland
| | - Eric Bacqué
- Therapeutic Area Infectious Diseases; Sanofi R&D; Marcy L'Etoile Frankreich
| | - Christoph Pöverlein
- R&D Integrated Drug Discovery; Sanofi-Aventis Deutschland GmbH; Industriepark Höchst 65926 Frankfurt am Main Deutschland
| | - Armin Bauer
- R&D Therapeutic Area Infectious Diseases; Sanofi-Aventis Deutschland GmbH; Industriepark Höchst 65926 Frankfurt am Main Deutschland
| |
Collapse
|
22
|
Jürjens G, Schuler SMM, Kurz M, Petit S, Couturier C, Jeannot F, Nguyen F, Wende RC, Hammann PE, Wilson DN, Bacqué E, Pöverlein C, Bauer A. Total Synthesis and Structural Revision of the Antibiotic Tetrapeptide GE81112A. Angew Chem Int Ed Engl 2018; 57:12157-12161. [PMID: 30004165 DOI: 10.1002/anie.201805901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Indexed: 01/14/2023]
Abstract
The total synthesis of the naturally occurring antibiotic GE81112A, a densely functionalized tetrapeptide, is reported. Comparison of spectral data with those of the natural product and the lack of biological activity of the synthesized compound led us to revise the published configuration of the 3-hydroxypipecolic acid moiety. This hypothesis was fully validated by the synthesis of the corresponding epimer.
Collapse
Affiliation(s)
- Gerrit Jürjens
- Fraunhofer IME, Projektgruppe Bioressourcen, Gießen, Germany.,Helmholtz Zentrum München, Institut für Medizinalchemie, Hannover, Germany
| | | | - Michael Kurz
- R&D Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Sylvain Petit
- Therapeutic Area Infectious Diseases, Sanofi R&D, Marcy L'Etoile, France.,Present address: UCB Pharma SA, Braine-L'Alleud, Belgium
| | - Cédric Couturier
- Therapeutic Area Infectious Diseases, Sanofi R&D, Marcy L'Etoile, France
| | - Frédéric Jeannot
- Therapeutic Area Infectious Diseases, Sanofi R&D, Marcy L'Etoile, France
| | - Fabian Nguyen
- Gene Center, Department for Biochemistry and Center for Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität, München, Germany.,Present address: Institut für Biochemie und Molekularbiologie, Universität Hamburg, Germany
| | - Raffael C Wende
- Fraunhofer IME, Projektgruppe Bioressourcen, Gießen, Germany
| | - Peter E Hammann
- R&D Therapeutic Area Infectious Diseases, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Daniel N Wilson
- Gene Center, Department for Biochemistry and Center for Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität, München, Germany.,Present address: Institut für Biochemie und Molekularbiologie, Universität Hamburg, Germany
| | - Eric Bacqué
- Therapeutic Area Infectious Diseases, Sanofi R&D, Marcy L'Etoile, France
| | - Christoph Pöverlein
- R&D Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Armin Bauer
- R&D Therapeutic Area Infectious Diseases, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| |
Collapse
|
23
|
Polikanov YS, Aleksashin NA, Beckert B, Wilson DN. The Mechanisms of Action of Ribosome-Targeting Peptide Antibiotics. Front Mol Biosci 2018; 5:48. [PMID: 29868608 PMCID: PMC5960728 DOI: 10.3389/fmolb.2018.00048] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022] Open
Abstract
The ribosome is one of the major targets in the cell for clinically used antibiotics. However, the increase in multidrug resistant bacteria is rapidly reducing the effectiveness of our current arsenal of ribosome-targeting antibiotics, highlighting the need for the discovery of compounds with new scaffolds that bind to novel sites on the ribosome. One possible avenue for the development of new antimicrobial agents is by characterization and optimization of ribosome-targeting peptide antibiotics. Biochemical and structural data on ribosome-targeting peptide antibiotics illustrates the large diversity of scaffolds, binding interactions with the ribosome as well as mechanism of action to inhibit translation. The availability of high-resolution structures of ribosomes in complex with peptide antibiotics opens the way to structure-based design of these compounds as novel antimicrobial agents.
Collapse
Affiliation(s)
- Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States.,Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL, United States
| | - Nikolay A Aleksashin
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Bertrand Beckert
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Daniel N Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
24
|
Mattay J, Hüttel W. Pipecolic Acid Hydroxylases: A Monophyletic Clade amongcis-Selective Bacterial Proline Hydroxylases that Discriminatesl-Proline. Chembiochem 2017; 18:1523-1528. [DOI: 10.1002/cbic.201700187] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Johanna Mattay
- Institute of Pharmaceutical Sciences; University of Freiburg; Albertstrasse 25 79104 Freiburg Germany
| | - Wolfgang Hüttel
- Institute of Pharmaceutical Sciences; University of Freiburg; Albertstrasse 25 79104 Freiburg Germany
| |
Collapse
|
25
|
da Mata ÉCG, Mourão CBF, Rangel M, Schwartz EF. Antiviral activity of animal venom peptides and related compounds. J Venom Anim Toxins Incl Trop Dis 2017; 23:3. [PMID: 28074089 PMCID: PMC5217322 DOI: 10.1186/s40409-016-0089-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/30/2016] [Indexed: 12/11/2022] Open
Abstract
Viruses exhibit rapid mutational capacity to trick and infect host cells, sometimes assisted through virus-coded peptides that counteract host cellular immune defense. Although a large number of compounds have been identified as inhibiting various viral infections and disease progression, it is urgent to achieve the discovery of more effective agents. Furthermore, proportionally to the great variety of diseases caused by viruses, very few viral vaccines are available, and not all are efficient. Thus, new antiviral substances obtained from natural products have been prospected, including those derived from venomous animals. Venoms are complex mixtures of hundreds of molecules, mostly peptides, that present a large array of biological activities and evolved to putatively target the biochemical machinery of different pathogens or host cellular structures. In addition, non-venomous compounds, such as some body fluids of invertebrate organisms, exhibit antiviral activity. This review provides a panorama of peptides described from animal venoms that present antiviral activity, thereby reinforcing them as important tools for the development of new therapeutic drugs.
Collapse
Affiliation(s)
- Élida Cleyse Gomes da Mata
- Laboratory of Toxinology, Department of Physiological Sciences, University of Brasília, Brasília, DF 70910-900 Brazil
| | | | - Marisa Rangel
- Laboratory of Toxinology, Department of Physiological Sciences, University of Brasília, Brasília, DF 70910-900 Brazil ; Laboratory of Immunopathology, Butantan Institute, São Paulo, SP 05508-900 Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratory of Toxinology, Department of Physiological Sciences, University of Brasília, Brasília, DF 70910-900 Brazil
| |
Collapse
|
26
|
Maio A, Brandi L, Donadio S, Gualerzi CO. The Oligopeptide Permease Opp Mediates Illicit Transport of the Bacterial P-site Decoding Inhibitor GE81112. Antibiotics (Basel) 2016; 5:antibiotics5020017. [PMID: 27231947 PMCID: PMC4929432 DOI: 10.3390/antibiotics5020017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 11/16/2022] Open
Abstract
GE81112 is a tetrapeptide antibiotic that binds to the 30S ribosomal subunit and specifically inhibits P-site decoding of the mRNA initiation codon by the fMet-tRNA anticodon. GE81112 displays excellent microbiological activity against some Gram-positive and Gram-negative bacteria in both minimal and complete, chemically defined, broth, but is essentially inactive in complete complex media. This is due to the presence of peptides that compete with the antibiotic for the oligopeptide permease system (Opp) responsible for its illicit transport into the bacterial cells as demonstrated in the cases of Escherichia coli and Bacillus subtilis. Mutations that inactivate the Opp system and confer GE81112 resistance arise spontaneously with a frequency of ca. 1 × 10(-6), similar to that of the mutants resistant to tri-l-ornithine, a known Opp substrate. On the contrary, cells expressing extrachromosomal copies of the opp genes are extremely sensitive to GE81112 in rich medium and GE81112-resistant mutations affecting the molecular target of the antibiotic were not detected upon examining >10⁸ cells of this type. However, some mutations introduced in the 16S rRNA to confer kasugamycin resistance were found to reduce the sensitivity of the cells to GE81112.
Collapse
Affiliation(s)
- Alessandro Maio
- Laboratory of Genetics, University of Camerino, via Gentile III da Varano, Camerino 62032 (MC), Italy.
| | - Letizia Brandi
- Laboratory of Genetics, University of Camerino, via Gentile III da Varano, Camerino 62032 (MC), Italy.
| | | | - Claudio O Gualerzi
- Laboratory of Genetics, University of Camerino, via Gentile III da Varano, Camerino 62032 (MC), Italy.
| |
Collapse
|
27
|
Inhibition of translation initiation complex formation by GE81112 unravels a 16S rRNA structural switch involved in P-site decoding. Proc Natl Acad Sci U S A 2016; 113:E2286-95. [PMID: 27071098 DOI: 10.1073/pnas.1521156113] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In prokaryotic systems, the initiation phase of protein synthesis is governed by the presence of initiation factors that guide the transition of the small ribosomal subunit (30S) from an unlocked preinitiation complex (30S preIC) to a locked initiation complex (30SIC) upon the formation of a correct codon-anticodon interaction in the peptidyl (P) site. Biochemical and structural characterization of GE81112, a translational inhibitor specific for the initiation phase, indicates that the main mechanism of action of this antibiotic is to prevent P-site decoding by stabilizing the anticodon stem loop of the initiator tRNA in a distorted conformation. This distortion stalls initiation in the unlocked 30S preIC state characterized by tighter IF3 binding and a reduced association rate for the 50S subunit. At the structural level we observe that in the presence of GE81112 the h44/h45/h24a interface, which is part of the IF3 binding site and forms ribosomal intersubunit bridges, preferentially adopts a disengaged conformation. Accordingly, the findings reveal that the dynamic equilibrium between the disengaged and engaged conformations of the h44/h45/h24a interface regulates the progression of protein synthesis, acting as a molecular switch that senses and couples the 30S P-site decoding step of translation initiation to the transition from an unlocked preIC to a locked 30SIC state.
Collapse
|
28
|
Skinnider MA, Dejong CA, Rees PN, Johnston CW, Li H, Webster ALH, Wyatt MA, Magarvey NA. Genomes to natural products PRediction Informatics for Secondary Metabolomes (PRISM). Nucleic Acids Res 2015; 43:9645-62. [PMID: 26442528 PMCID: PMC4787774 DOI: 10.1093/nar/gkv1012] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 09/24/2015] [Indexed: 12/05/2022] Open
Abstract
Microbial natural products are an invaluable source of evolved bioactive small molecules and pharmaceutical agents. Next-generation and metagenomic sequencing indicates untapped genomic potential, yet high rediscovery rates of known metabolites increasingly frustrate conventional natural product screening programs. New methods to connect biosynthetic gene clusters to novel chemical scaffolds are therefore critical to enable the targeted discovery of genetically encoded natural products. Here, we present PRISM, a computational resource for the identification of biosynthetic gene clusters, prediction of genetically encoded nonribosomal peptides and type I and II polyketides, and bio- and cheminformatic dereplication of known natural products. PRISM implements novel algorithms which render it uniquely capable of predicting type II polyketides, deoxygenated sugars, and starter units, making it a comprehensive genome-guided chemical structure prediction engine. A library of 57 tailoring reactions is leveraged for combinatorial scaffold library generation when multiple potential substrates are consistent with biosynthetic logic. We compare the accuracy of PRISM to existing genomic analysis platforms. PRISM is an open-source, user-friendly web application available at http://magarveylab.ca/prism/.
Collapse
Affiliation(s)
- Michael A Skinnider
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Chris A Dejong
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Philip N Rees
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Chad W Johnston
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Haoxin Li
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Andrew L H Webster
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Morgan A Wyatt
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Nathan A Magarvey
- Departments of Biochemistry and Biomedical Sciences and Chemistry and Chemical Biology, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| |
Collapse
|
29
|
Vingadassalon A, Lorieux F, Juguet M, Le Goff G, Gerbaud C, Pernodet JL, Lautru S. Natural combinatorial biosynthesis involving two clusters for the synthesis of three pyrrolamides in Streptomyces netropsis. ACS Chem Biol 2015; 10:601-10. [PMID: 25415678 DOI: 10.1021/cb500652n] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The pyrrolamides constitute a small family of secondary metabolites that are known for their ability to bind noncovalently to the DNA minor groove with some sequence specificity. To date, only a single pyrrolamide biosynthetic gene cluster has been reported, directing the synthesis of congocidine (netropsin) in Streptomyces ambofaciens. In this study, we improve our understanding of pyrrolamide biosynthesis through the identification and characterization of the gene cluster responsible for the production of distamycin in Streptomyces netropsis DSM40846. We discover that the strain produces two other pyrrolamides, the well-characterized congocidine and a congocidine/distamycin hybrid that we named disgocidine. S. netropsis DSM40846 genome analysis led to the identification of two distinct pyrrolamide-like biosynthetic gene clusters. We show here that these two clusters are reciprocally dependent for the production of the three pyrrolamide molecules. Furthermore, based on detailed functional analysis of these clusters, we propose a biosynthetic route to congocidine and distamycin and an updated model for pyrrolamide assembly. The synthesis of disgocidine, the distamycin/congocidine hybrid, appears to constitute the first example of "natural combinatorial biosynthesis" between two related biosynthetic pathways. Finally, we analyze the genomic context of the two biosynthetic gene clusters and suggest that the presently interdependent clusters result from the coevolution of two ancestral independent pyrrolamide gene clusters.
Collapse
Affiliation(s)
- Audrey Vingadassalon
- Université Paris-Sud, Institut de Génétique
et Microbiologie (UMR 8621), Orsay F-91405 Cedex, France
- CNRS, Institut de Génétique
et Microbiologie
(UMR 8621), Orsay F-91405 Cedex, France
| | - Florence Lorieux
- Université Paris-Sud, Institut de Génétique
et Microbiologie (UMR 8621), Orsay F-91405 Cedex, France
- CNRS, Institut de Génétique
et Microbiologie
(UMR 8621), Orsay F-91405 Cedex, France
| | - Maud Juguet
- Université Paris-Sud, Institut de Génétique
et Microbiologie (UMR 8621), Orsay F-91405 Cedex, France
- CNRS, Institut de Génétique
et Microbiologie
(UMR 8621), Orsay F-91405 Cedex, France
| | - Géraldine Le Goff
- CNRS, Institut de Chimie des Substances Naturelles (UPR 2301), Gif-sur-Yvette F-91198
Cedex, France
| | - Claude Gerbaud
- Université Paris-Sud, Institut de Génétique
et Microbiologie (UMR 8621), Orsay F-91405 Cedex, France
- CNRS, Institut de Génétique
et Microbiologie
(UMR 8621), Orsay F-91405 Cedex, France
| | - Jean-Luc Pernodet
- Université Paris-Sud, Institut de Génétique
et Microbiologie (UMR 8621), Orsay F-91405 Cedex, France
- CNRS, Institut de Génétique
et Microbiologie
(UMR 8621), Orsay F-91405 Cedex, France
| | - Sylvie Lautru
- Université Paris-Sud, Institut de Génétique
et Microbiologie (UMR 8621), Orsay F-91405 Cedex, France
- CNRS, Institut de Génétique
et Microbiologie
(UMR 8621), Orsay F-91405 Cedex, France
| |
Collapse
|
30
|
Kang HK, Seo CH, Park Y. Marine peptides and their anti-infective activities. Mar Drugs 2015; 13:618-54. [PMID: 25603351 PMCID: PMC4306955 DOI: 10.3390/md13010618] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/01/2015] [Indexed: 12/29/2022] Open
Abstract
Marine bioresources are a valuable source of bioactive compounds with industrial and nutraceutical potential. Numerous clinical trials evaluating novel chemotherapeutic agents derived from marine sources have revealed novel mechanisms of action. Recently, marine-derived bioactive peptides have attracted attention owing to their numerous beneficial effects. Moreover, several studies have reported that marine peptides exhibit various anti-infective activities, such as antimicrobial, antifungal, antimalarial, antiprotozoal, anti-tuberculosis, and antiviral activities. In the last several decades, studies of marine plants, animals, and microbes have revealed tremendous number of structurally diverse and bioactive secondary metabolites. However, the treatments available for many infectious diseases caused by bacteria, fungi, and viruses are limited. Thus, the identification of novel antimicrobial peptides should be continued, and all possible strategies should be explored. In this review, we will present the structures and anti-infective activity of peptides isolated from marine sources (sponges, algae, bacteria, fungi and fish) from 2006 to the present.
Collapse
Affiliation(s)
- Hee Kyoung Kang
- Department of Biomedical Science, Chosun University, Gwangju 501-759, Korea.
| | - Chang Ho Seo
- Department of Bioinformatics, Kongju National University, Kongju 314-701, Korea.
| | - Yoonkyung Park
- Department of Biomedical Science, Chosun University, Gwangju 501-759, Korea.
| |
Collapse
|
31
|
Dai YN, Zhou K, Cao DD, Jiang YL, Meng F, Chi CB, Ren YM, Chen Y, Zhou CZ. Crystal structures and catalytic mechanism of theC-methyltransferase Coq5 provide insights into a key step of the yeast coenzyme Q synthesis pathway. ACTA ACUST UNITED AC 2014; 70:2085-92. [DOI: 10.1107/s1399004714011559] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/19/2014] [Indexed: 11/11/2022]
Abstract
Saccharomyces cerevisiaeCoq5 is anS-adenosyl methionine (SAM)-dependent methyltransferase (SAM-MTase) that catalyzes the onlyC-methylation step in the coenzyme Q (CoQ) biosynthesis pathway, in which 2-methoxy-6-polyprenyl-1,4-benzoquinone (DDMQH2) is converted to 2-methoxy-5-methyl-6-polyprenyl-1,4-benzoquinone (DMQH2). Crystal structures of Coq5 were determined in the apo form (Coq5-apo) at 2.2 Å resolution and in the SAM-bound form (Coq5-SAM) at 2.4 Å resolution, representing the first pair of structures for the yeast CoQ biosynthetic enzymes. Coq5 displays a typical class I SAM-MTase structure with two minor variations beyond the core domain, both of which are considered to participate in dimerization and/or substrate recognition. Slight conformational changes at the active-site pocket were observed upon binding of SAM. Structure-based computational simulation using an analogue of DDMQH2enabled us to identify the binding pocket and entrance tunnel of the substrate. Multiple-sequence alignment showed that the residues contributing to the dimeric interface and the SAM- and DDMQH2-binding sites are highly conserved in Coq5 and homologues from diverse species. A putative catalytic mechanism of Coq5 was proposed in which Arg201 acts as a general base to initiate catalysis with the help of a water molecule.
Collapse
|
32
|
Pon CL, Fabbretti A, Brandi L. Antibiotics Targeting Translation Initiation in Prokaryotes. Antibiotics (Basel) 2013. [DOI: 10.1002/9783527659685.ch17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
33
|
Desriac F, Jégou C, Balnois E, Brillet B, Le Chevalier P, Fleury Y. Antimicrobial peptides from marine proteobacteria. Mar Drugs 2013; 11:3632-60. [PMID: 24084784 PMCID: PMC3826127 DOI: 10.3390/md11103632] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 07/30/2013] [Accepted: 08/05/2013] [Indexed: 01/03/2023] Open
Abstract
After years of inadequate use and the emergence of multidrug resistant (MDR) strains, the efficiency of "classical" antibiotics has decreased significantly. New drugs to fight MDR strains are urgently needed. Bacteria hold much promise as a source of unusual bioactive metabolites. However, the potential of marine bacteria, except for Actinomycetes and Cyanobacteria, has been largely underexplored. In the past two decades, the structures of several antimicrobial compounds have been elucidated in marine Proteobacteria. Of these compounds, polyketides (PKs), synthesised by condensation of malonyl-coenzyme A and/or acetyl-coenzyme A, and non-ribosomal peptides (NRPs), obtained through the linkage of (unusual) amino acids, have recently generated particular interest. NRPs are good examples of naturally modified peptides. Here, we review and compile the data on the antimicrobial peptides isolated from marine Proteobacteria, especially NRPs.
Collapse
Affiliation(s)
- Florie Desriac
- University of Brest, LUBEM EA 3882, SFR 148, Quimper 29000, France.
| | | | | | | | | | | |
Collapse
|
34
|
Capturing Linear Intermediates and C-Terminal Variants during Maturation of the Thiopeptide GE2270. ACTA ACUST UNITED AC 2013; 20:1067-77. [DOI: 10.1016/j.chembiol.2013.07.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/05/2013] [Accepted: 07/10/2013] [Indexed: 11/18/2022]
|
35
|
Condurso HL, Bruner SD. Structure and noncanonical chemistry of nonribosomal peptide biosynthetic machinery. Nat Prod Rep 2012; 29:1099-110. [PMID: 22729219 PMCID: PMC3442147 DOI: 10.1039/c2np20023f] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Structural biology has provided significant insights into the complex chemistry and macromolecular organization of nonribosomal peptide synthetases. In addition, novel pathways are continually described, expanding the knowledge of known biosynthetic chemistry.
Collapse
Affiliation(s)
- Heather L. Condurso
- Department of Chemistry, University of Florida, Gainesville, Florida, 32611, USA. Fax: 352 392 8758; Tel: 352 392 0525
| | - Steven D. Bruner
- Department of Chemistry, University of Florida, Gainesville, Florida, 32611, USA. Fax: 352 392 8758; Tel: 352 392 0525
| |
Collapse
|
36
|
|