1
|
Wang S, Wu M. Decoding the link between microbial secondary metabolites and colorectal cancer. Comput Biol Chem 2025; 115:108372. [PMID: 39923290 DOI: 10.1016/j.compbiolchem.2025.108372] [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: 05/10/2024] [Revised: 10/29/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Colorectal cancer (CRC) is a prevalent form of cancer in humans, with the gut microbiota playing a significant role in its pathogenesis. Although previous research has primarily focused on the role of primary metabolites produced by gut microbes in CRC development, the role of secondary metabolites remains largely unexplored. Secondary metabolites are known to mediate crucial interactions between the microbiota and the host, potentially influencing CRC progression. However, their specific relationship to CRC pathogenesis is poorly understood. To address this gap, we performed a meta-analysis using fecal metagenomic data from a cohort of CRC patients and healthy controls, aiming to identify CRC-associated microbial secondary metabolite biosynthetic gene clusters (BGCs). Our findings not only provide valuable insights into the pathogenicity and carcinogenicity of CRC but also shed light on the potential mechanisms underlying its development.
Collapse
Affiliation(s)
- Shengqin Wang
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, Wenzhou University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Subtropical Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China; College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Mingjiang Wu
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, Wenzhou University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Subtropical Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China; College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
2
|
Arias-Orozco P, Cebrián R, de Jong A, Kuipers OP. Synechococsins: Lanthipeptides acting as defensive signals to disarm offensive competitors? Microbiol Res 2025; 291:127965. [PMID: 39612774 DOI: 10.1016/j.micres.2024.127965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 12/01/2024]
Abstract
Synechococsins represent a diverse group of class II lanthipeptides from the prochlorosin family, produced by the marine picocyanobacterium Synechococcus. A single strain can produce multiple SyncA peptides through modification by SyncM, a bifunctional lanthipeptide synthetase. Despite the prevalence of these lanthipeptides in nature, their biological functions remain elusive, even for the most studied group, Prochlorococcus MIT9313. This study investigated the transcriptomic response of the marine SyncA-producing strain Synechococcus sp. RS9116 to the characterized and purified SyncA6 peptide from Synechococcus sp. MITS9509. Intriguingly, the analysis of gene expression revealed a strong down-regulation of genes that encode putative ribosomally produced antimicrobial peptides, such as coculture-responsive genes (CCRG-2) and microcin-C-like bacteriocins. This study suggests a potential biological role for synechococsins as interspecific gene modulators, improving the fitness of the producing strain in a competitive and resource-limited environment.
Collapse
Affiliation(s)
- Patricia Arias-Orozco
- Department of Molecular Genetics, University of Groningen, Groningen, Nijenborgh 7, Groningen 9747AG, the Netherlands
| | - Rubén Cebrián
- Department of Clinical Microbiology, Instituto de Investigación Biosanitaria ibs. GRANADA, San Cecilio University Hospital, Av. De la Innovación s/n, Granada 18016, Spain; CIBER de Enfermedades Infecciosas, CIBERINFEC, ISCIII, Madrid, Spain
| | - Anne de Jong
- Department of Molecular Genetics, University of Groningen, Groningen, Nijenborgh 7, Groningen 9747AG, the Netherlands
| | - Oscar P Kuipers
- Department of Molecular Genetics, University of Groningen, Groningen, Nijenborgh 7, Groningen 9747AG, the Netherlands.
| |
Collapse
|
3
|
Wieder C, Künzer M, Wiechert R, Seipp K, Andresen K, Stark P, Schüffler A, Opatz T, Thines E. Biosynthesis of the Antifungal Polyhydroxy-Polyketide Acrophialocinol. Org Lett 2025; 27:1036-1041. [PMID: 39842789 PMCID: PMC11791885 DOI: 10.1021/acs.orglett.4c04656] [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: 12/12/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025]
Abstract
Bioactivity-guided isolation identified the main antifungal compounds produced by Acrophialophora levis as the new polyhydroxy-polyketides acrophialocinol (1) and acrophialocin (2). Their biosynthesis was elucidated by heterologous reconstitution in Aspergillus oryzae and involves an α-ketoglutarate-dependent dioxygenase-catalyzed α-hydroxylation, resulting in the formation of a tertiary alcohol that is indispensable for antifungal activity. Furthermore, self-resistance toward the polyhydroxy-polyketides is mediated by a conserved RTA1-like protein encoded in the acr biosynthetic gene cluster.
Collapse
Affiliation(s)
- Carsten Wieder
- Institute
of Molecular Physiology, Johannes Gutenberg-University, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
- Institut
für Biotechnologie und Wirkstoff-Forschung gGmbH, Mainz, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
| | - Moritz Künzer
- Institute
of Molecular Physiology, Johannes Gutenberg-University, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
| | - Rainer Wiechert
- Department
of Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Kevin Seipp
- Department
of Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Karsten Andresen
- Institute
of Molecular Physiology, Johannes Gutenberg-University, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
| | - Petra Stark
- Institut
für Biotechnologie und Wirkstoff-Forschung gGmbH, Mainz, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
| | - Anja Schüffler
- Institut
für Biotechnologie und Wirkstoff-Forschung gGmbH, Mainz, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
| | - Till Opatz
- Department
of Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Eckhard Thines
- Institute
of Molecular Physiology, Johannes Gutenberg-University, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
- Institut
für Biotechnologie und Wirkstoff-Forschung gGmbH, Mainz, Hanns-Dieter-Huesch-Weg 17, D-55128 Mainz, Germany
| |
Collapse
|
4
|
Mohite OS, Jørgensen TS, Booth TJ, Charusanti P, Phaneuf PV, Weber T, Palsson BO. Pangenome mining of the Streptomyces genus redefines species' biosynthetic potential. Genome Biol 2025; 26:9. [PMID: 39810189 PMCID: PMC11734326 DOI: 10.1186/s13059-024-03471-9] [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: 03/06/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Streptomyces is a highly diverse genus known for the production of secondary or specialized metabolites with a wide range of applications in the medical and agricultural industries. Several thousand complete or nearly complete Streptomyces genome sequences are now available, affording the opportunity to deeply investigate the biosynthetic potential within these organisms and to advance natural product discovery initiatives. RESULTS We perform pangenome analysis on 2371 Streptomyces genomes, including approximately 1200 complete assemblies. Employing a data-driven approach based on genome similarities, the Streptomyces genus was classified into 7 primary and 42 secondary Mash-clusters, forming the basis for comprehensive pangenome mining. A refined workflow for grouping biosynthetic gene clusters (BGCs) redefines their diversity across different Mash-clusters. This workflow also reassigns 2729 known BGC families to only 440 families, a reduction caused by inaccuracies in BGC boundary detections. When the genomic location of BGCs is included in the analysis, a conserved genomic structure, or synteny, among BGCs becomes apparent within species and Mash-clusters. This synteny suggests that vertical inheritance is a major factor in the diversification of BGCs. CONCLUSIONS Our analysis of a genomic dataset at a scale of thousands of genomes refines predictions of BGC diversity using Mash-clusters as a basis for pangenome analysis. The observed conservation in the order of BGCs' genomic locations shows that the BGCs are vertically inherited. The presented workflow and the in-depth analysis pave the way for large-scale pangenome investigations and enhance our understanding of the biosynthetic potential of the Streptomyces genus.
Collapse
Affiliation(s)
- Omkar S Mohite
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Tue S Jørgensen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Thomas J Booth
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Pep Charusanti
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Patrick V Phaneuf
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark.
| | - Bernhard O Palsson
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
5
|
Zdouc M, Blin K, Louwen NL, Navarro J, Loureiro C, Bader C, Bailey C, Barra L, Booth T, Bozhüyük KJ, Cediel-Becerra JD, Charlop-Powers Z, Chevrette M, Chooi YH, D’Agostino P, de Rond T, Del Pup E, Duncan K, Gu W, Hanif N, Helfrich EN, Jenner M, Katsuyama Y, Korenskaia A, Krug D, Libis V, Lund G, Mantri S, Morgan K, Owen C, Phan CS, Philmus B, Reitz Z, Robinson S, Singh KS, Teufel R, Tong Y, Tugizimana F, Ulanova D, Winter J, Aguilar C, Akiyama D, Al-Salihi SA, Alanjary M, Alberti F, Aleti G, Alharthi S, Rojo MA, Arishi A, Augustijn H, Avalon N, Avelar-Rivas J, Axt K, Barbieri H, Barbosa J, Barboza Segato LG, Barrett S, Baunach M, Beemelmanns C, Beqaj D, Berger T, Bernaldo-Agüero J, Bettenbühl S, Bielinski V, Biermann F, Borges R, Borriss R, Breitenbach M, Bretscher K, Brigham M, Buedenbender L, Bulcock B, Cano-Prieto C, Capela J, Carrion V, Carter R, Castelo-Branco R, Castro-Falcón G, Chagas F, Charria-Girón E, Chaudhri AA, Chaudhry V, Choi H, Choi Y, Choupannejad R, Chromy J, Donahey MC, Collemare J, Connolly J, Creamer K, Crüsemann M, Cruz A, Cumsille A, Dallery JF, Damas-Ramos L, Damiani T, de Kruijff M, Martín BD, Sala GD, Dillen J, Doering D, Dommaraju S, Durusu S, Egbert S, Ellerhorst M, Faussurier B, Fetter A, Feuermann M, Fewer D, Foldi J, Frediansyah A, Garza E, Gavriilidou A, Gentile A, Gerke J, Gerstmans H, Gomez-Escribano JP, González-Salazar L, Grayson N, Greco C, Gomez JG, Guerra S, Flores SG, Gurevich A, Gutiérrez-García K, Hart L, Haslinger K, He B, Hebra T, Hemmann J, Hindra H, Höing L, Holland D, Holme J, Horch T, Hrab P, Hu J, Huynh TH, Hwang JY, Iacovelli R, Iftime D, Iorio M, Jayachandran S, Jeong E, Jing J, Jung J, Kakumu Y, Kalkreuter E, Kang KB, Kang S, Kim W, Kim GJ, Kim H, Kim H, Klapper M, Koetsier R, Kollten C, Kovács Á, Kriukova Y, Kubach N, Kunjapur A, Kushnareva A, Kust A, Lamber J, Larralde M, Larsen N, Launay A, Le NTH, Lebeer S, Lee BT, Lee K, Lev K, Li SM, Li YX, Licona-Cassani C, Lien A, Liu J, Lopez J, Machushynets N, Macias M, Mahmud T, Maleckis M, Martinez-Martinez AM, Mast Y, Maximo M, McBride C, McLellan R, Bhatt KM, Melkonian C, Merrild A, Metsä-Ketelä M, Mitchell D, Müller A, Nguyen GS, Nguyen H, Niedermeyer T, O’Hare J, Ossowicki A, Ostash B, Otani H, Padva L, Paliyal S, Pan X, Panghal M, Parade DS, Park J, Parra J, Rubio MP, Pham H, Pidot S, Piel J, Pourmohsenin B, Rakhmanov M, Ramesh S, Rasmussen M, Rego A, Reher R, Rice A, Rigolet A, Romero-Otero A, Rosas-Becerra L, Rosiles P, Rutz A, Ryu B, Sahadeo LA, Saldanha M, Salvi L, Sánchez-Carvajal E, Santos-Medellin C, Sbaraini N, Schoellhorn S, Schumm C, Sehnal L, Selem N, Shah A, Shishido T, Sieber S, Silviani V, Singh G, Singh H, Sokolova N, Sonnenschein E, Sosio M, Sowa S, Steffen K, Stegmann E, Streiff A, Strüder A, Surup F, Svenningsen T, Sweeney D, Szenei J, Tagirdzhanov A, Tan B, Tarnowski M, Terlouw B, Rey T, Thome N, Torres Ortega LR, Tørring T, Trindade M, Truman A, Tvilum M, Udwary D, Ulbricht C, Vader L, van Wezel G, Walmsley M, Warnasinghe R, Weddeling H, Weir AM, Williams K, Williams S, Witte T, Rocca SW, Yamada K, Yang D, Yang D, Yu J, Zhou Z, Ziemert N, Zimmer L, Zimmermann A, Zimmermann C, van der Hooft JJ, Linington R, Weber T, Medema M. MIBiG 4.0: advancing biosynthetic gene cluster curation through global collaboration. Nucleic Acids Res 2025; 53:D678-D690. [PMID: 39657789 PMCID: PMC11701617 DOI: 10.1093/nar/gkae1115] [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: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
Specialized or secondary metabolites are small molecules of biological origin, often showing potent biological activities with applications in agriculture, engineering and medicine. Usually, the biosynthesis of these natural products is governed by sets of co-regulated and physically clustered genes known as biosynthetic gene clusters (BGCs). To share information about BGCs in a standardized and machine-readable way, the Minimum Information about a Biosynthetic Gene cluster (MIBiG) data standard and repository was initiated in 2015. Since its conception, MIBiG has been regularly updated to expand data coverage and remain up to date with innovations in natural product research. Here, we describe MIBiG version 4.0, an extensive update to the data repository and the underlying data standard. In a massive community annotation effort, 267 contributors performed 8304 edits, creating 557 new entries and modifying 590 existing entries, resulting in a new total of 3059 curated entries in MIBiG. Particular attention was paid to ensuring high data quality, with automated data validation using a newly developed custom submission portal prototype, paired with a novel peer-reviewing model. MIBiG 4.0 also takes steps towards a rolling release model and a broader involvement of the scientific community. MIBiG 4.0 is accessible online at https://mibig.secondarymetabolites.org/.
Collapse
Affiliation(s)
- Mitja M Zdouc
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Kai Blin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Nico L L Louwen
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Jorge Navarro
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Catarina Loureiro
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Chantal D Bader
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Constance B Bailey
- School of Chemistry, Chemistry Building, University of Sydney, Eastern Ave, Camperdown NSW 2050, Sydney, New South Wales, Australia
| | - Lena Barra
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464 Konstanz, Germany
| | - Thomas J Booth
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Kenan A J Bozhüyük
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Myria Biosciences AG, Tech Park Basel, Hochbergstrasse 60C, 4057 Basel, Switzerland
| | - José D D Cediel-Becerra
- Department of Microbiology and Cell Science, University of Florida, 1355 Museum Drive, Gainesville, Florida, 32611, USA
| | | | - Marc G Chevrette
- Department of Microbiology and Cell Science, University of Florida, 1355 Museum Drive, Gainesville, Florida, 32611, USA
- University of Florida Genetics Institute, University of Florida, 2033 Mowry Rd, Gainesville, FL 32611, USA
| | - Yit Heng Chooi
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth 6009, Australia
| | - Paul M D’Agostino
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Chair of Technical Biochemistry, Technical University of Dresden, Bergstraße 66, 01069 Dresden, Germany
| | - Tristan de Rond
- School of Chemical Sciences, University of Auckland, 23 Symonds St, Auckland 1010, New Zealand
| | - Elena Del Pup
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Katherine R Duncan
- Newcastle University, Biosciences Institute, Catherine Cookson Building, Newcastle upon Tyne, NE2 4HH, UK
| | - Wenjia Gu
- Sutro Biopharma, 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Novriyandi Hanif
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Gedung Kimia Wing 1 Lantai 3, Jalan Tanjung Kampus IPB Dramaga, Bogor, Jawa Barat 16680, Indonesia
| | - Eric J N Helfrich
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
- Senckenberg Society for Nature Research, Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Matthew Jenner
- Department of Chemistry, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Yohei Katsuyama
- Department of Biotechnology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Aleksandra Korenskaia
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Interfaculty Institute for Biomedical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Daniel Krug
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Vincent Libis
- Université Paris Cité - Inserm Unit 1284, 75015 Paris, France
- Translational Genome Mining for Natural Products, Generare Bioscience, 75011 Paris, Île-de-France, France
| | - George A Lund
- Sustainable Soils and Crops, Rothamsted Research, West Common, Harpenden, Hertfordshire, AL5 2JQ, UK
| | - Shrikant Mantri
- Computational Biology Lab, National Agri-Food and Biomanufacturing Institute (NABI), Sector 81, S.A.S. Nagar, Mohali, Punjab 140306, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad - 121001 Haryana (NCR Delhi), India
| | - Kalindi D Morgan
- Department of Chemistry and Biochemistry, University of Northern British Columbia, 3333 University Way, Prince George, BC, V2N 4Z9, Canada
| | - Charlotte Owen
- Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK
| | - Chin-Soon Phan
- Latvian Institute of Organic Synthesis, Aizkraukles street 21, LV-1006 Riga, Latvia
| | - Benjamin Philmus
- Department of Pharmaceutical Sciences, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331-3507, USA
| | - Zachary L Reitz
- Department of Ecology, Evolution and Marine Biology, University of California, 1169 Biological Sciences II, Santa Barbara, CA 93106, USA
| | - Serina L Robinson
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology, Ueberlandstrasse 133, 8600 Duebendorf, Switzerland
| | - Kumar Saurabh Singh
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Plant-Microbe Interactions, Institute of Environmental Biology, Utrecht University, 3584 CH Utrecht, The Netherlands
- Faculty of Environment, Science and Economy, University of Exeter, TR10 9FE, Penryn, Cornwall, UK
| | - Robin Teufel
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Yaojun Tong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minhang District, Shanghai 200240, China
| | - Fidele Tugizimana
- Department of Biochemistry, University of Johannesburg, C2 Lab Building 224, Kingsway Campus, Cnr University & Kingsway Road, Auckland Park, Johannesburg 2006, South Africa
- International Research and Development (R&D) Division, Omnia Nutriology, Omnia Holdings Ltd, Building H, Monte Circle, 178 Montecasino Blvd, Fourways, Sandton, 2055, South Africa
| | - Dana Ulanova
- Department of Marine Resource Science, Faculty of Agriculture and Marine Science, Kochi University, 200 Otsu, Monobe, Nankoku-shi, Kochi, 783-8502, Japan
- Marine Core Research Institute, Kochi University, 200 Otsu, Monobe, Nankoku-shi, Kochi, 783-8502, Japan
| | - Jaclyn M Winter
- Department of Pharmacology and Toxicology, University of Utah, 30 S 2000 E, Salt Lake City, Utah, 84112, USA
| | - César Aguilar
- Industrial Genomics Laboratory, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501sur, Nuevo Leon, 64700, México
- Department of Chemistry, Purdue University, 610 Purdue Mall, West Lafayette, IN, 47907, USA
| | - Daniel Y Akiyama
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Rua Monteiro Lobato 270, Campinas, São Paulo, 13.083-862, Brazil
| | - Suhad A A Al-Salihi
- Department of Applied Sciences, University of Technology, Al-Sina’a St., 10066, Baghdad, Iraq
| | - Mohammad Alanjary
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Fabrizio Alberti
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Gajender Aleti
- Department of Food and Animal Sciences, College of Agriculture, Tennessee State University, Nashville, TN 37209, USA
| | - Shumukh A Alharthi
- Department of Biosciences, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Mariela Y Arias Rojo
- Escuela Nacional de Estudios Superiores Unidad Leon, Universidad Nacional Autonoma de Mexico, Blv. UNAM #2011, Predio El Saucillo y, Comunidad de los Tepetates, El Potrero, 37684 León de los Aldama, Gto., Mexico
| | - Amr A Arishi
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth 6009, Australia
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Hannah E Augustijn
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
| | - Nicole E Avalon
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0212, USA
| | - J Abraham Avelar-Rivas
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Mexico
| | - Kyle K Axt
- Department of Pharmaceutical Sciences, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331-3507, USA
| | - Hellen B Barbieri
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Rua Monteiro Lobato 270, Campinas, São Paulo, 13.083-862, Brazil
| | - Julio Cesar J Barbosa
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Rua Monteiro Lobato 270, Campinas, São Paulo, 13.083-862, Brazil
| | | | - Susanna E Barrett
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Matthews Ave, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, 1206 W. Gregory Drive, Urbana, IL 61801, USA
| | - Martin Baunach
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Christine Beemelmanns
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Dardan Beqaj
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Microbial Bioactive Compounds, University of Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Tim Berger
- Institute of Pharmaceutical Biology and Biotechnology, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Jordan Bernaldo-Agüero
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Chamilpa, 62210 Cuernavaca, Morelos, México
- Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Germán Amézaga S/N, Ciudad Universitaria, UNMSM, Lima 15081, Perú
| | - Sandra M Bettenbühl
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Vincent A Bielinski
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos, no 149, Rio de Janeiro, 21941-909, Brazil
| | - Friederike Biermann
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Ricardo M Borges
- Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, CCS - Av. Carlos Chagas Filho, 373 - Bloco H - Cidade Universitária, Rio de Janeiro - RJ, 21941-599, Brazil
| | - Rainer Borriss
- Department of Biology, Humboldt University Berlin, Invaliden-Str. 42, 10115 Berlin, Germany
- Institute of Marine Biotechnology (IMaB), University of Greifswald,Walter-Rathenau-Str. 49A, 17489 Greifswald, Germany
| | - Milena Breitenbach
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Kevin M Bretscher
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- Department of Crop Protection, Instituto de Hortofruticultura Subtropical y Mediterránea “La Mayora” (IHSM-UMA-CSIC), Campus Universitario de Teatinos, 29010, Málaga, Spain
- Department of Microbial Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10 6708 PB Wageningen, The Netherlands
| | - Michael W Brigham
- School of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Leeds, 6 Clarendon Way, Woodhouse, Leeds, LS2 3AA, UK
| | - Larissa Buedenbender
- CICA – Centro Interdisciplinar de Química e Bioloxía, Universidade da Coruña, As Carballeiras, s/n, Campus de Elviña, 15071 A Coruña, Spain
| | - Brodie W Bulcock
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth 6009, Australia
| | - Carolina Cano-Prieto
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - João Capela
- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
| | - Victor J Carrion
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- Department of Crop Protection, Instituto de Hortofruticultura Subtropical y Mediterránea “La Mayora” (IHSM-UMA-CSIC), Campus Universitario de Teatinos, 29010, Málaga, Spain
- Department of Microbiology, Faculty of Science, Campus Universitario de Teatinos s/n, University of Málaga, 29010 Málaga, Spain
| | - Riley S Carter
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Matthews Ave, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, 1206 W. Gregory Drive, Urbana, IL 61801, USA
| | - Raquel Castelo-Branco
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, 4450-208 Matosinhos, Portugal
| | - Gabriel Castro-Falcón
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0212, USA
| | - Fernanda O Chagas
- Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, CCS - Av. Carlos Chagas Filho, 373 - Bloco H - Cidade Universitária, Rio de Janeiro - RJ, 21941-599, Brazil
| | - Esteban Charria-Girón
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Ayesha Ahmed Chaudhri
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Vasvi Chaudhry
- Microbial Interactions in Plant Ecosystems, IMIT/ZMBP, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Hyukjae Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-ro Gyeongsan-Si, Gyeongsangbuk-do 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro Gyeongsan-Si, Gyeongsangbuk-do 38541, Republic of Korea
| | - Yukyung Choi
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Roya Choupannejad
- Fungal Natural Products, Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - Jakub Chromy
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Melinda S Chue Donahey
- Department of Microbiology and Cell Science, University of Florida, 1355 Museum Drive, Gainesville, Florida, 32611, USA
| | - Jérôme Collemare
- Fungal Natural Products, Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - Jack A Connolly
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Kaitlin E Creamer
- Innovative Genomics Institute, University of California Berkeley, 2151Berkeley Way, Berkeley, CA 94720, USA
- Department of Earth and Planetary Science, University of California, 307 McCone Hall Berkeley, CA 94720-4767, USA
| | - Max Crüsemann
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Andres Arredondo Cruz
- Escuela Nacional de Estudios Superiores Unidad Leon, Universidad Nacional Autonoma de Mexico, Blv. UNAM #2011, Predio El Saucillo y, Comunidad de los Tepetates, El Potrero, 37684 León de los Aldama, Gto., Mexico
| | - Andres Cumsille
- Department of Microbiology and Cell Science, University of Florida, 1355 Museum Drive, Gainesville, Florida, 32611, USA
| | - Jean-Felix Dallery
- Université Paris-Saclay, INRAE, UR BIOGER, 22 place de l’Agronomie, 91120 Palaiseau, France
| | - Luis Caleb Damas-Ramos
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Tito Damiani
- IOCB Prague, Czech Academy of Science, Flemingovo náměstí 542/2, 160 00 Praha 6, Czech Republic
| | - Martinus de Kruijff
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Belén Delgado Martín
- Department of Microbiology, Faculty of Science, Campus Universitario de Teatinos s/n, University of Málaga, 29010 Málaga, Spain
| | - Gerardo Della Sala
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Giardini del Molosiglio, Via A.F. Acton 55, 80133 Naples, Italy
| | - Jelle Dillen
- Department of Bioscience Engineering, Research Group Laboratory of Applied Microbiology and Biotechnology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Drew T Doering
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Shravan R Dommaraju
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Matthews Ave, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, 1206 W. Gregory Drive, Urbana, IL 61801, USA
| | - Suhan Durusu
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Susan Egbert
- Department of Chemistry, University of Manitoba, 66 Chancellors Circle, Winnipeg, MB R3T 2N2, Canada
| | - Mark Ellerhorst
- Biochemistry of Microorganisms, Faculty of Life Sciences: Food, Nutrition, and Health, University of Bayreuth, Fritz-Hornschuch-Straße 13, 95326 Kulmbach, Germany
| | - Baptiste Faussurier
- Université Paris Cité - Inserm Unit 1284, 75015 Paris, France
- Translational Genome Mining for Natural Products, Generare Bioscience, 75011 Paris, Île-de-France, France
| | - Artem Fetter
- Newcastle University, Biosciences Institute, Catherine Cookson Building, Newcastle upon Tyne, NE2 4HH, UK
| | - Marc Feuermann
- SIB Swiss Institute of Bioinformatics, Centre Medical Universitaire, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | - David P Fewer
- Department of Microbiology, University of Helsinki, Viikinkaari 9, 00017, Helsinki, Finland
| | - Jonathan Foldi
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Andri Frediansyah
- Research Center for Food Technology and Processing, Research Organization of Agriculture and Food, National Research and Innovation Agency (BRIN), Jl. Jogja - Wonosari Km 31.5, DI. Yogyakarta 55861, Indonesia
- Microbial Metabolites for Food Research Group, National Research and Innovation Agency (BRIN), Jl. Jogja - Wonosari Km 31.5, Gunungkidul, DI. Yogyakarta 55861, Indonesia
| | - Erin A Garza
- Microbial and Environmental Genomics Group, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA
| | - Athina Gavriilidou
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Interfaculty Institute for Biomedical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Andrea Gentile
- NAICONS Srl, viale Ortles 22/4, 20139 Milan, Italy
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Biosciences complex – Pavilion 02, Sciences and Technology Campus 11/A, 43124 Parma, Italy
| | - Jennifer Gerke
- Institute for Organic Chemistry, Leibniz Universität Hannover, Schneiderberg 38, 30167 Hannover, Germany
| | - Hans Gerstmans
- VIB-KU Leuven Center for Microbiology, Flanders Institute for Biotechnology, Kasteelpark Arenberg 31, 3001 Leuven, Belgium
- Department of Biology, Laboratory for Biomolecular Discovery & Engineering, KU Leuven, Kasteelpark Arenberg 31, 3001 Leuven, Belgium
- Department of Biosystems, Biosensors Group, KU Leuven, Willem de Croylaan 42, box 2428, 3001 Leuven, Belgium
| | - Juan Pablo Gomez-Escribano
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures GmbH, Inhoffenstraße 7B, 38124Braunschweig, Germany
| | - Luz A González-Salazar
- Industrial Genomics Laboratory, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501sur, Nuevo Leon, 64700, México
| | - Natalie E Grayson
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0212, USA
| | - Claudio Greco
- Department of Biosciences, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Juan E Gris Gomez
- Industrial Genomics Laboratory, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501sur, Nuevo Leon, 64700, México
| | - Sebastian Guerra
- Department of Microbiology and Cell Science, University of Florida, 1355 Museum Drive, Gainesville, Florida, 32611, USA
- University of Florida Genetics Institute, University of Florida, 2033 Mowry Rd, Gainesville, FL 32611, USA
| | - Shaday Guerrero Flores
- Centro de Ciencias Matemáticas National Autonomous University of Mexico (UNAM) Antigua Carretera a Pátzcuaro # 8701, Sin Nombre, Residencial San José de la Huerta, 58089 Morelia, Mich
| | - Alexey Gurevich
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Karina Gutiérrez-García
- Biosphere Sciences and Engineering Division, Carnegie Institution for Science, 3520 San Martin Dr, Baltimore, MD, 21218, USA
| | - Lauren Hart
- Program in Chemical Biology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, Michigan, USA
| | - Kristina Haslinger
- Department of Chemical and Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Beibei He
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Teo Hebra
- IOCB Prague, Czech Academy of Science, Flemingovo náměstí 542/2, 160 00 Praha 6, Czech Republic
| | - Jethro L Hemmann
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Hindra Hindra
- Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S4L8, Canada
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S4K1, Canada
| | - Lars Höing
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Darren C Holland
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth 6009, Australia
| | - Jonathan E Holme
- Department of Biotechnology and Nanomedicine, SINTEF Industry, P.O.Box 4760 Torgard, N-7465 Trondheim, Norway
| | - Therese Horch
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Pavlo Hrab
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Microbiology Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Jie Hu
- Corteva Agriscience, 9330 Zionsville Road, Indianapolis, Indiana 46268, USA
| | - Thanh-Hau Huynh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 101 Daehak-ro, Jongro-gu, Seoul 110-744, Korea
| | - Ji-Yeon Hwang
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, USA
| | - Riccardo Iacovelli
- Production Host Engineering Team, VTT Technical Research Centre of Finland Ltd, Maarintie 3, 02150 Espoo, Finland
| | - Dumitrita Iftime
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Microbial Bioactive Compounds, University of Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | | | - Sidharth Jayachandran
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Eunah Jeong
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Jiayi Jing
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Jung J Jung
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 101 Daehak-ro, Jongro-gu, Seoul 110-744, Korea
| | - Yuya Kakumu
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Edward Kalkreuter
- Department of Chemistry and Biochemistry, Laboratories of Molecular Recognition, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306, USA
| | - Kyo Bin Kang
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Sangwook Kang
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 101 Daehak-ro, Jongro-gu, Seoul 110-744, Korea
| | - Wonyong Kim
- Department of Applied Biology, College of Agriculture and Life Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, South Korea
| | - Geum Jin Kim
- Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro Gyeongsan-Si, Gyeongsangbuk-do 38541, Republic of Korea
- Department of Pharmacology, College of Medicine, Dongguk University, Dongdae-ro 123, Gyeongju-si, Gyeongsangbuk-do 38066, Republic of Korea
| | - Hyunwoo Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Dongguk-ro 32, Goyang 10326, Republic of Korea
| | - Hyun Uk Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Martin Klapper
- Department of Paleobiotechnology, Leibniz Institute for Natural Product Research and Infection Biology Hans Knöll Institute, Beutenbergstr. 11a, 07745 Jena, Germany
| | - Robert A Koetsier
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Cassandra Kollten
- Chair of Technical Biochemistry, Technical University of Dresden, Bergstraße 66, 01069 Dresden, Germany
| | - Ákos T Kovács
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- DTU Bioengineering, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Yelyzaveta Kriukova
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Chair of Technical Biochemistry, Technical University of Dresden, Bergstraße 66, 01069 Dresden, Germany
| | - Noel Kubach
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Interfaculty Institute for Biomedical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Aditya M Kunjapur
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Aleksandra K Kushnareva
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Andreja Kust
- Innovative Genomics Institute, University of California Berkeley, 2151Berkeley Way, Berkeley, CA 94720, USA
- Department of Earth and Planetary Science, University of California, 307 McCone Hall Berkeley, CA 94720-4767, USA
| | - Jessica Lamber
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Inhoffenstr. 7, 38124 Hannover-Braunschweig, Germany
| | - Martin Larralde
- Center of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Niels J Larsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Adrien P Launay
- Bioinformatics department, Endogenomiks, El Marqués, Querétaro, Mexico
| | | | - Sarah Lebeer
- Department of Bioscience Engineering, Research Group Laboratory of Applied Microbiology and Biotechnology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
- U-MaMi Centre of Excellence, Middelheimlaan 1, 2020 Antwerp, Belgium
| | - Byung Tae Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Kyungha Lee
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Katherine L Lev
- Program in Chemical Biology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, Michigan, USA
| | - Shu-Ming Li
- Institute of Pharmaceutical Biology and Biotechnology, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Yong-Xin Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Cuauhtémoc Licona-Cassani
- Industrial Genomics Laboratory, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501sur, Nuevo Leon, 64700, México
- Integrative Biology Research Unit, The Institute for Obesity Research, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501 sur, Nuevo Leon, 64700. México
| | - Annette Lien
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Jing Liu
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Julius Adam V Lopez
- School of Innovation and Sustainability, De La Salle University, Laguna Boulevard, LTI Spine Road, Brgys. Biñan and Malamig, 4024 Biñan City, Laguna, Philippines
- Department of Chemistry, De La Salle University, 2401 Taft Avenue, 0922 Manila, Philippines
| | | | - Marla I Macias
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, G4 0RE UK
| | - Taifo Mahmud
- Department of Pharmaceutical Sciences, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331-3507, USA
| | - Matiss Maleckis
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 221, 2800 Kgs. Lyngby, Denmark
| | - Añadir Maharai Martinez-Martinez
- Industrial Genomics Laboratory, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501sur, Nuevo Leon, 64700, México
| | - Yvonne Mast
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures GmbH, Inhoffenstraße 7B, 38124Braunschweig, Germany
- Technical University of Braunschweig, Institute of Microbiology, Rebenring 56, 38106 Braunschweig, Germany
| | - Marina F Maximo
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Rua Monteiro Lobato 270, Campinas, São Paulo, 13.083-862, Brazil
| | - Christina M McBride
- Program in Chemical Biology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, Michigan, USA
| | - Rose M McLellan
- Ferrier Research Institute, Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand
| | - Khyati Mehta Bhatt
- Fungal Natural Products, Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - Chrats Melkonian
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Aske Merrild
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Mikko Metsä-Ketelä
- Department of Life Technologies, University of Turku, Tykistökatu 6, FIN-20520 Turku, Finland
| | - Douglas A Mitchell
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Matthews Ave, Urbana, IL 61801, USA
| | - Alison V Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Inhoffenstr. 7, 38124 Hannover-Braunschweig, Germany
| | - Giang-Son Nguyen
- Department of Biotechnology and Nanomedicine, SINTEF Industry, P.O.Box 4760 Torgard, N-7465 Trondheim, Norway
| | - Hera T Nguyen
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth 6009, Australia
| | - Timo H J Niedermeyer
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Julia H O’Hare
- Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, CCS - Av. Carlos Chagas Filho, 373 - Bloco H - Cidade Universitária, Rio de Janeiro - RJ, 21941-599, Brazil
| | - Adam Ossowicki
- Department of Crop Protection, Instituto de Hortofruticultura Subtropical y Mediterránea “La Mayora” (IHSM-UMA-CSIC), Campus Universitario de Teatinos, 29010, Málaga, Spain
- Department of Microbiology, Faculty of Science, Campus Universitario de Teatinos s/n, University of Málaga, 29010 Málaga, Spain
| | - Bohdan O Ostash
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, Hrushevskoho st 4, Lviv 79005, Ukraine
- German-Ukrainian Core of Excellence in Natural Products Research, Zelena str. 20, Lviv 79005, Ukraine
| | - Hiroshi Otani
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Leo Padva
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Sunaina Paliyal
- Computational Biology Lab, National Agri-Food and Biomanufacturing Institute (NABI), Sector 81, S.A.S. Nagar, Mohali, Punjab 140306, India
| | - Xinya Pan
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- Department of Crop Protection, Instituto de Hortofruticultura Subtropical y Mediterránea “La Mayora” (IHSM-UMA-CSIC), Campus Universitario de Teatinos, 29010, Málaga, Spain
- Department of Microbial Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10 6708 PB Wageningen, The Netherlands
| | - Mohit Panghal
- Computational Biology Lab, National Agri-Food and Biomanufacturing Institute (NABI), Sector 81, S.A.S. Nagar, Mohali, Punjab 140306, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad - 121001 Haryana (NCR Delhi), India
| | - Dana S Parade
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Jiyoon Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 101 Daehak-ro, Jongro-gu, Seoul 110-744, Korea
| | - Jonathan Parra
- Centro de Investigaciones en Productos Naturales (CIPRONA), Universidad de Costa Rica, San José, 11501-2060, Costa Rica
- Centro Nacional de Innovaciones Biotecnológicas (CENIBiot), CeNAT-CONARE, 1174-1200, San José, Costa Rica
- Instituto de Investigaciones Farmacéuticas (INIFAR), Facultad de Farmacia, Universidad de Costa Rica, San José, 11501-2060, Costa Rica
| | - Marcos Pedraza Rubio
- Department of Crop Protection, Instituto de Hortofruticultura Subtropical y Mediterránea “La Mayora” (IHSM-UMA-CSIC), Campus Universitario de Teatinos, 29010, Málaga, Spain
- Department of Microbiology, Faculty of Science, Campus Universitario de Teatinos s/n, University of Málaga, 29010 Málaga, Spain
| | - Huong T Pham
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Sacha J Pidot
- Department of Microbiology and Immunology at the Doherty Institute, University of Melbourne, Grattan Street, Parkville Victoria, Victoria 3000, Australia
| | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zurich, Vladimir-Prelog Weg 4, 8093 Zurich, Switzerland
| | - Bita Pourmohsenin
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Interfaculty Institute for Biomedical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Malik Rakhmanov
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Sangeetha Ramesh
- Department of Plant Pathology, University of California Davis, One Shields Avenue, Davis, CA 95616-8751, USA
| | - Michelle H Rasmussen
- Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000, Aarhus C, Denmark
| | - Adriana Rego
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, 4450-208 Matosinhos, Portugal
| | - Raphael Reher
- Institute of Pharmaceutical Biology and Biotechnology, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Andrew J Rice
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Matthews Ave, Urbana, IL 61801, USA
| | - Augustin Rigolet
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
| | - Adriana Romero-Otero
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 221, 2800 Kgs. Lyngby, Denmark
| | - Luis Rodrigo Rosas-Becerra
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Mexico
| | - Pablo Y Rosiles
- Bioinformatics department, Endogenomiks, El Marqués, Querétaro, Mexico
| | - Adriano Rutz
- Institute for Molecular Systems Biology, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Byeol Ryu
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0212, USA
| | - Libby-Ann Sahadeo
- Department of Chemistry, University of Manitoba, 66 Chancellors Circle, Winnipeg, MB R3T 2N2, Canada
| | - Murrel Saldanha
- Department of Microbiology and Cell Science, University of Florida, 1355 Museum Drive, Gainesville, Florida, 32611, USA
| | - Luca Salvi
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Microbial Bioactive Compounds, University of Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Eduardo Sánchez-Carvajal
- Laboratorio de Microbiología Molecular y Biotecnología Ambiental, Centro de Biotecnología DAL, Universidad Técnica Federico Santa María, Avenida España 1680, 2390123, Valparaíso, Chile
| | | | - Nicolau Sbaraini
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth 6009, Australia
| | - Sydney M Schoellhorn
- Department of Chemistry and BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, USA
| | - Clemens Schumm
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Inhoffenstr. 7, 38124 Hannover-Braunschweig, Germany
- Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Ludek Sehnal
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Microbial Bioactive Compounds, University of Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
- Masaryk University, Faculty of Science, RECETOX, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - Nelly Selem
- Centro de Ciencias Matemáticas National Autonomous University of Mexico (UNAM) Antigua Carretera a Pátzcuaro # 8701, Sin Nombre, Residencial San José de la Huerta, 58089 Morelia, Mich
| | - Anjali D Shah
- School of Biological Sciences, University of Bristol, Life Sciences Building, 24 Tyndall Avenue, Bristol, BS8 1TQ, UK
- School of Chemistry, University of Bristol, Cantock's Close, Bristol, BS8 1TS, UK
| | - Tania K Shishido
- Fungal Natural Products, Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 5, 00790, Helsinki, Finland
| | - Simon Sieber
- Department of Chemistry, University of Zurich,Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Velina Silviani
- College of Pharmacy, Yeungnam University, 280 Daehak-ro Gyeongsan-Si, Gyeongsangbuk-do 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro Gyeongsan-Si, Gyeongsangbuk-do 38541, Republic of Korea
| | - Garima Singh
- Department of Biology, University of Padova, Via U. Bassi, 58/B, 35121 Padova, Italy
- Botanical Garden, University of Padova, Via Orto Botanico 15, 35123 Padova, Italy
| | - Hemant Singh
- Computational Biology Lab, National Agri-Food and Biomanufacturing Institute (NABI), Sector 81, S.A.S. Nagar, Mohali, Punjab 140306, India
| | - Nika Sokolova
- Department of Chemical and Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Eva C Sonnenschein
- Department of Biosciences, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | | | - Sven T Sowa
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Karin Steffen
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, 465 21st Ave S, Nashville, TN 37232, USA
| | - Evi Stegmann
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Microbial Bioactive Compounds, University of Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
| | - Alena B Streiff
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zurich, Vladimir-Prelog Weg 4, 8093 Zurich, Switzerland
| | - Alena Strüder
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Microbial Bioactive Compounds, University of Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Frank Surup
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Tiziana Svenningsen
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Douglas Sweeney
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0212, USA
| | - Judit Szenei
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Azat Tagirdzhanov
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Bin Tan
- Institute for Molecular Bio Science, Goethe University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, 60325 Frankfurt am Main, Germany
| | - Matthew J Tarnowski
- Department of Biosciences, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Barbara R Terlouw
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Thomas Rey
- DE SANGOSSE, Bonnel, 47480, Pont-Du-Casse, France
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, Université Toulouse III, Toulouse INP, 24 Chemin de Borde Rouge, Auzeville, 31320, Auzeville-Tolosane, France
| | - Nicola U Thome
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- Department of Biology, Laboratory for Biomolecular Discovery & Engineering, KU Leuven, Kasteelpark Arenberg 31, 3001 Leuven, Belgium
| | - Laura Rosina Torres Ortega
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Thomas Tørring
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Marla Trindade
- Institute for Microbial Biotechnology and Metagenomics, Department of Biotechnology, University of the Western Cape, Robert Sobukwe Rd, Bellville, 7535, South Africa
| | - Andrew W Truman
- Department of Molecular Microbiology, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, United Kingdome, Norwich, UK
| | - Marie Tvilum
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Daniel W Udwary
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Christoph Ulbricht
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Lisa Vader
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Gilles P van Wezel
- Institute of Biology, Leiden University, Sylviusweg 72, 2333BE Leiden, The Netherlands
- Department of Microbial Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10 6708 PB Wageningen, The Netherlands
| | - Max Walmsley
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Randika Warnasinghe
- Department of Chemistry, University of Manitoba, 66 Chancellors Circle, Winnipeg, MB R3T 2N2, Canada
| | - Heiner G Weddeling
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Angus N M Weir
- VIB-KU Leuven Center for Microbiology, Flanders Institute for Biotechnology, Kasteelpark Arenberg 31, 3001 Leuven, Belgium
- The Rosalind Franklin Institute, R113 Rutherford Appleton Laboratory, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0QX, UK
| | - Katherine Williams
- School of Biological Sciences, University of Bristol, Life Sciences Building, 24 Tyndall Avenue, Bristol, BS8 1TQ, UK
- School of Applied Sciences, University of the West of England, Frenchay Campus, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Sam E Williams
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Thomas E Witte
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Private, Ottawa, Ontario, K1N 6N5, Canada
| | - Steffaney M Wood Rocca
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0212, USA
- Microbial and Environmental Genomics Group, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA
| | - Keith Yamada
- Department of Life Technologies, University of Turku, Tykistökatu 6, FIN-20520 Turku, Finland
| | - Dong Yang
- Natural Products Discovery Center, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, USA
| | - Dongsoo Yang
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Anam-ro 145, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jingwei Yu
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen 518055, P.R. China
| | - Zhenyi Zhou
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Chaowang Raod 18, Hangzhou 310014, China
| | - Nadine Ziemert
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Interfaculty Institute for Biomedical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Lukas Zimmer
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Alina Zimmermann
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures GmbH, Inhoffenstraße 7B, 38124Braunschweig, Germany
| | - Christian Zimmermann
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorfer Strasse 1a, 1060 Vienna, Austria
| | - Justin J J van der Hooft
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Department of Biochemistry, University of Johannesburg, C2 Lab Building 224, Kingsway Campus, Cnr University & Kingsway Road, Auckland Park, Johannesburg 2006, South Africa
| | - Roger G Linington
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Marnix H Medema
- Bioinformatics Group, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
6
|
Roberts Kingman GA, Kipness JL, Rothschild LJ. Raiding nature's genetic toolbox for UV-C resistance by functional metagenomics. Sci Rep 2025; 15:223. [PMID: 39747236 PMCID: PMC11695868 DOI: 10.1038/s41598-024-83952-w] [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: 08/04/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
As we assess the habitability of other worlds, we are limited by being able to only study terrestrial life adapted to terrestrial conditions. The environments found on Earth, though tremendously diverse, do not approach the multitude of potentially habitable environments beyond Earth, and so limited terrestrial adaptive capabilities tell us little about the fundamental biochemical boundaries of life. One approach to this problem is to use experimental laboratory evolution to adapt microbes to these novel environmental conditions. This approach can be dramatically improved through functional metagenomics, large-scale introduction of foreign genetic material to screen for phenotypes in a new host organism. This takes advantage of Earth's immense biological diversity with high-throughput screening for genetic tools that can facilitate adaptation. We address a key gap in functional metagenomics work by exploring the impact of the experimental parameters chosen for functional metagenomics libraries. Experimental design dictates both fragment size and copy number, and we show that both can have outsized effects on the resultant phenotypes in non-intuitive ways. These results highlight the potential of functional metagenomics for adapting life rapidly to challenging new environments, with important implications in both astrobiology and bioindustry, while also emphasizing the impacts of decisions in experimental design.
Collapse
Affiliation(s)
| | - Justin L Kipness
- Department of Cell Biology & Biochemistry, Brown University, Providence, RI, USA
| | - Lynn J Rothschild
- NASA Ames Research Center, Planetary Systems Branch, Moffett Field, CA, USA.
| |
Collapse
|
7
|
Klapper M, Stallforth P. Accessing microbial natural products of the past. MICROLIFE 2024; 5:uqae023. [PMID: 39660047 PMCID: PMC11630838 DOI: 10.1093/femsml/uqae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/09/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Microbial natural products-low molecular weight compounds biosynthesized by microorganisms-form the foundation of important modern therapeutics, including antibiotics, immunomodulators, and anti-cancer agents. This perspective discusses and contrasts two emerging approaches for uncovering natural products of the past. On the one hand, ancestral sequence reconstruction allows recreating biosynthetic pathways that date back hundreds of millions of years. On the other hand, sequencing and de novo assembly of ancient DNA reveals the biosynthetic potential of ancient microbial communities up to 100 000 years. Together, these approaches unveil an otherwise hidden reservoir of functional and structural molecular diversity. They also offer new opportunities to study the biological function and evolution of these molecules within an archaeological context.
Collapse
Affiliation(s)
- Martin Klapper
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, Department of Paleobiotechnology, Beutenbergstraße 11a, D-07745 Jena, Germany
| | - Pierre Stallforth
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, Department of Paleobiotechnology, Beutenbergstraße 11a, D-07745 Jena, Germany
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromolecular Chemistry, Humboldtstraße 10, D-07743 Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Fürstengraben 1, D-07743 Jena, Germany
| |
Collapse
|
8
|
Gamberi C, Leverette CL, Davis AC, Ismail M, Piccialli I, Borbone N, Oliviero G, Vicidomini C, Palumbo R, Roviello GN. Oceanic Breakthroughs: Marine-Derived Innovations in Vaccination, Therapy, and Immune Health. Vaccines (Basel) 2024; 12:1263. [PMID: 39591167 PMCID: PMC11598900 DOI: 10.3390/vaccines12111263] [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: 10/10/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The vast, untapped potential of the world's oceans is revealing groundbreaking advancements in human health and vaccination. Microalgae such as Nannochloropsis spp. and Dunaliella salina are emerging as resources for recombinant vaccine development with specific and heterologous genetic tools used to boost production of functional recombinant antigens in Dunaliella salina and Nannochloropsis spp. to induce immunoprotection. In humans, several antigens produced in microalgae have shown potential in combating diseases caused by the human papillomavirus, human immunodeficiency virus, hepatitis B virus, influenza virus, Zika virus, Zaire Ebola virus, Plasmodium falciparum, and Staphylococcus aureus. For animals, microalgae-derived vaccine prototypes have been developed to fight against the foot-and-mouth disease virus, classical swine fever virus, vibriosis, white spot syndrome virus, and Histophilus somni. Marine organisms offer unique advantages, including the ability to express complex antigens and sustainable production. Additionally, the oceans provide an array of bioactive compounds that serve as therapeutics, potent adjuvants, delivery systems, and immunomodulatory agents. These innovations from the sea not only enhance vaccine efficacy but also contribute to broader immunological and general health. This review explores the transformative role of marine-derived substances in modern medicine, emphasizing their importance in the ongoing battle against infectious diseases.
Collapse
Affiliation(s)
- Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Chad L. Leverette
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Alexis C. Davis
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Moayad Ismail
- Faculty of Medicine, European University, 76 Guramishvili Ave., 0141 Tbilisi, Georgia;
| | - Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Nicola Borbone
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Giorgia Oliviero
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| | - Rosanna Palumbo
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| |
Collapse
|
9
|
Chiang CH, Wang Y, Hussain A, Brooks CL, Narayan ARH. Ancestral Sequence Reconstruction to Enable Biocatalytic Synthesis of Azaphilones. J Am Chem Soc 2024; 146:30194-30203. [PMID: 39441831 DOI: 10.1021/jacs.4c08761] [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: 10/25/2024]
Abstract
Biocatalysis can be powerful in organic synthesis but is often limited by enzymes' substrate scope and selectivity. Developing a biocatalytic step involves identifying an initial enzyme for the target reaction followed by optimization through rational design, directed evolution, or both. These steps are time consuming, resource-intensive, and require expertise beyond typical organic chemistry. Thus, an effective strategy for streamlining the process from enzyme identification to implementation is essential to expanding biocatalysis. Here, we present a strategy combining bioinformatics-guided enzyme mining and ancestral sequence reconstruction (ASR) to resurrect enzymes for biocatalytic synthesis. Specifically, we achieve an enantioselective synthesis of azaphilone natural products using two ancestral enzymes: a flavin-dependent monooxygenase (FDMO) for stereodivergent oxidative dearomatization and a substrate-selective acyltransferase (AT) for the acylation of the enzymatically installed hydroxyl group. This cascade, stereocomplementary to established chemoenzymatic routes, expands access to enantiomeric linear tricyclic azaphilones. By leveraging the co-occurrence and coevolution of FDMO and AT in azaphilone biosynthetic pathways, we identified an AT candidate, CazE, and addressed its low solubility and stability through ASR, obtaining a more soluble, stable, promiscuous, and reactive ancestral AT (AncAT). Sequence analysis revealed AncAT as a chimeric composition of its descendants with enhanced reactivity likely due to ancestral promiscuity. Flexible receptor docking and molecular dynamics simulations showed that the most reactive AncAT promotes a reactive geometry between substrates. We anticipate that our bioinformatics-guided, ASR-based approach can be broadly applied in target-oriented synthesis, reducing the time required to develop biocatalytic steps and efficiently access superior biocatalysts.
Collapse
Affiliation(s)
- Chang-Hwa Chiang
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ye Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Azam Hussain
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Charles L Brooks
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Enhanced Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Alison R H Narayan
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
10
|
Choufa C, Gascht P, Leblond H, Gauthier A, Vos M, Bontemps C, Leblond P. Conjugation Mediates Large-Scale Chromosomal Transfer in Streptomyces Driving Diversification of Antibiotic Biosynthetic Gene Clusters. Mol Biol Evol 2024; 41:msae236. [PMID: 39506544 PMCID: PMC11571958 DOI: 10.1093/molbev/msae236] [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: 07/09/2024] [Revised: 10/01/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Streptomyces are ubiquitous soil-dwelling bacteria with large, linear genomes that are of special importance as a source of metabolites used in human and veterinary medicine, agronomy, and industry. Conjugative elements (actinomycetes integrative and conjugative elements, AICEs) are the main drivers of Streptomyces Horizontal Gene Transfer. AICE transfer has long been known to be accompanied by mobilization of chromosomal DNA. However, the magnitude of DNA transfer, or the localization of acquired DNA across their linear chromosome, has remained undetermined. We here show that conjugative crossings in sympatric strains of Streptomyces result in the large-scale, genome-wide distributed replacement of up to one-third of the recipient chromosome, a phenomenon for which we propose the name "Streptomyces Chromosomal Transfer" (SCT). Such chromosome blending results in the acquisition, loss, and hybridization of Specialized Metabolite Biosynthetic Gene Clusters, leading to a novel metabolic arsenal in exconjugant offspring. Harnessing conjugation-mediated specialized metabolite biosynthesis gene cluster diversification holds great promise in the discovery of new bioactive compounds including antibiotics.
Collapse
Affiliation(s)
- Caroline Choufa
- Université de Lorraine, INRAe, DynAMic, Nancy, F-54000, France
| | - Pauline Gascht
- Université de Lorraine, INRAe, DynAMic, Nancy, F-54000, France
| | - Hugo Leblond
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, F-54000, France
| | | | - Michiel Vos
- European Centre for Environment and Human Health, Penryn Campus, Penryn TR10 9FE, UK
| | - Cyril Bontemps
- Université de Lorraine, INRAe, DynAMic, Nancy, F-54000, France
| | - Pierre Leblond
- Université de Lorraine, INRAe, DynAMic, Nancy, F-54000, France
| |
Collapse
|
11
|
Ancajas CMF, Oyedele AS, Butt CM, Walker AS. Advances, opportunities, and challenges in methods for interrogating the structure activity relationships of natural products. Nat Prod Rep 2024; 41:1543-1578. [PMID: 38912779 PMCID: PMC11484176 DOI: 10.1039/d4np00009a] [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: 02/27/2024] [Indexed: 06/25/2024]
Abstract
Time span in literature: 1985-early 2024Natural products play a key role in drug discovery, both as a direct source of drugs and as a starting point for the development of synthetic compounds. Most natural products are not suitable to be used as drugs without further modification due to insufficient activity or poor pharmacokinetic properties. Choosing what modifications to make requires an understanding of the compound's structure-activity relationships. Use of structure-activity relationships is commonplace and essential in medicinal chemistry campaigns applied to human-designed synthetic compounds. Structure-activity relationships have also been used to improve the properties of natural products, but several challenges still limit these efforts. Here, we review methods for studying the structure-activity relationships of natural products and their limitations. Specifically, we will discuss how synthesis, including total synthesis, late-stage derivatization, chemoenzymatic synthetic pathways, and engineering and genome mining of biosynthetic pathways can be used to produce natural product analogs and discuss the challenges of each of these approaches. Finally, we will discuss computational methods including machine learning methods for analyzing the relationship between biosynthetic genes and product activity, computer aided drug design techniques, and interpretable artificial intelligence approaches towards elucidating structure-activity relationships from models trained to predict bioactivity from chemical structure. Our focus will be on these latter topics as their applications for natural products have not been extensively reviewed. We suggest that these methods are all complementary to each other, and that only collaborative efforts using a combination of these techniques will result in a full understanding of the structure-activity relationships of natural products.
Collapse
Affiliation(s)
| | | | - Caitlin M Butt
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.
| | - Allison S Walker
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
12
|
Deng RX, Li HL, Sheng CL, Wang W, Hu HB, Zhang XH. Characterization of Lomofungin Gene Cluster Enables the Biosynthesis of Related Phenazine Derivatives. ACS Synth Biol 2024; 13:2982-2991. [PMID: 39250825 DOI: 10.1021/acssynbio.4c00394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Phenazine-based small molecules are nitrogen-containing heterocyclic compounds with diverse bioactivities and electron transfer properties that exhibit promising applications in pharmaceutical and electrochemical industries. However, the biosynthetic mechanism of highly substituted natural phenazines remains poorly understood. In this study, we report the direct cloning and heterologous expression of the lomofungin biosynthetic gene cluster (BGC) from Streptomyces lomondensis S015. Reconstruction and overexpression of the BGCs in Streptomyces coelicolor M1152 resulted in eight phenazine derivatives including two novel hybrid phenazine metabolites, and the biosynthetic pathway of lomofungin was proposed. Furthermore, gene deletion suggested that NAD(P)H-dependent oxidoreductase gene lomo14 is a nonessential gene in the biosynthesis of lomofungin. Cytotoxicity evaluation of the isolated phenazines and lomofungin was performed. Specifically, lomofungin shows substantial inhibition against two human cancer cells, HCT116 and 5637. These results provide insights into the biosynthetic mechanism of lomofungin, which will be useful for the directed biosynthesis of natural phenazine derivatives.
Collapse
Affiliation(s)
- Ru-Xiang Deng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hui-Ling Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao-Lan Sheng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hong-Bo Hu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- National Experimental Teaching Center for Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xue-Hong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
13
|
Hernández Delgado JG, Acedos MG, de la Calle F, Rodríguez P, García JL, Galán B. Regulation of Safracin Biosynthesis and Transport in Pseudomonas poae PMA22. Mar Drugs 2024; 22:418. [PMID: 39330299 PMCID: PMC11432991 DOI: 10.3390/md22090418] [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: 07/24/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Pseudomonas poae PMA22 produces safracins, a family of compounds with potent broad-spectrum anti-bacterial and anti-tumor activities. The safracins' biosynthetic gene cluster (BGC sac) consists of 11 ORFs organized in two divergent operons (sacABCDEFGHK and sacIJ) that are controlled by Pa and Pi promoters. Contiguous to the BGC sac, we have located a gene that encodes a putative global regulator of the LysR family annotated as MexT that was originally described as a transcriptional activator of the MexEF-OprN multidrug efflux pump in Pseudomonas. Through both in vitro and in vivo experiments, we have demonstrated the involvement of the dual regulatory system MexT-MexS on the BGC sac expression acting as an activator and a repressor, respectively. The MexEF-OprN transport system of PMA22, also controlled by MexT, was shown to play a fundamental role in the metabolism of safracin. The overexpression of mexEF-oprN in PMA22 resulted in fourfold higher production levels of safracin. These results illustrate how a pleiotropic regulatory system can be critical to optimizing the production of tailored secondary metabolites, not only through direct interaction with the BGC promoters, but also by controlling their transport.
Collapse
Affiliation(s)
- J Gerardo Hernández Delgado
- Department of Biothecnology, Centro de Investigaciones Biológicas Margarita Salas, Agencia Estatal Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Miguel G Acedos
- Department of Biothecnology, Centro de Investigaciones Biológicas Margarita Salas, Agencia Estatal Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | | | - Pilar Rodríguez
- Research and Development Department, PharmaMar S.A., 28770 Madrid, Spain
| | - José Luis García
- Department of Biothecnology, Centro de Investigaciones Biológicas Margarita Salas, Agencia Estatal Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Beatriz Galán
- Department of Biothecnology, Centro de Investigaciones Biológicas Margarita Salas, Agencia Estatal Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| |
Collapse
|
14
|
Salamzade R, Tran PQ, Martin C, Manson AL, Gilmore MS, Earl AM, Anantharaman K, Kalan LR. zol & fai: large-scale targeted detection and evolutionary investigation of gene clusters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.07.544063. [PMID: 37333121 PMCID: PMC10274777 DOI: 10.1101/2023.06.07.544063] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Many universally and conditionally important genes are genomically aggregated within clusters. Here, we introduce fai and zol, which together enable large-scale comparative analysis of different types of gene clusters and mobile-genetic elements (MGEs), such as biosynthetic gene clusters (BGCs) or viruses. Fundamentally, they overcome a current bottleneck to reliably perform comprehensive orthology inference at large scale across broad taxonomic contexts and thousands of genomes. First, fai allows the identification of orthologous instances of a query gene cluster of interest amongst a database of target genomes. Subsequently, zol enables reliable, context-specific inference of ortholog groups for individual protein-encoding genes across gene cluster instances. In addition, zol performs functional annotation and computes a variety of evolutionary statistics for each inferred ortholog group. Importantly, in comparison to tools for visual exploration of homologous relationships between gene clusters, zol can scale to thousands of gene cluster instances and produce detailed reports that are easy to digest. To showcase fai and zol, we apply them for: (i) longitudinal tracking of a virus in metagenomes, (ii) discovering novel population-level genetic insights of two common BGCs in the fungal species Aspergillus flavus, and (iii) uncovering large-scale evolutionary trends of a virulence-associated gene cluster across thousands of genomes from a diverse bacterial genus.
Collapse
Affiliation(s)
- Rauf Salamzade
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Patricia Q. Tran
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Freshwater and Marine Science Doctoral Program, University of Wisconsin-Madison, WI, USA
| | - Cody Martin
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Abigail L. Manson
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Michael S. Gilmore
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School and Mass Eye and Ear, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School and Mass Eye and Ear, Boston, Massachusetts, USA
| | - Ashlee M. Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Lindsay R. Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
15
|
Li Z, Ióca LP, He R, Donia MS. Natural diversifying evolution of nonribosomal peptide synthetases in a defensive symbiont reveals nonmodular functional constraints. PNAS NEXUS 2024; 3:pgae384. [PMID: 39346623 PMCID: PMC11428043 DOI: 10.1093/pnasnexus/pgae384] [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: 03/04/2024] [Accepted: 07/19/2024] [Indexed: 10/01/2024]
Abstract
The modular architecture of nonribosomal peptide synthetases (NRPSs) has inspired efforts to study their evolution and engineering. In this study, we analyze in detail a unique family of NRPSs from the defensive intracellular bacterial symbiont, Candidatus Endobryopsis kahalalidifaciens (Ca. E. kahalalidifaciens). We show that intensive and indiscriminate recombination events erase trivial sequence covariations induced by phylogenetic relatedness, revealing nonmodular functional constraints and clear recombination units. Moreover, we reveal unique substrate specificity determinants for multiple enzymatic domains, allowing us to accurately predict and experimentally discover the products of an orphan NRPS in Ca. E. kahalalidifaciens directly from environmental samples of its algal host. Finally, we expanded our analysis to 1,531 diverse NRPS pathways and revealed similar functional constraints to those observed in Ca. E. kahalalidifaciens' NRPSs. Our findings reveal the sequence bases of genetic exchange, functional constraints, and substrate specificity in Ca. E. kahalalidifaciens' NRPSs, and highlight them as a uniquely primed system for diversifying evolution.
Collapse
Affiliation(s)
- Zhiyuan Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking 8 University, Beijing 100871, China
- Center for the Physics of Biological Function, Princeton University, Princeton, NJ 08544, USA
| | - Laura P Ióca
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Ruolin He
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
16
|
Wang M, Li Y, Pan T, Jia N. Plant natural compounds in the cancer treatment: A systematic bibliometric analysis. Heliyon 2024; 10:e34462. [PMID: 39104486 PMCID: PMC11298917 DOI: 10.1016/j.heliyon.2024.e34462] [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: 04/13/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024] Open
Abstract
Plant-derived natural compounds are significant resources for the discovery of potential anticancer drugs. While research in the plant-based anticancer field has surged in recent years, systematic bibliometric analyses covering a longer period and containing up-to-date publications remain scarce. Here, we conducted a bibliometric analysis of literature on the anticancer properties of plant natural compounds over the past three decades, leveraging the bibliometric framework and open-access platform, KNIME. Our findings showed that the number of plant anticancer-related publications underwent an accelerating growth from 1992 to 2023. The country and institution analyses revealed that countries with traditional medical systems contributed a large portion of publications in the plant anticancer field, such as India, China, and South Korea. This study also highlighted the top ten eminent researchers and publications, assisting researchers in identifying pivotal literature. The primary publications were domains of chemistry and biology-related fields, such as Pharmacology & Pharmacy, Plant Sciences, and Biochemistry & Molecular Biology. Additionally, we noted that flavonoids have been focal plant compounds in anticancer, with strong anticancer potential. Our study provides new insights into the progress and trends in the plant anticancer field and will assist researchers in grasping the future research direction.
Collapse
Affiliation(s)
- Mengting Wang
- Ningbo Key Laboratory of Agricultural Germplasm Resources Mining and Environmental Regulation, College of Science and Technology, Ningbo University, Cixi, China
| | - Yinshuai Li
- School of Design, Shanghai Jiao Tong University, Shanghai, China
| | - Tiejun Pan
- Ningbo Key Laboratory of Agricultural Germplasm Resources Mining and Environmental Regulation, College of Science and Technology, Ningbo University, Cixi, China
| | - Nan Jia
- Center for Systems Integration and Sustainability, Department of Fisheries and Wildlife, Environmental Science and Policy Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
17
|
Wu Z, Wang W, Li J, Ma C, Chen L, Che Q, Zhang G, Zhu T, Li D. Evolution-Based Discovery of Polyketide Acylated Valine from a Cytochalasin-Like Gene Cluster in Simplicillium lamelliciola HDN13430. JOURNAL OF NATURAL PRODUCTS 2024; 87:1222-1229. [PMID: 38447096 DOI: 10.1021/acs.jnatprod.3c01202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Utilizing a gene evolution-oriented approach for gene cluster mining, a cryptic cytochalasin-like gene cluster (sla) in Antarctic-derived Simplicillium lamelliciola HDN13430 was identified. Compared with the canonical cytochalasin biosynthetic gene clusters (BGCs), the sla gene cluster lacks the key α,β-hydrolase gene. Heterologous expression of the sla gene cluster led to the discovery of a new compound, slamysin (1), characterized by an N-acylated amino acid structure and demonstrating weak anti-Bacillus cereus activity. These findings underscore the potential of genetic evolution in uncovering novel compounds and indicating specific adaptive evolution within specialized habitats.
Collapse
Affiliation(s)
- Zuodong Wu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
| | - Wenxue Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
| | - Jilong Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
| | - Chuanteng Ma
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
| | - Liangzhen Chen
- Qingdao Vland Biotech Group Co., Ltd. Qingdao, Shandong 266102, People's Republic of China
| | - Qian Che
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
| | - Guojian Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, Shandong 266237, People's Republic of China
| | - Tianjiao Zhu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Sanya, Hainan 572025, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, Shandong 266237, People's Republic of China
| |
Collapse
|
18
|
Arias-Orozco P, Zhou L, Yi Y, Cebrián R, Kuipers OP. Uncovering the diversity and distribution of biosynthetic gene clusters of prochlorosins and other putative RiPPs in marine Synechococcus strains. Microbiol Spectr 2024; 12:e0361123. [PMID: 38088546 PMCID: PMC10783134 DOI: 10.1128/spectrum.03611-23] [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: 10/12/2023] [Accepted: 11/06/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE Genome mining studies have revealed the remarkable combinatorial diversity of ribosomally synthesized and post-translationally modified peptides (RiPPs) in marine bacteria, including prochlorosins. However, mining strategies also prove valuable in investigating the genomic landscape of associated genes within biosynthetic gene cluster (BGC) specific to targeted RiPPs of interest. Our study contributes to the enrichment of knowledge regarding prochlorosin diversity. It offers insights into potential mechanisms involved in their biosynthesis and modification, such as hyper-modification, which may give rise to active lantibiotics. Additionally, our study uncovers putative novel promiscuous post-translational enzymes, thereby expanding the chemical space explored within the Synechococcus genus. Moreover, this research extends the applications of mining techniques beyond the discovery of new RiPP-like clusters, allowing for a deeper understanding of genomics and diversity. Furthermore, it holds the potential to reveal previously unknown functions within the intriguing RiPP families, particularly in the case of prochlorosins.
Collapse
Affiliation(s)
- Patricia Arias-Orozco
- Department of Molecular Genetics, University of Groningen, Nijenborgh, Groningen, The Netherlands
| | - Lu Zhou
- Department of Molecular Genetics, University of Groningen, Nijenborgh, Groningen, The Netherlands
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Yunhai Yi
- Department of Molecular Genetics, University of Groningen, Nijenborgh, Groningen, The Netherlands
| | - Rubén Cebrián
- Department of Molecular Genetics, University of Groningen, Nijenborgh, Groningen, The Netherlands
- Department of Clinical Microbiology, Instituto de Investigación Biosanitaria ibs.GRANADA, San Cecilio University Hospital, Granada, Spain
- CIBER de Enfermedades Infecciosas, CIBERINFEC, ISCIII, Madrid, Spain
| | - Oscar P. Kuipers
- Department of Molecular Genetics, University of Groningen, Nijenborgh, Groningen, The Netherlands
| |
Collapse
|
19
|
Jaarsma AH, Zervas A, Sipes K, Campuzano Jiménez F, Smith AC, Svendsen LV, Thøgersen MS, Stougaard P, Benning LG, Tranter M, Anesio AM. The undiscovered biosynthetic potential of the Greenland Ice Sheet microbiome. Front Microbiol 2023; 14:1285791. [PMID: 38149278 PMCID: PMC10749974 DOI: 10.3389/fmicb.2023.1285791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/24/2023] [Indexed: 12/28/2023] Open
Abstract
The Greenland Ice Sheet is a biome which is mainly microbially driven. Several different niches can be found within the glacial biome for those microbes able to withstand the harsh conditions, e.g., low temperatures, low nutrient conditions, high UV radiation in summer, and contrasting long and dark winters. Eukaryotic algae can form blooms during the summer on the ice surface, interacting with communities of bacteria, fungi, and viruses. Cryoconite holes and snow are also habitats with their own microbial community. Nevertheless, the microbiome of supraglacial habitats remains poorly studied, leading to a lack of representative genomes from these environments. Under-investigated extremophiles, like those living on the Greenland Ice Sheet, may provide an untapped reservoir of chemical diversity that is yet to be discovered. In this study, an inventory of the biosynthetic potential of these organisms is made, through cataloging the presence of biosynthetic gene clusters in their genomes. There were 133 high-quality metagenome-assembled genomes (MAGs) and 28 whole genomes of bacteria obtained from samples of the ice sheet surface, cryoconite, biofilm, and snow using culturing-dependent and -independent approaches. AntiSMASH and BiG-SCAPE were used to mine these genomes and subsequently analyze the resulting predicted gene clusters. Extensive sets of predicted Biosynthetic Gene Clusters (BGCs) were collected from the genome collection, with limited overlap between isolates and MAGs. Additionally, little overlap was found in the biosynthetic potential among different environments, suggesting specialization of organisms in specific habitats. The median number of BGCs per genome was significantly higher for the isolates compared to the MAGs. The most talented producers were found among Proteobacteria. We found evidence for the capacity of these microbes to produce antimicrobials, carotenoid pigments, siderophores, and osmoprotectants, indicating potential survival mechanisms to cope with extreme conditions. The majority of identified BGCs, including those in the most prevalent gene cluster families, have unknown functions, presenting a substantial potential for bioprospecting. This study underscores the diverse biosynthetic potential in Greenland Ice Sheet genomes, revealing insights into survival strategies and highlighting the need for further exploration and characterization of these untapped resources.
Collapse
Affiliation(s)
- Ate H. Jaarsma
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Athanasios Zervas
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Katie Sipes
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | | | | | | | | | - Peter Stougaard
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Liane G. Benning
- German Research Centre for Geosciences, Helmholtz Centre Potsdam, Potsdam, Germany
- Department of Earth Sciences, Freie Universität Berlin, Berlin, Germany
| | - Martyn Tranter
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | | |
Collapse
|
20
|
Chen H, Bai X, Sun T, Wang X, Zhang Y, Bian X, Zhou H. The Genomic-Driven Discovery of Glutarimide-Containing Derivatives from Burkholderia gladioli. Molecules 2023; 28:6937. [PMID: 37836780 PMCID: PMC10574677 DOI: 10.3390/molecules28196937] [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: 08/04/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Glutarimide-containing polyketides exhibiting potent antitumor and antimicrobial activities were encoded via conserved module blocks in various strains that favor the genomic mining of these family compounds. The bioinformatic analysis of the genome of Burkholderia gladioli ATCC 10248 showed a silent trans-AT PKS biosynthetic gene cluster (BGC) on chromosome 2 (Chr2C8), which was predicted to produce new glutarimide-containing derivatives. Then, the silent polyketide synthase gene cluster was successfully activated via in situ promoter insertion and heterologous expression. As a result, seven glutarimide-containing analogs, including five new ones, gladiofungins D-H (3-7), and two known gladiofungin A/gladiostatin (1) and 2 (named gladiofungin C), were isolated from the fermentation of the activated mutant. Their structures were elucidated through the analysis of HR-ESI-MS and NMR spectroscopy. The structural diversities of gladiofungins may be due to the degradation of the butenolide group in gladiofungin A (1) during the fermentation and extraction process. Bioactivity screening showed that 2 and 4 had moderate anti-inflammatory activities. Thus, genome mining combined with promoter engineering and heterologous expression were proved to be effective strategies for the pathway-specific activation of the silent BGCs for the directional discovery of new natural products.
Collapse
Affiliation(s)
- Hanna Chen
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
- School of Medicine, Linyi University, Shuangling Road, Linyi 276000, China
| | - Xianping Bai
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
| | - Tao Sun
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
| | - Xingyan Wang
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
| | - Youming Zhang
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoying Bian
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
| | - Haibo Zhou
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (H.C.); (X.B.); (T.S.); (X.W.)
| |
Collapse
|
21
|
Arsın S, Delbaje E, Jokela J, Wahlsten M, Farrar ZM, Permi P, Fewer D. A Plastic Biosynthetic Pathway for the Production of Structurally Distinct Microbial Sunscreens. ACS Chem Biol 2023; 18:1959-1967. [PMID: 37603862 PMCID: PMC10510106 DOI: 10.1021/acschembio.3c00112] [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: 02/20/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023]
Abstract
Mycosporine-like amino acids (MAAs) are small, colorless, and water-soluble secondary metabolites. They have high molar extinction coefficients and a unique UV radiation absorption mechanism that make them effective sunscreens. Here we report the discovery of two structurally distinct MAAs from the lichen symbiont strain Nostoc sp. UHCC 0926. We identified these MAAs as aplysiapalythine E (C23H38N2O15) and tricore B (C34H53N4O15) using a combination of high-resolution liquid chromatography-mass spectrometry (HR-LCMS) analysis and nuclear magnetic resonance (NMR) spectroscopy. We obtained a 8.3 Mb complete genome sequence of Nostoc sp. UHCC 0926 to gain insights into the genetic basis for the biosynthesis of these two structural distinct MAAs. We identified MAA biosynthetic genes encoded in three separate locations of the genome. The organization of biosynthetic enzymes in Nostoc sp. UHCC 0926 necessitates a branched biosynthetic pathway to produce two structurally distinct MAAs. We detected the presence of such discontiguous MAA biosynthetic gene clusters in 12% of the publicly available complete cyanobacterial genomes. Bioinformatic analysis of public MAA biosynthetic gene clusters suggests that they are subject to rapid evolutionary processes resulting in highly plastic biosynthetic pathways that are responsible for the chemical diversity in this family of microbial sunscreens.
Collapse
Affiliation(s)
- Sıla Arsın
- University
of Helsinki, Department of Microbiology,
Faculty of Agriculture and Forestry, 00014 Helsinki, Finland
| | - Endrews Delbaje
- University
of São Paulo, Center for Nuclear
Energy in Agriculture, Avenida Centenário 303, 13400-970 Piracicaba, São Paulo, Brazil
| | - Jouni Jokela
- University
of Helsinki, Department of Microbiology,
Faculty of Agriculture and Forestry, 00014 Helsinki, Finland
| | - Matti Wahlsten
- University
of Helsinki, Department of Microbiology,
Faculty of Agriculture and Forestry, 00014 Helsinki, Finland
| | - Zoë M. Farrar
- University
of Helsinki, Department of Microbiology,
Faculty of Agriculture and Forestry, 00014 Helsinki, Finland
| | - Perttu Permi
- Department
of Chemistry, University of Jyväskylä, 40014 Jyväskylä, Finland
- Department
of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - David Fewer
- University
of Helsinki, Department of Microbiology,
Faculty of Agriculture and Forestry, 00014 Helsinki, Finland
| |
Collapse
|
22
|
Wan J, Ma N, Yuan H. Recent advances in the direct cloning of large natural product biosynthetic gene clusters. ENGINEERING MICROBIOLOGY 2023; 3:100085. [PMID: 39628928 PMCID: PMC11611023 DOI: 10.1016/j.engmic.2023.100085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 12/06/2024]
Abstract
Large-scale genome-mining analyses have revealed that microbes potentially harbor a huge reservoir of uncharacterized natural product (NP) biosynthetic gene clusters (BGCs), and this has spurred a renaissance of novel drug discovery. However, the majority of these BGCs are often poorly or not at all expressed in their native hosts under laboratory conditions, and thus are regarded as silent/orphan BGCs. Currently, connecting silent BGCs to their corresponding NPs quickly and on a large scale is particularly challenging because of the lack of universal strategies and enabling technologies. Generally, the heterologous host-based genome mining strategy is believed to be a suitable alternative to the native host-based approach for prioritization of the vast and ever-increasing number of uncharacterized BGCs. In the last ten years, a variety of methods have been reported for the direct cloning of BGCs of interest, which is the first and rate-limiting step in the heterologous expression strategy. Essentially, each method requires that the following three issues be resolved: 1) how to prepare genomic DNA; 2) how to digest the bilateral boundaries for release of the target BGC; and 3) how to assemble the BGC and the capture vector. Here, we summarize recent reports regarding how to directly capture a BGC of interest and briefly discuss the advantages and disadvantages of each method, with an emphasis on the notion that direct cloning is very beneficial for accelerating genome mining research and large-scale drug discovery.
Collapse
Affiliation(s)
- Jiaying Wan
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Nan Ma
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Hua Yuan
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| |
Collapse
|
23
|
Ahearne A, Phillips KE, Knehans T, Hoing M, Dowd SE, Stevens DC. Chromosomal organization of biosynthetic gene clusters, including those of nine novel species, suggests plasticity of myxobacterial specialized metabolism. Front Microbiol 2023; 14:1227206. [PMID: 37601375 PMCID: PMC10435759 DOI: 10.3389/fmicb.2023.1227206] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/11/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Natural products discovered from bacteria provide critically needed therapeutic leads for drug discovery, and myxobacteria are an established source for metabolites with unique chemical scaffolds and biological activities. Myxobacterial genomes accommodate an exceptional number and variety of biosynthetic gene clusters (BGCs) which encode for features involved in specialized metabolism. Methods In this study, we describe the collection, sequencing, and genome mining of 20 myxobacteria isolated from rhizospheric soil samples collected in North America. Results Nine isolates were determined to be novel species of myxobacteria including representatives from the genera Archangium, Myxococcus, Nannocystis, Polyangium, Pyxidicoccus, Sorangium, and Stigmatella. Growth profiles, biochemical assays, and descriptions were provided for all proposed novel species. We assess the BGC content of all isolates and observe differences between Myxococcia and Polyangiia clusters. Discussion Continued discovery and sequencing of novel myxobacteria from the environment provide BGCs for the genome mining pipeline. Utilizing complete or near-complete genome sequences, we compare the chromosomal organization of BGCs of related myxobacteria from various genera and suggest that the spatial proximity of hybrid, modular clusters contributes to the metabolic adaptability of myxobacteria.
Collapse
Affiliation(s)
- Andrew Ahearne
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Kayleigh E. Phillips
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Thomas Knehans
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Miranda Hoing
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Scot E. Dowd
- Molecular Research LP (MR DNA), Shallowater, TX, United States
| | - David Cole Stevens
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, United States
| |
Collapse
|
24
|
Barona-Gómez F, Chevrette MG, Hoskisson PA. On the evolution of natural product biosynthesis. Adv Microb Physiol 2023; 83:309-349. [PMID: 37507161 DOI: 10.1016/bs.ampbs.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Natural products are the raw material for drug discovery programmes. Bioactive natural products are used extensively in medicine and agriculture and have found utility as antibiotics, immunosuppressives, anti-cancer drugs and anthelminthics. Remarkably, the natural role and what mechanisms drive evolution of these molecules is relatively poorly understood. The exponential increase in genome and chemical data in recent years, coupled with technical advances in bioinformatics and genetics have enabled progress to be made in understanding the evolution of biosynthetic gene clusters and the products of their enzymatic machinery. Here we discuss the diversity of natural products, incorporating the mechanisms that govern evolution of metabolic pathways and how this can be applied to biosynthetic gene clusters. We build on the nomenclature of natural products in terms of primary, integrated, secondary and specialised metabolism and place this within an ecology-evolutionary-developmental biology framework. This eco-evo-devo framework we believe will help to clarify the nature and use of the term specialised metabolites in the future.
Collapse
Affiliation(s)
| | - Marc G Chevrette
- Department of Microbiology and Cell Sciences, University of Florida, Museum Drive, Gainesville, FL, United States; University of Florida Genetics Institute, University of Florida, Mowry Road, Gainesville, FL, United States
| | - Paul A Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Cathedral Street, Glasgow, United Kingdom.
| |
Collapse
|
25
|
Ahearne A, Phillips K, Knehans T, Hoing M, Dowd SE, Stevens DC. Chromosomal organization of biosynthetic gene clusters suggests plasticity of myxobacterial specialized metabolism including descriptions for nine novel species: Archangium lansinium sp. nov., Myxococcus landrumus sp. nov., Nannocystis bainbridgea sp. nov., Nannocystis poenicansa sp. nov., Nannocystis radixulma sp. nov., Polyangium mundeleinium sp. nov., Pyxidicoccus parkwaysis sp. nov., Sorangium aterium sp. nov., Stigmatella ashevillena sp. nov. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531766. [PMID: 36945379 PMCID: PMC10028903 DOI: 10.1101/2023.03.08.531766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Natural products discovered from bacteria provide critically needed therapeutic leads for drug discovery, and myxobacteria are an established source for metabolites with unique chemical scaffolds and biological activities. Myxobacterial genomes accommodate an exceptional number and variety of biosynthetic gene clusters (BGCs) which encode for features involved in specialized metabolism. Continued discovery and sequencing of novel myxobacteria from the environment provides BGCs for the genome mining pipeline. Herein, we describe the collection, sequencing, and genome mining of 20 myxobacteria isolated from rhizospheric soil samples collected in North America. Nine isolates where determined to be novel species of myxobacteria including representatives from the genera Archangium, Myxococcus, Nannocystis, Polyangium, Pyxidicoccus, Sorangium, and Stigmatella. Growth profiles, biochemical assays, and descriptions are provided for all proposed novel species. We assess the BGC content of all isolates and observe differences between Myxococcia and Polyangiia clusters. Utilizing complete or near complete genome sequences we compare the chromosomal organization of BGCs of related myxobacteria from various genera and suggest spatial proximity of hybrid, modular clusters contributes to the metabolic adaptability of myxobacteria.
Collapse
Affiliation(s)
- Andrew Ahearne
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Kayleigh Phillips
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Thomas Knehans
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Miranda Hoing
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Scot E. Dowd
- MR DNA, Molecular Research LP, Shallowater, TX 79363, USA
| | - D. Cole Stevens
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| |
Collapse
|
26
|
Tseng CC, Chen L, Lee C, Tu Z, Lin CH, Lin HC. Characterization and catalytic investigation of fungal single-module nonribosomal peptide synthetase in terpene-amino acid meroterpenoid biosynthesis. J Ind Microbiol Biotechnol 2023; 50:kuad043. [PMID: 38049376 PMCID: PMC10720950 DOI: 10.1093/jimb/kuad043] [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: 10/18/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023]
Abstract
Hybrid natural products are compounds that originate from diverse biosynthetic pathways and undergo a conjugation process, which enables them to expand their chemical diversity and biological functionality. Terpene-amino acid meroterpenoids have garnered increasing attention in recent years, driven by the discovery of noteworthy examples such as the anthelmintic CJ-12662, the insecticidal paeciloxazine, and aculene A (1). In the biosynthesis of terpene-amino acid natural products, single-module nonribosomal peptide synthetases (NRPSs) have been identified to be involved in the esterification step, catalyzing the fusion of modified terpene and amino acid components. Despite prior investigations into these NRPSs through gene deletion or in vivo experiments, the enzymatic basis and mechanistic insights underlying this family of single-module NRPSs remain unclear. In this study, we performed biochemical characterization of AneB by in vitro characterization, molecular docking, and site-directed mutagenesis. The enzyme reaction analyses, performed with L-proline and daucane/nordaucane sesquiterpene substrates, revealed that AneB specifically esterifies the C10-OH of aculenes with L-proline. Notably, in contrast to ThmA in CJ-12662 biosynthesis, which exclusively recognizes oxygenated amorpha-4,11-diene sesquiterpenes for L-tryptophan transfer, AneB demonstrates broad substrate selectivity, including oxygenated amorpha-4,11-diene and 2-phenylethanol, resulting in the production of diverse unnatural prolyl compounds. Furthermore, site-directed mutagenesis experiments indicated the involvement of H794 and D798 in the esterification catalyzed by AneB. Lastly, domain swapping between AneB and ThmA unveiled that the A‒T domains of ThmA can be effectively harnessed by the C domain of AneB for L-tryptophan transfer, thus highlighting the potential of the C domain of AneB for generating various terpene-amino acid meroterpenoid derivatives. ONE-SENTENCE SUMMARY The enzymatic basis and mechanistic insights into AneB, a single-module NRPS, highlight its capacity to generate various terpene-amino acid meroterpenoid derivatives.
Collapse
Affiliation(s)
- Cheng-Chung Tseng
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
- School of Pharmacy, National Taiwan University, Taipei 100, Taiwan R.O.C
| | - Li‐Xun Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
- Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan R.O.C
| | - Chi‐Fang Lee
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
- Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan R.O.C
| | - Zhijay Tu
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
- Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan R.O.C
| | - Hsiao-Ching Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
- School of Pharmacy, National Taiwan University, Taipei 100, Taiwan R.O.C
- Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan R.O.C
| |
Collapse
|
27
|
Louwen JJR, Kautsar SA, van der Burg S, Medema MH, van der Hooft JJJ. iPRESTO: Automated discovery of biosynthetic sub-clusters linked to specific natural product substructures. PLoS Comput Biol 2023; 19:e1010462. [PMID: 36758069 PMCID: PMC9946207 DOI: 10.1371/journal.pcbi.1010462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/22/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Microbial specialised metabolism is full of valuable natural products that are applied clinically, agriculturally, and industrially. The genes that encode their biosynthesis are often physically clustered on the genome in biosynthetic gene clusters (BGCs). Many BGCs consist of multiple groups of co-evolving genes called sub-clusters that are responsible for the biosynthesis of a specific chemical moiety in a natural product. Sub-clusters therefore provide an important link between the structures of a natural product and its BGC, which can be leveraged for predicting natural product structures from sequence, as well as for linking chemical structures and metabolomics-derived mass features to BGCs. While some initial computational methodologies have been devised for sub-cluster detection, current approaches are not scalable, have only been run on small and outdated datasets, or produce an impractically large number of possible sub-clusters to mine through. Here, we constructed a scalable method for unsupervised sub-cluster detection, called iPRESTO, based on topic modelling and statistical analysis of co-occurrence patterns of enzyme-coding protein families. iPRESTO was used to mine sub-clusters across 150,000 prokaryotic BGCs from antiSMASH-DB. After annotating a fraction of the resulting sub-cluster families, we could predict a substructure for 16% of the antiSMASH-DB BGCs. Additionally, our method was able to confirm 83% of the experimentally characterised sub-clusters in MIBiG reference BGCs. Based on iPRESTO-detected sub-clusters, we could correctly identify the BGCs for xenorhabdin and salbostatin biosynthesis (which had not yet been annotated in BGC databases), as well as propose a candidate BGC for akashin biosynthesis. Additionally, we show for a collection of 145 actinobacteria how substructures can aid in linking BGCs to molecules by correlating iPRESTO-detected sub-clusters to MS/MS-derived Mass2Motifs substructure patterns. This work paves the way for deeper functional and structural annotation of microbial BGCs by improved linking of orphan molecules to their cognate gene clusters, thus facilitating accelerated natural product discovery.
Collapse
Affiliation(s)
- Joris J. R. Louwen
- Bioinformatics Group, Wageningen University, Wageningen, the Netherlands
| | - Satria A. Kautsar
- Bioinformatics Group, Wageningen University, Wageningen, the Netherlands
| | | | - Marnix H. Medema
- Bioinformatics Group, Wageningen University, Wageningen, the Netherlands
- * E-mail: (MHM); (JJJvdH)
| | - Justin J. J. van der Hooft
- Bioinformatics Group, Wageningen University, Wageningen, the Netherlands
- Department of Biochemistry, University of Johannesburg, Johannesburg, South Africa
- * E-mail: (MHM); (JJJvdH)
| |
Collapse
|
28
|
Devkar HU, Thakur NL, Kaur P. Marine-derived antimicrobial molecules from the sponges and their associated bacteria. Can J Microbiol 2023; 69:1-16. [PMID: 36288610 DOI: 10.1139/cjm-2022-0147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antimicrobial resistance (AMR) is one of the leading global health issues that demand urgent attention. Very soon the world will have to bear the consequences of increased drug resistance if new anti-infectives are not pumped into the clinical pipeline in a short period. This presses on the need for novel chemical entities, and the marine environment is one such hotspot to look for. The Ocean harbours a variety of organisms, of which from this aspect, "Sponges (Phylum Porifera)" are of particular interest. To tackle the stresses faced due to their sessile and filter-feeding lifestyle, sponges produce various bioactive compounds, which can be tapped for human use. The sponges harbour several microorganisms of different types and in most cases; the microbial symbionts are the actual producers of the bioactive compounds. This review describes the alarming need for the development of new antimicrobials and how marine sponges can contribute to this. Selected antimicrobial compounds from the marine sponges and their associated bacteria have been described. Additionally, measures to tackle the supply problem have been covered, which is the primary obstacle in marine natural product drug discovery.
Collapse
Affiliation(s)
- Heena U Devkar
- CSIR- National Institute of Oceanography, Dona Paula 403004, Goa, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Narsinh L Thakur
- CSIR- National Institute of Oceanography, Dona Paula 403004, Goa, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Parvinder Kaur
- Foundation for Neglected Disease Research, Bangalore 561203, Karnataka, India
| |
Collapse
|
29
|
Alam K, Islam MM, Islam S, Hao J, Abbasi MN, Hayat M, Shoaib M, Zhang Y, Li A. Comparative genomics with evolutionary lineage in Streptomyces bacteria reveals high biosynthetic potentials. World J Microbiol Biotechnol 2022; 39:64. [PMID: 36581678 DOI: 10.1007/s11274-022-03433-y] [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: 05/10/2022] [Accepted: 10/06/2022] [Indexed: 12/31/2022]
Abstract
Genome mining in silico approaches allow scientists to proficiently evaluate the genomic potency of secondary bioactive chemical producers and find new bioactive compounds in different bacteria. Streptomyces is one of the most ubiquitous bacterial genera in the environments, and well-known as prolific producers of diverse and valuable natural products (NPs) with significant biological activities. Mining and prioritizing of NP biosynthetic gene clusters (BGCs) would be the most important stage in the identification of novel compounds. Comparative genomics and genetic similarity network analysis of 62 Streptomyces public reference genomes demonstrated that individuals of these species exhibit a huge number of distinct NP BGCs, the most of which are cryptic and unconnected to any reported NPs with high phylogenetic variation among individuals. It was assumed that substantial heterogeneity across the varieties of species of Streptomyces drives outstanding biosynthetic and metabolic potential, making them plausible candidates for the identification of novel molecules.
Collapse
Affiliation(s)
- Khorshed Alam
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Md Mahmudul Islam
- Department of Microbiology, Rajshahi Institute of Biosciences (RIB), Affiliated University of Rajshahi, Rajshahi, 6212, Bangladesh
| | - Saiful Islam
- Chattogram Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram, 4220, Bangladesh
| | - Jinfang Hao
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Muhammad Nazeer Abbasi
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Muhammad Hayat
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Muhammad Shoaib
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Youming Zhang
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Aiying Li
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, 266237, People's Republic of China.
| |
Collapse
|
30
|
Scott TA, Verest M, Farnung J, Forneris CC, Robinson SL, Ji X, Hubrich F, Chepkirui C, Richter DU, Huber S, Rust P, Streiff AB, Zhang Q, Bode JW, Piel J. Widespread microbial utilization of ribosomal β-amino acid-containing peptides and proteins. Chem 2022. [DOI: 10.1016/j.chempr.2022.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
31
|
Ullah R, Zhu B, Kakar KU, Nawaz Z, Mushtaq M, Durrani TS, Islam ZU, Nawaz F. Micro-synteny conservation analysis revealed the evolutionary history of bacterial biphenyl degradation pathway. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:494-505. [PMID: 35560986 DOI: 10.1111/1758-2229.13081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 06/15/2023]
Abstract
Phenolic compounds have been enlisted by the United States Environmental Protection Agency (USEPA) and the European Union (EU) as pollutants of priority concern. The biphenyl degradation pathway plays an essential role in prokaryote polychlorinated biphenyls degradation. Our understanding of prokaryotic pathways and their evolution has dramatically increased in recent years with the advancements in prokaryotic genome sequencing and analysis tools. In this work, we applied bioinformatics tools to study the evolution of the biphenyl degradation pathway focusing on the phylogeny and initiation of four representative species (Burkholderia xenovorans LB400, Polaromonas naphthalenivorans CJ2, Pseudomonas putida F1 and Rhodococcus jostii RHA1). These species contained partial or full concatenated genes from bph gene cluster (i.e. bphRbphA1A2A3A4BCKHJID). The aim was to establish this pathway's origin and development mode in the prokaryotic world. Genomic screening revealed that many bacterial species possess genes for the biphenyl degradation pathway. However, the micro-synteny conservation analysis indicated that massive gene recruitment events might have occurred during the evolution of the biphenyl degradation pathway. Combining with the phylogenetic positions, this work points to the evolutionary process of acquiring the biphenyl degradation pathway by different fragments through horizontal gene transfer in these bacterial groups. This study reports the first-ever evidence of the birth of this pathway in the represented species.
Collapse
Affiliation(s)
- Raqeeb Ullah
- Department of Environmental Science, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, 87300, Pakistan
| | - Bo Zhu
- Key Laboratory of Urban Agriculture by Ministry of Agriculture of China, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Kaleem U Kakar
- Department of Microbiology, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, 87300, Pakistan
| | - Zarqa Nawaz
- Department of Botany, University of Central Punjab, Rawalpindi, Pakistan
| | - Muhammd Mushtaq
- Department of Biotechnology, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, 87300, Pakistan
| | - Taimoor Shah Durrani
- Department of Environmental Science, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, 87300, Pakistan
| | - Zia Ul Islam
- Department of Civil and Environmental Engineering, The University of Toledo, Toledo, OH, USA
| | - Faheem Nawaz
- Department of Environmental Science, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, 87300, Pakistan
| |
Collapse
|
32
|
Entfellner E, Li R, Jiang Y, Ru J, Blom J, Deng L, Kurmayer R. Toxic/Bioactive Peptide Synthesis Genes Rearranged by Insertion Sequence Elements Among the Bloom-Forming Cyanobacteria Planktothrix. Front Microbiol 2022; 13:901762. [PMID: 35966708 PMCID: PMC9366434 DOI: 10.3389/fmicb.2022.901762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022] Open
Abstract
It has been generally hypothesized that mobile elements can induce genomic rearrangements and influence the distribution and functionality of toxic/bioactive peptide synthesis pathways in microbes. In this study, we performed in depth genomic analysis by completing the genomes of 13 phylogenetically diverse strains of the bloom-forming freshwater cyanobacteria Planktothrix spp. to investigate the role of insertion sequence (IS) elements in seven pathways. Chromosome size varied from 4.7-4.8 Mbp (phylogenetic Lineage 1 of P. agardhii/P. rubescens thriving in shallow waterbodies) to 5.4-5.6 Mbp (Lineage 2 of P. agardhii/P. rubescens thriving in deeper physically stratified lakes and reservoirs) and 6.3-6.6 Mbp (Lineage 3, P. pseudagardhii/P. tepida including planktic and benthic ecotypes). Although the variation in chromosome size was positively related to the proportion of IS elements (1.1-3.7% on chromosome), quantitatively, IS elements and other paralogs only had a minor share in chromosome size variation. Thus, the major part of genomic variation must have resulted from gene loss processes (ancestor of Lineages 1 and 2) and horizontal gene transfer (HGT). Six of seven peptide synthesis gene clusters were found located on the chromosome and occurred already in the ancestor of P. agardhii/P. rubescens, and became partly lost during evolution of Lineage 1. In general, no increased IS element frequency in the vicinity of peptide synthesis gene clusters was observed. We found a higher proportion of IS elements in ten breaking regions related to chromosomal rearrangements and a tendency for colocalization of toxic/bioactive peptide synthesis gene clusters on the chromosome.
Collapse
Affiliation(s)
| | - Ruibao Li
- Research Department for Limnology, University of Innsbruck, Mondsee, Austria
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- Department of Ecology and Institute of Hydrobiology, Jinan University, Guangzhou, China
| | - Yiming Jiang
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Jinlong Ru
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Jochen Blom
- Bioinformatics and Systems Biology, Justus-Liebig-University, Giessen, Germany
| | - Li Deng
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Rainer Kurmayer
- Research Department for Limnology, University of Innsbruck, Mondsee, Austria
| |
Collapse
|
33
|
Henriksen NNSE, Lindqvist LL, Wibowo M, Sonnenschein EC, Bentzon-Tilia M, Gram L. Role is in the eye of the beholder-the multiple functions of the antibacterial compound tropodithietic acid produced by marine Rhodobacteraceae. FEMS Microbiol Rev 2022; 46:fuac007. [PMID: 35099011 PMCID: PMC9075582 DOI: 10.1093/femsre/fuac007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Many microbial secondary metabolites have been studied for decades primarily because of their antimicrobial properties. However, several of these metabolites also possess nonantimicrobial functions, both influencing the physiology of the producer and their ecological neighbors. An example of a versatile bacterial secondary metabolite with multiple functions is the tropone derivative tropodithietic acid (TDA). TDA is a broad-spectrum antimicrobial compound produced by several members of the Rhodobacteraceae family, a major marine bacterial lineage, within the genera Phaeobacter, Tritonibacter, and Pseudovibrio. The production of TDA is governed by the mode of growth and influenced by the availability of nutrient sources. The antibacterial effect of TDA is caused by disruption of the proton motive force of target microorganisms and, potentially, by its iron-chelating properties. TDA also acts as a signaling molecule, affecting gene expression in other bacteria, and altering phenotypic traits such as motility, biofilm formation, and antibiotic production in the producer. In microbial communities, TDA-producing bacteria cause a reduction of the relative abundance of closely related species and some fast-growing heterotrophic bacteria. Here, we summarize the current understanding of the chemical ecology of TDA, including the environmental niches of TDA-producing bacteria, and the molecular mechanisms governing the function and regulation of TDA.
Collapse
Affiliation(s)
- Nathalie N S E Henriksen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts, Plads Bldg. 221, DK-2800 Kgs. Lyngby, Denmark
| | - Laura L Lindqvist
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts, Plads Bldg. 221, DK-2800 Kgs. Lyngby, Denmark
| | - Mario Wibowo
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts, Plads Bldg. 221, DK-2800 Kgs. Lyngby, Denmark
| | - Eva C Sonnenschein
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts, Plads Bldg. 221, DK-2800 Kgs. Lyngby, Denmark
| | - Mikkel Bentzon-Tilia
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts, Plads Bldg. 221, DK-2800 Kgs. Lyngby, Denmark
| | - Lone Gram
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts, Plads Bldg. 221, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
34
|
Wenski SL, Thiengmag S, Helfrich EJ. Complex peptide natural products: Biosynthetic principles, challenges and opportunities for pathway engineering. Synth Syst Biotechnol 2022; 7:631-647. [PMID: 35224231 PMCID: PMC8842026 DOI: 10.1016/j.synbio.2022.01.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/03/2023] Open
Abstract
Complex peptide natural products exhibit diverse biological functions and a wide range of physico-chemical properties. As a result, many peptides have entered the clinics for various applications. Two main routes for the biosynthesis of complex peptides have evolved in nature: ribosomally synthesized and post-translationally modified peptide (RiPP) biosynthetic pathways and non-ribosomal peptide synthetases (NRPSs). Insights into both bioorthogonal peptide biosynthetic strategies led to the establishment of universal principles for each of the two routes. These universal rules can be leveraged for the targeted identification of novel peptide biosynthetic blueprints in genome sequences and used for the rational engineering of biosynthetic pathways to produce non-natural peptides. In this review, we contrast the key principles of both biosynthetic routes and compare the different biochemical strategies to install the most frequently encountered peptide modifications. In addition, the influence of the fundamentally different biosynthetic principles on past, current and future engineering approaches is illustrated. Despite the different biosynthetic principles of both peptide biosynthetic routes, the arsenal of characterized peptide modifications encountered in RiPP and NRPS systems is largely overlapping. The continuous expansion of the biocatalytic toolbox of peptide modifying enzymes for both routes paves the way towards the production of complex tailor-made peptides and opens up the possibility to produce NRPS-derived peptides using the ribosomal route and vice versa.
Collapse
Affiliation(s)
- Sebastian L. Wenski
- Institute for Molecular Bio Science, Goethe University Frankfurt, 60438, Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), 60325, Frankfurt am Main, Germany
| | - Sirinthra Thiengmag
- Institute for Molecular Bio Science, Goethe University Frankfurt, 60438, Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), 60325, Frankfurt am Main, Germany
| | - Eric J.N. Helfrich
- Institute for Molecular Bio Science, Goethe University Frankfurt, 60438, Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), 60325, Frankfurt am Main, Germany
| |
Collapse
|
35
|
Claus S, Jezierska S, Elbourne LDH, Van Bogaert I. Exploring the transportome of the biosurfactant producing yeast Starmerella bombicola. BMC Genomics 2022; 23:22. [PMID: 34998388 PMCID: PMC8742932 DOI: 10.1186/s12864-021-08177-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Starmerella bombicola is a non-conventional yeast mainly known for its capacity to produce high amounts of the glycolipids 'sophorolipids'. Although its product has been used as biological detergent for a couple of decades, the genetics of S. bombicola are still largely unknown. Computational analysis of the yeast's genome enabled us to identify 254 putative transporter genes that make up the entire transportome. For each of them, a potential substrate was predicted using homology analysis, subcellular localization prediction and RNA sequencing in different stages of growth. One transporter family is of exceptional importance to this yeast: the ATP Binding Cassette (ABC) transporter Superfamily, because it harbors the main driver behind the highly efficient sophorolipid export. Furthermore, members of this superfamily translocate a variety of compounds ranging from antibiotics to hydrophobic molecules. We conducted an analysis of this family by creating deletion mutants to understand their role in the export of hydrophobic compounds, antibiotics and sophorolipids. Doing this, we could experimentally confirm the transporters participating in the efflux of medium chain fatty alcohols, particularly decanol and undecanol, and identify a second sophorolipid transporter that is located outside the sophorolipid biosynthetic gene cluster.
Collapse
Affiliation(s)
- Silke Claus
- Centre for Synthetic Biology, Department of Biotechnology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Sylwia Jezierska
- Centre for Synthetic Biology, Department of Biotechnology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Liam D H Elbourne
- Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW, 2109, Australia
| | - Inge Van Bogaert
- Centre for Synthetic Biology, Department of Biotechnology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
| |
Collapse
|
36
|
Vertical Inheritance Facilitates Interspecies Diversification in Biosynthetic Gene Clusters and Specialized Metabolites. mBio 2021; 12:e0270021. [PMID: 34809466 PMCID: PMC8609351 DOI: 10.1128/mbio.02700-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
While specialized metabolites are thought to mediate ecological interactions, the evolutionary processes driving chemical diversification, particularly among closely related lineages, remain poorly understood. Here, we examine the evolutionary dynamics governing the distribution of natural product biosynthetic gene clusters (BGCs) among 118 strains representing all nine currently named species of the marine actinobacterial genus Salinispora. While much attention has been given to the role of horizontal gene transfer (HGT) in structuring BGC distributions, we find that vertical descent facilitates interspecies BGC diversification over evolutionary timescales. Moreover, we identified a distinct phylogenetic signal among Salinispora species at both the BGC and metabolite level, indicating that specialized metabolism represents a conserved phylogenetic trait. Using a combination of genomic analyses and liquid chromatography–high-resolution tandem mass spectrometry (LC-MS/MS) targeting nine experimentally characterized BGCs and their small molecule products, we identified gene gain/loss events, constrained interspecies recombination, and other evolutionary processes associated with vertical inheritance as major contributors to BGC diversification. These evolutionary dynamics had direct consequences for the compounds produced, as exemplified by species-level differences in salinosporamide production. Together, our results support the concept that specialized metabolites, and their cognate BGCs, can represent phylogenetically conserved functional traits with chemical diversification proceeding in species-specific patterns over evolutionary time frames.
Collapse
|
37
|
Genome Mining of Pseudomonas Species: Diversity and Evolution of Metabolic and Biosynthetic Potential. Molecules 2021; 26:molecules26247524. [PMID: 34946606 PMCID: PMC8704066 DOI: 10.3390/molecules26247524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022] Open
Abstract
Microbial genome sequencing has uncovered a myriad of natural products (NPs) that have yet to be explored. Bacteria in the genus Pseudomonas serve as pathogens, plant growth promoters, and therapeutically, industrially, and environmentally important microorganisms. Though most species of Pseudomonas have a large number of NP biosynthetic gene clusters (BGCs) in their genomes, it is difficult to link many of these BGCs with products under current laboratory conditions. In order to gain new insights into the diversity, distribution, and evolution of these BGCs in Pseudomonas for the discovery of unexplored NPs, we applied several bioinformatic programming approaches to characterize BGCs from Pseudomonas reference genome sequences available in public databases along with phylogenetic and genomic comparison. Our research revealed that most BGCs in the genomes of Pseudomonas species have a high diversity for NPs at the species and subspecies levels and built the correlation of species with BGC taxonomic ranges. These data will pave the way for the algorithmic detection of species- and subspecies-specific pathways for NP development.
Collapse
|
38
|
Chakraborty P. Gene cluster from plant to microbes: Their role in genome architecture, organism's development, specialized metabolism and drug discovery. Biochimie 2021; 193:1-15. [PMID: 34890733 DOI: 10.1016/j.biochi.2021.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023]
Abstract
Plants and microbes fulfil our daily requirements through different high-value chemicals, e.g., nutraceuticals, pharmaceuticals, cosmetics, and through varieties of fruits, crops, vegetables, and many more. Utmost care would therefore be taken for growth, development and sustainability of these important crops and medicinal plants and microbes. Homeobox genes and HOX clusters and their recently characterized expanded family members, including newly discovered homeobox, WOX gene from medicinal herb, Panax ginseng, significantly contributes in the growth and development of these organisms. On the other hand, secondary metabolites produced through secondary metabolism of plants and microbes are used as organisms defense as well as drugs/drug-like molecules for humans. Both the developmental HOX cluster and the biosynthetic gene-cluster (BGC) for secondary metabolites are organised in organisms genome. Genome mining and genomewide analysis of these clusters will definitely identify and characterize many more important molecules from unexplored plants and microbes and underexplored human microbiota and the evolution studies of these clusters will indicate their source of origin. Although genomics revolution now continues at a pace, till date only few hundred plant genome sequences are available. However, next-generation sequencing (NGS) technology now in market and may be applied even for plants with recalcitrant genomes, eventually may discover genomic potential towards production of secondary metabolites of diverse plants and micro-organisms present in the environment and microbiota. Additionally, the development of tools for genome mining e.g., antiSMASH, plantiSMASH, and more and more computational approaches that predicts hundreds of secondary metabolite BGCs will be discussed.
Collapse
Affiliation(s)
- Prasanta Chakraborty
- Kalpana Chawla Center for Space and Nanoscience, Kolkata, Indian Institute of Chemical Biology (retd.), Kolkata, 700032, India.
| |
Collapse
|
39
|
Transporter Gene-mediated Typing for Detection and Genome Mining of Lipopeptide-producing Pseudomonas. Appl Environ Microbiol 2021; 88:e0186921. [PMID: 34731056 PMCID: PMC8788793 DOI: 10.1128/aem.01869-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Pseudomonas lipopeptides (LPs) are involved in diverse ecological functions and have biotechnological application potential associated with their antimicrobial and/or antiproliferative activities. They are synthesized by multimodular nonribosomal peptide synthetases which, together with transport and regulatory proteins, are encoded by large biosynthetic gene clusters (BGCs). These secondary metabolites are classified in distinct families based on the sequence and length of the oligopeptide and size of the macrocycle, if present. The phylogeny of PleB, the MacB-like transporter that is part of a dedicated ATP-dependent tripartite efflux system driving export of Pseudomonas LPs, revealed a strong correlation with LP chemical diversity. As each LP BGC carries its cognate pleB, PleB is suitable as a diagnostic sequence for genome mining, allowing assignment of the putative metabolite to a particular LP family. In addition, pleB proved to be a suitable target gene for an alternative PCR method for detecting LP-producing Pseudomonas sp. and did not rely on amplification of catalytic domains of the biosynthetic enzymes. Combined with amplicon sequencing, this approach enabled typing of Pseudomonas strains as potential producers of a LP belonging to one of the known LP families, underscoring its value for strain prioritization. This finding was validated by chemical characterization of known LPs from three different families secreted by novel producers isolated from the rice or maize rhizosphere, namely, the type strains of Pseudomonas fulva (putisolvin), Pseudomonas zeae (tensin), and Pseudomonas xantholysinigenes (xantholysin). In addition, a new member of the Bananamide family, prosekin, was discovered in the type strain of Pseudomonas prosekii, which is an Antarctic isolate. IMPORTANCEPseudomonas spp. are ubiquitous bacteria able to thrive in a wide range of ecological niches, and lipopeptides often support their lifestyle but also their interaction with other micro- and macro-organisms. Therefore, the production of lipopeptides is widespread among Pseudomonas strains. Consequently, Pseudomonas lipopeptide research not only affects chemists and microbiologists but also touches a much broader audience, including biochemists, ecologists, and plant biologists. In this study, we present a reliable transporter gene-guided approach for the detection and/or typing of Pseudomonas lipopeptide producers. Indeed, it allows us to readily assess the lipopeptide diversity among sets of Pseudomonas isolates and differentiate strains likely to produce known lipopeptides from producers of potentially novel lipopeptides. This work provides a valuable tool that can also be integrated in a genome mining strategy and adapted for the typing of other specialized metabolites.
Collapse
|
40
|
Bojarska J, Mieczkowski A, Ziora ZM, Skwarczynski M, Toth I, Shalash AO, Parang K, El-Mowafi SA, Mohammed EHM, Elnagdy S, AlKhazindar M, Wolf WM. Cyclic Dipeptides: The Biological and Structural Landscape with Special Focus on the Anti-Cancer Proline-Based Scaffold. Biomolecules 2021; 11:1515. [PMID: 34680148 PMCID: PMC8533947 DOI: 10.3390/biom11101515] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cyclic dipeptides, also know as diketopiperazines (DKP), the simplest cyclic forms of peptides widespread in nature, are unsurpassed in their structural and bio-functional diversity. DKPs, especially those containing proline, due to their unique features such as, inter alia, extra-rigid conformation, high resistance to enzyme degradation, increased cell permeability, and expandable ability to bind a diverse of targets with better affinity, have emerged in the last years as biologically pre-validated platforms for the drug discovery. Recent advances have revealed their enormous potential in the development of next-generation theranostics, smart delivery systems, and biomaterials. Here, we present an updated review on the biological and structural profile of these appealing biomolecules, with a particular emphasis on those with anticancer properties, since cancers are the main cause of death all over the world. Additionally, we provide a consideration on supramolecular structuring and synthons, based on the proline-based DKP privileged scaffold, for inspiration in the design of compound libraries in search of ideal ligands, innovative self-assembled nanomaterials, and bio-functional architectures.
Collapse
Affiliation(s)
- Joanna Bojarska
- Faculty of Chemistry, Institute of General & Inorganic Chemistry, Technical University of Lodz, 90-924 Lodz, Poland;
| | - Adam Mieczkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland;
| | - Zyta M. Ziora
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (Z.M.Z.); (I.T.)
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.S.); (A.O.S.)
| | - Istvan Toth
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (Z.M.Z.); (I.T.)
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.S.); (A.O.S.)
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Ahmed O. Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.S.); (A.O.S.)
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, School of Pharmacy, Chapman University, Irvine, CA 92618, USA; (K.P.); (S.A.E.-M.); (E.H.M.M.)
| | - Shaima A. El-Mowafi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, School of Pharmacy, Chapman University, Irvine, CA 92618, USA; (K.P.); (S.A.E.-M.); (E.H.M.M.)
| | - Eman H. M. Mohammed
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, School of Pharmacy, Chapman University, Irvine, CA 92618, USA; (K.P.); (S.A.E.-M.); (E.H.M.M.)
| | - Sherif Elnagdy
- Botany Department, Faculty of Science, Cairo University, Giza 12613, Egypt; (S.E.); (M.A.)
| | - Maha AlKhazindar
- Botany Department, Faculty of Science, Cairo University, Giza 12613, Egypt; (S.E.); (M.A.)
| | - Wojciech M. Wolf
- Faculty of Chemistry, Institute of General & Inorganic Chemistry, Technical University of Lodz, 90-924 Lodz, Poland;
| |
Collapse
|
41
|
Vignolle GA, Schaffer D, Zehetner L, Mach RL, Mach-Aigner AR, Derntl C. FunOrder: A robust and semi-automated method for the identification of essential biosynthetic genes through computational molecular co-evolution. PLoS Comput Biol 2021; 17:e1009372. [PMID: 34570757 PMCID: PMC8476034 DOI: 10.1371/journal.pcbi.1009372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/23/2021] [Indexed: 11/24/2022] Open
Abstract
Secondary metabolites (SMs) are a vast group of compounds with different structures and properties that have been utilized as drugs, food additives, dyes, and as monomers for novel plastics. In many cases, the biosynthesis of SMs is catalysed by enzymes whose corresponding genes are co-localized in the genome in biosynthetic gene clusters (BGCs). Notably, BGCs may contain so-called gap genes, that are not involved in the biosynthesis of the SM. Current genome mining tools can identify BGCs, but they have problems with distinguishing essential genes from gap genes. This can and must be done by expensive, laborious, and time-consuming comparative genomic approaches or transcriptome analyses. In this study, we developed a method that allows semi-automated identification of essential genes in a BGC based on co-evolution analysis. To this end, the protein sequences of a BGC are blasted against a suitable proteome database. For each protein, a phylogenetic tree is created. The trees are compared by treeKO to detect co-evolution. The results of this comparison are visualized in different output formats, which are compared visually. Our results suggest that co-evolution is commonly occurring within BGCs, albeit not all, and that especially those genes that encode for enzymes of the biosynthetic pathway are co-evolutionary linked and can be identified with FunOrder. In light of the growing number of genomic data available, this will contribute to the studies of BGCs in native hosts and facilitate heterologous expression in other organisms with the aim of the discovery of novel SMs. The discovery and description of novel fungal secondary metabolites promises novel antibiotics, pharmaceuticals, and other useful compounds. A way to identify novel secondary metabolites is to express the corresponding genes in a suitable expression host. Consequently, a detailed knowledge or an accurate prediction of these genes is necessary. In fungi, the genes are co-localized in so-called biosynthetic gene clusters. Notably, the clusters may also contain genes that are not necessary for the biosynthesis of the secondary metabolites, so-called gap genes. We developed a method to detect co-evolved genes within the clusters and demonstrated that essential genes are co-evolving and can thus be differentiated from the gap genes. This adds an additional layer of information, which can support researchers with their decisions on which genes to study and express for the discovery of novel secondary metabolites.
Collapse
Affiliation(s)
- Gabriel A. Vignolle
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | - Denise Schaffer
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | - Leopold Zehetner
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | - Robert L. Mach
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | - Astrid R. Mach-Aigner
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | - Christian Derntl
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
- * E-mail:
| |
Collapse
|
42
|
Biology and applications of co-produced, synergistic antimicrobials from environmental bacteria. Nat Microbiol 2021; 6:1118-1128. [PMID: 34446927 DOI: 10.1038/s41564-021-00952-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 07/21/2021] [Indexed: 02/07/2023]
Abstract
Environmental bacteria, such as Streptomyces spp., produce specialized metabolites that are potent antibiotics and therapeutics. Selected specialized antimicrobials are co-produced and function together synergistically. Co-produced antimicrobials comprise multiple chemical classes and are produced by a wide variety of bacteria in different environmental niches, suggesting that their combined functions are ecologically important. Here, we highlight the exquisite mechanisms that underlie the simultaneous production and functional synergy of 16 sets of co-produced antimicrobials. To date, antibiotic and antifungal discovery has focused mainly on single molecules, but we propose that methods to target co-produced antimicrobials could widen the scope and applications of discovery programs.
Collapse
|
43
|
Baunach M, Chowdhury S, Stallforth P, Dittmann E. The Landscape of Recombination Events That Create Nonribosomal Peptide Diversity. Mol Biol Evol 2021; 38:2116-2130. [PMID: 33480992 PMCID: PMC8097286 DOI: 10.1093/molbev/msab015] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nonribosomal peptides (NRP) are crucial molecular mediators in microbial ecology and provide indispensable drugs. Nevertheless, the evolution of the flexible biosynthetic machineries that correlates with the stunning structural diversity of NRPs is poorly understood. Here, we show that recombination is a key driver in the evolution of bacterial NRP synthetase (NRPS) genes across distant bacterial phyla, which has guided structural diversification in a plethora of NRP families by extensive mixing and matching of biosynthesis genes. The systematic dissection of a large number of individual recombination events did not only unveil a striking plurality in the nature and origin of the exchange units but allowed the deduction of overarching principles that enable the efficient exchange of adenylation (A) domain substrates while keeping the functionality of the dynamic multienzyme complexes. In the majority of cases, recombination events have targeted variable portions of the Acore domains, yet domain interfaces and the flexible Asub domain remained untapped. Our results strongly contradict the widespread assumption that adenylation and condensation (C) domains coevolve and significantly challenge the attributed role of C domains as stringent selectivity filter during NRP synthesis. Moreover, they teach valuable lessons on the choice of natural exchange units in the evolution of NRPS diversity, which may guide future engineering approaches.
Collapse
Affiliation(s)
- Martin Baunach
- Institute for Biochemistry and Biology, University of Potsdam, Potsdam-Golm, Germany
| | - Somak Chowdhury
- Department of Paleobiotechnology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
| | - Pierre Stallforth
- Department of Paleobiotechnology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
| | - Elke Dittmann
- Institute for Biochemistry and Biology, University of Potsdam, Potsdam-Golm, Germany
| |
Collapse
|
44
|
Davis EW, Okrent RA, Manning VA, Trippe KM. Unexpected distribution of the 4-formylaminooxyvinylglycine (FVG) biosynthetic pathway in Pseudomonas and beyond. PLoS One 2021; 16:e0247348. [PMID: 33891610 PMCID: PMC8064604 DOI: 10.1371/journal.pone.0247348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/13/2021] [Indexed: 11/29/2022] Open
Abstract
The biological herbicide and antibiotic 4-formylaminooxyvinylglycine (FVG) was originally isolated from several rhizosphere-associated strains of Pseudomonas fluorescens. Biosynthesis of FVG is dependent on the gvg biosynthetic gene cluster in P. fluorescens. In this investigation, we used comparative genomics to identify strains with the genetic potential to produce FVG due to presence of a gvg gene cluster. These strains primarily belong to two groups of Pseudomonas, P. fluorescens and P. syringae, however, a few strains with the gvg cluster were found outside of Pseudomonas. Mass spectrometry confirmed that all tested strains of the P. fluorescens species group produced FVG. However, P. syringae strains did not produce FVG under standard conditions. Several lines of evidence regarding the transmission of the gvg cluster including a robust phylogenetic analysis suggest that it was introduced multiple times through horizontal gene transfer within the Pseudomonas lineage as well as in select lineages of Thiomonas, Burkholderia and Pantoea. Together, these data broaden our understanding of the evolution and diversity of FVG biosynthesis. In the course of this investigation, additional gene clusters containing only a subset of the genes required to produce FVG were identified in a broad range of bacteria, including many non-pseudomonads.
Collapse
Affiliation(s)
- Edward W. Davis
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, United States of America
| | - Rachel A. Okrent
- Forage Seed and Cereal Research Unit, United States Department of Agriculture, Agricultural Research Service, Corvallis, OR, United States of America
| | - Viola A. Manning
- Forage Seed and Cereal Research Unit, United States Department of Agriculture, Agricultural Research Service, Corvallis, OR, United States of America
| | - Kristin M. Trippe
- Forage Seed and Cereal Research Unit, United States Department of Agriculture, Agricultural Research Service, Corvallis, OR, United States of America
- Department of Crop and Soil Sciences, Oregon State University, Corvallis, OR, United States of America
| |
Collapse
|
45
|
|
46
|
Cleto S, Haslinger K, Prather KLJ, Lu TK. Natural combinatorial genetics and prolific polyamine production enable siderophore diversification in Serratia plymuthica. BMC Biol 2021; 19:46. [PMID: 33722216 PMCID: PMC7962358 DOI: 10.1186/s12915-021-00971-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/31/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Iron is essential for bacterial survival. Bacterial siderophores are small molecules with unmatched capacity to scavenge iron from proteins and the extracellular milieu, where it mostly occurs as insoluble Fe3+. Siderophores chelate Fe3+ for uptake into the cell, where it is reduced to soluble Fe2+. Siderophores are key molecules in low soluble iron conditions. The ability of bacteria to synthesize proprietary siderophores may have increased bacterial evolutionary fitness; one way that bacteria diversify siderophore structure is by incorporating different polyamine backbones while maintaining the catechol moieties. RESULTS We report that Serratia plymuthica V4 produces a variety of siderophores, which we term the siderome, and which are assembled by the concerted action of enzymes encoded in two independent gene clusters. Besides assembling serratiochelin A and B with diaminopropane, S. plymuthica utilizes putrescine and the same set of enzymes to assemble photobactin, a siderophore found in the bacterium Photorhabdus luminescens. The enzymes encoded by one of the gene clusters can independently assemble enterobactin. A third, independent operon is responsible for biosynthesis of the hydroxamate siderophore aerobactin, initially described in Enterobacter aerogenes. Mutant strains not synthesizing polyamine-siderophores significantly increased enterobactin production levels, though lack of enterobactin did not impact the production of serratiochelins. Knocking out SchF0, an enzyme involved in the assembly of enterobactin alone, significantly reduced bacterial fitness. CONCLUSIONS This study shows the natural occurrence of serratiochelins, photobactin, enterobactin, and aerobactin in a single bacterial species and illuminates the interplay between siderophore biosynthetic pathways and polyamine production, indicating routes of molecular diversification. Given its natural yields of diaminopropane (97.75 μmol/g DW) and putrescine (30.83 μmol/g DW), S. plymuthica can be exploited for the industrial production of these compounds.
Collapse
Affiliation(s)
- Sara Cleto
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kristina Haslinger
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical and Pharmaceutical Biology, University of Groningen, Groningen, The Netherlands
| | - Kristala L J Prather
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Timothy K Lu
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
47
|
Hewage RT, Huang RJ, Lai SJ, Lien YC, Weng SH, Li D, Chen YJ, Wu SH, Chein RJ, Lin HC. An Enzyme-Mediated Aza-Michael Addition Is Involved in the Biosynthesis of an Imidazoyl Hybrid Product of Conidiogenone B. Org Lett 2021; 23:1904-1909. [PMID: 33570417 DOI: 10.1021/acs.orglett.1c00330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Meleagrin B is a terpene-alkaloid hybrid natural product that contains both the conidiogenone and meleagrin scaffold. Their derivatives show diverse biological activities. We characterized the biosynthesis of (-)-conidiogenone B (1), which involves a diterpene synthase and a P450 monooxygenase. In addition, an α,β-hydrolase (Con-ABH) was shown to catalyze an aza-Michael addition between 1 and imidazole to give 3S-imidazolyl conidiogenone B (6). Compound 6 was more potent than 1 against Staphylococcus aureus strains.
Collapse
Affiliation(s)
- Ranuka T Hewage
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C.,Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan R.O.C.,Department of Chemistry, National Taiwan University, Taipei 106, Taiwan R.O.C
| | - Rou-Jie Huang
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan R.O.C
| | - Shu-Jung Lai
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan R.O.C.,Research Center for Cancer Biology, China Medical University, Taichung 404, Taiwan R.O.C
| | - Ya-Chu Lien
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
| | - Shao-Hsing Weng
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P.R. China
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
| | - Shih-Hsiung Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C.,Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan R.O.C.,Department of Chemistry, National Taiwan University, Taipei 106, Taiwan R.O.C
| | - Rong-Jie Chein
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan R.O.C.,Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C
| | - Hsiao-Ching Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan R.O.C.,Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan R.O.C
| |
Collapse
|
48
|
Waglechner N, Culp EJ, Wright GD. Ancient Antibiotics, Ancient Resistance. EcoSal Plus 2021; 9:eESP-0027-2020. [PMID: 33734062 PMCID: PMC11163840 DOI: 10.1128/ecosalplus.esp-0027-2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
As the spread of antibiotic resistance threatens our ability to treat infections, avoiding the return of a preantibiotic era requires the discovery of new drugs. While therapeutic use of antibiotics followed by the inevitable selection of resistance is a modern phenomenon, these molecules and the genetic determinants of resistance were in use by environmental microbes long before humans discovered them. In this review, we discuss evidence that antibiotics and resistance were present in the environment before anthropogenic use, describing techniques including direct sampling of ancient DNA and phylogenetic analyses that are used to reconstruct the past. We also pay special attention to the ecological and evolutionary forces that have shaped the natural history of antibiotic biosynthesis, including a discussion of competitive versus signaling roles for antibiotics, proto-resistance, and substrate promiscuity of biosynthetic and resistance enzymes. Finally, by applying an evolutionary lens, we describe concepts governing the origins and evolution of biosynthetic gene clusters and cluster-associated resistance determinants. These insights into microbes' use of antibiotics in nature, a game they have been playing for millennia, can provide inspiration for discovery technologies and management strategies to combat the growing resistance crisis.
Collapse
Affiliation(s)
- Nicholas Waglechner
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | - Elizabeth J. Culp
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | - Gerard D. Wright
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| |
Collapse
|
49
|
Vij R, Hube B, Brunke S. Uncharted territories in the discovery of antifungal and antivirulence natural products from bacteria. Comput Struct Biotechnol J 2021; 19:1244-1252. [PMID: 33680363 PMCID: PMC7905183 DOI: 10.1016/j.csbj.2021.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/26/2022] Open
Abstract
Many fungi can cause deadly diseases in humans, and nearly every human will suffer from some kind of fungal infection in their lives. Only few antifungals are available, and some of these fail to treat intrinsically resistant species and the ever-increasing number of fungal strains that have acquired resistance. In nature, bacteria and fungi display versatile interactions that range from friendly co-existence to predation. The first antifungal drugs, nystatin and amphotericin B, were discovered in bacteria as mediators of such interactions, and bacteria continue to be an important source of antifungals. To learn more about the ecological bacterial-fungal interactions that drive the evolution of natural products and exploit them, we need to identify environments where such interactions are pronounced, and diverse. Here, we systematically analyze historic and recent developments in this field to identify potentially under-investigated niches and resources. We also discuss alternative strategies to treat fungal infections by utilizing the antagonistic potential of bacteria to target fungal stress pathways and virulence factors, and thereby suppress the evolution of antifungal resistance.
Collapse
Affiliation(s)
- Raghav Vij
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI), Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI), Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI), Germany
| |
Collapse
|
50
|
Crits-Christoph A, Bhattacharya N, Olm MR, Song YS, Banfield JF. Transporter genes in biosynthetic gene clusters predict metabolite characteristics and siderophore activity. Genome Res 2021; 31:239-250. [PMID: 33361114 PMCID: PMC7849407 DOI: 10.1101/gr.268169.120] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022]
Abstract
Biosynthetic gene clusters (BGCs) are operonic sets of microbial genes that synthesize specialized metabolites with diverse functions, including siderophores and antibiotics, which often require export to the extracellular environment. For this reason, genes for transport across cellular membranes are essential for the production of specialized metabolites and are often genomically colocalized with BGCs. Here, we conducted a comprehensive computational analysis of transporters associated with characterized BGCs. In addition to known exporters, in BGCs we found many importer-specific transmembrane domains that co-occur with substrate binding proteins possibly for uptake of siderophores or metabolic precursors. Machine learning models using transporter gene frequencies were predictive of known siderophore activity, molecular weights, and a measure of lipophilicity (log P) for corresponding BGC-synthesized metabolites. Transporter genes associated with BGCs were often equally or more predictive of metabolite features than biosynthetic genes. Given the importance of siderophores as pathogenicity factors, we used transporters specific for siderophore BGCs to identify both known and uncharacterized siderophore-like BGCs in genomes from metagenomes from the infant and adult gut microbiome. We find that 23% of microbial genomes from premature infant guts have siderophore-like BGCs, but only 3% of those assembled from adult gut microbiomes do. Although siderophore-like BGCs from the infant gut are predominantly associated with Enterobacteriaceae and Staphylococcus, siderophore-like BGCs can be identified from taxa in the adult gut microbiome that have rarely been recognized for siderophore production. Taken together, these results show that consideration of BGC-associated transporter genes can inform predictions of specialized metabolite structure and function.
Collapse
Affiliation(s)
- Alexander Crits-Christoph
- Department of Plant and Microbial Biology, University of California, Berkeley, California 94720, USA
- Innovative Genomics Institute, Berkeley, California 94720, USA
| | - Nicholas Bhattacharya
- Department of Mathematics, University of California, Berkeley, California 94720, USA
| | - Matthew R Olm
- Department of Microbiology and Immunology, Stanford University, California 94305, USA
| | - Yun S Song
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
- Department of Statistics, University of California, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Jillian F Banfield
- Innovative Genomics Institute, Berkeley, California 94720, USA
- Department of Microbiology and Immunology, Stanford University, California 94305, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
- Department of Environmental Science, Policy, and Management, University of California, Berkeley, California 94720, USA
- Earth Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| |
Collapse
|