1
|
Zhao Y, Duan YT, Zang J, Raadam MH, Pateraki I, Miettinen K, Staerk D, Kampranis SC. Structure-Agnostic Bioactivity-Driven Combinatorial Biosynthesis Reveals New Antidiabetic and Anticancer Triterpenoids. Angew Chem Int Ed Engl 2025; 64:e202416218. [PMID: 39297433 DOI: 10.1002/anie.202416218] [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: 08/23/2024] [Indexed: 11/01/2024]
Abstract
Although combinatorial biosynthesis can dramatically expand the chemical structures of bioactive natural products to identify molecules with improved characteristics, progress in this direction has been hampered by the difficulty in isolating and characterizing the numerous produced compounds. This challenge could be overcome with improved designs that enable the analysis of the bioactivity of the produced metabolites ahead of the time-consuming isolation procedures. Herein, we showcase a structure-agnostic bioactivity-driven combinatorial biosynthesis workflow that introduces bioactivity assessment as a selection-driving force to guide iterative combinatorial biosynthesis rounds towards enzyme combinations with increasing bioactivity. We apply this approach to produce triterpenoids with potent bioactivity against PTP1B, a promising molecular target for diabetes and cancer treatment. We demonstrate that the bioactivity-guided workflow can expedite the combinatorial process by enabling the narrowing down of more than 1000 possible combinations to only five highly potent candidates. By focusing the isolation and structural elucidation effort on only these five strains, we reveal 20 structurally diverse triterpenoids, including four new compounds and a novel triterpenoid-anthranilic acid hybrid, as potent PTP1B inhibitors. This workflow expedites hit identification by combinatorial biosynthesis and is applicable to many other types of bioactive natural products, therefore providing a strategy for accelerated drug discovery.
Collapse
Affiliation(s)
- Yong Zhao
- Biochemical Engineering Group, Department of Plant and Environment Sciences, Faculty of Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark
| | - Yao-Tao Duan
- Biochemical Engineering Group, Department of Plant and Environment Sciences, Faculty of Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark
| | - Jie Zang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen, Denmark
| | - Morten H Raadam
- Biochemical Engineering Group, Department of Plant and Environment Sciences, Faculty of Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark
| | - Irini Pateraki
- Biochemical Engineering Group, Department of Plant and Environment Sciences, Faculty of Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark
| | - Karel Miettinen
- Biochemical Engineering Group, Department of Plant and Environment Sciences, Faculty of Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark
| | - Dan Staerk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen, Denmark
| | - Sotirios C Kampranis
- Biochemical Engineering Group, Department of Plant and Environment Sciences, Faculty of Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark
| |
Collapse
|
2
|
Zhang JM, Yuan GY, Zou Y. Enzymatic ester bond formation strategies in fungal macrolide skeletons. Nat Prod Rep 2025. [PMID: 39831437 DOI: 10.1039/d4np00050a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Covering: up to August 2024Macrolides, the core skeletons of numerous marketed drugs and bioactive natural products, have garnered considerable scientific interest owing to their structural diversity and broad spectrum of pharmaceutical activities. The formation of intramolecular ester bonds is a critical biocatalytic step in constructing macrolide skeletons. Here, we summarised enzymatic ester bond formation strategies in fungal polyketide (PK)-type, nonribosomal peptide (NRP)-type, and PK-NRP hybrid-type macrolides. In PK-type macrolides, ester bond formation is commonly catalysed by a trans-acting thioesterase (TE) or a cis-acting TE domain during the product release process. In NRP-type and PK-NRP hybrid-type macrolides, the ester bond is usually introduced through condensation (C) domain-catalysed esterification during the elongation or product release step. Although the TE and C domains share similarities in their catalytic mechanism, using hydroxyl groups as nucleophiles in an intramolecular nucleophilic attack, they differ in terms of the hydroxyl origin, the timing of ester bond formation, and domain location. Furthermore, some TE domains are utilized as chemoenzymatic catalysts to construct macrolides with different ring sizes. A comparison of ester bond formation between fungi and bacteria is also discussed. Exploring the biosynthetic pathways of fungal macrolides, elucidating the diverse strategies employed in the formation of ester bonds, and understanding the application of enzymes/domains in chemoenzymatic synthesis hold promise for the discovery of new bioactive macrolides in the future.
Collapse
Affiliation(s)
- Jin-Mei Zhang
- College of Pharmaceutical Sciences, Southwest University, 400715 Chongqing, China.
| | - Guan-Yin Yuan
- College of Pharmaceutical Sciences, Southwest University, 400715 Chongqing, China.
| | - Yi Zou
- College of Pharmaceutical Sciences, Southwest University, 400715 Chongqing, China.
| |
Collapse
|
3
|
Graziano N, Arce-López B, Barbeyron T, Delage L, Gerometta E, Roullier C, Burgaud G, Poirier E, Martinelli L, Jany JL, Hymery N, Meslet-Cladiere L. Identification and Characterization of Two Aryl Sulfotransferases from Deep-Sea Marine Fungi and Their Implications in the Sulfation of Secondary Metabolites. Mar Drugs 2024; 22:572. [PMID: 39728146 DOI: 10.3390/md22120572] [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: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Sulfation plays a critical role in the biosynthesis of small molecules, regulatory mechanisms such as hormone signaling, and detoxification processes (phase II enzymes). The sulfation reaction is catalyzed by a broad family of enzymes known as sulfotransferases (SULTs), which have been extensively studied in animals due to their medical importance, but also in plant key processes. Despite the identification of some sulfated metabolites in fungi, the mechanisms underlying fungal sulfation remain largely unknown. To address this knowledge gap, we conducted a comprehensive search of available genomes, resulting in the identification of 174 putative SULT genes in the Ascomycota phylum. Phylogenetic analysis and structural modeling revealed that these SULTs belong to the aryl sulfotransferase family, and they are divided into two potential distinct clusters of PAPS-dependent SULTs within the fungal kingdom. SULT genes from two marine fungi isolated from deep-sea hydrothermal vents, Hortaea werneckii UBOCC-A-208029 (HwSULT) and Aspergillus sydowii UBOCC-A-108050 SULT (AsSULT), were selected as representatives of each cluster. Recombinant proteins were expressed in Escherichia coli and biochemically characterized. HwSULT demonstrated high and versatile activity, while AsSULT appeared more substrate-specific. Here, HwSULT was used to sulfate the mycotoxin zearalenone, enhancing its cytotoxicity toward healthy feline intestinal cells.
Collapse
Affiliation(s)
- Nicolas Graziano
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| | - Beatriz Arce-López
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| | - Tristan Barbeyron
- Laboratory of Integrative Biology of Marine Models (LBI2M), Station Biologique de Roscoff (SBR), CNRS, Sorbonne Université, F-29688 Roscoff, France
| | - Ludovic Delage
- Laboratory of Integrative Biology of Marine Models (LBI2M), Station Biologique de Roscoff (SBR), CNRS, Sorbonne Université, F-29688 Roscoff, France
| | - Elise Gerometta
- Institut des Substances et Organismes de la Mer, Nantes Université, ISOMER, UR 2160, F-44000 Nantes, France
| | - Catherine Roullier
- Institut des Substances et Organismes de la Mer, Nantes Université, ISOMER, UR 2160, F-44000 Nantes, France
| | - Gaëtan Burgaud
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| | - Elisabeth Poirier
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| | - Laure Martinelli
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Hans-Knöll Strasse 8, 07455 Jena, Germany
| | - Jean-Luc Jany
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| | - Nolwenn Hymery
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| | - Laurence Meslet-Cladiere
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Écologie Microbienne, F-29280 Plouzané, France
| |
Collapse
|
4
|
Skellam E, Rajendran S, Li L. Combinatorial biosynthesis for the engineering of novel fungal natural products. Commun Chem 2024; 7:89. [PMID: 38637654 PMCID: PMC11026467 DOI: 10.1038/s42004-024-01172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Natural products are small molecules synthesized by fungi, bacteria and plants, which historically have had a profound effect on human health and quality of life. These natural products have evolved over millions of years resulting in specific biological functions that may be of interest for pharmaceutical, agricultural, or nutraceutical use. Often natural products need to be structurally modified to make them suitable for specific applications. Combinatorial biosynthesis is a method to alter the composition of enzymes needed to synthesize a specific natural product resulting in structurally diversified molecules. In this review we discuss different approaches for combinatorial biosynthesis of natural products via engineering fungal enzymes and biosynthetic pathways. We highlight the biosynthetic knowledge gained from these studies and provide examples of new-to-nature bioactive molecules, including molecules synthesized using combinations of fungal and non-fungal enzymes.
Collapse
Affiliation(s)
- Elizabeth Skellam
- Department of Chemistry, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA.
- BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA.
- Department of Biological Sciences, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA.
| | - Sanjeevan Rajendran
- Department of Chemistry, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA
- BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA
| | - Lei Li
- Department of Chemistry, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA
- BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX, 76203, USA
| |
Collapse
|
5
|
Li L, Zhong W, Liu H, Espinosa-Artiles P, Xu YM, Wang C, Robles JMV, Paz TA, Inácio MC, Chen F, Xu Y, Gunatilaka AAL, Molnár I. Biosynthesis of Cytosporones in Leotiomycetous Filamentous Fungi. J Am Chem Soc 2024; 146:6189-6198. [PMID: 38386630 PMCID: PMC11106036 DOI: 10.1021/jacs.3c14066] [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] [Indexed: 02/24/2024]
Abstract
Polyketides with the isochroman-3-one pharmacophore are rare among fungal natural products as their biosynthesis requires an unorthodox S-type aromatic ring cyclization. Genome mining uncovered a conserved gene cluster in select leotiomycetous fungi that encodes the biosynthesis of cytosporones, including isochroman-3-one congeners. Combinatorial biosynthesis in total biosynthetic and biocatalytic formats in Saccharomyces cerevisiae and in vitro reconstitution of key reactions with purified enzymes revealed how cytosporone structural and bioactivity diversity is generated. The S-type acyl dihydroxyphenylacetic acid (ADA) core of cytosporones is assembled by a collaborating polyketide synthase pair. Thioesterase domain-catalyzed transesterification releases ADA esters, some of which are known Nur77 modulators. Alternatively, hydrolytic release allows C6 hydroxylation by a flavin-dependent monooxygenase, yielding a trihydroxybenzene moiety. Reduction of the C9 carbonyl by a short chain dehydrogenase/reductase initiates isochroman-3-one formation, affording cytosporones with cytotoxic and antimicrobial activity. Enoyl di- or trihydroxyphenylacetic acids are generated as shunt products, while isocroman-3,4-diones are formed by autoxidation. The cytosporone pathway offers novel polyketide biosynthetic enzymes for combinatorial synthetic biology to advance the production of "unnatural" natural products for drug discovery.
Collapse
Affiliation(s)
- Li Li
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
- College of Life Science, Yangtze University, Jingzhou 434025, P. R. China
| | - Weimao Zhong
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
| | - Hang Liu
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P. R. China
| | - Patricia Espinosa-Artiles
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
| | - Ya-ming Xu
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
| | - Chen Wang
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P. R. China
| | - Jose Manuel Verdugo Robles
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
| | - Tiago Antunes Paz
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Marielle Cascaes Inácio
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
| | - Fusheng Chen
- School of Life Sciences, Guizhou Normal University, Guiyang 550025, P. R. China
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Yuquan Xu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P. R. China
| | - A. A. Leslie Gunatilaka
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
| | - István Molnár
- Southwest Center for Natural Products Research, University of Arizona, Tucson 85719, Arizona, United States
- VTT Technical Research Center of Finland Ltd., Espoo 02150, Finland
| |
Collapse
|
6
|
Cox RJ. Engineered and total biosynthesis of fungal specialized metabolites. Nat Rev Chem 2024; 8:61-78. [PMID: 38172201 DOI: 10.1038/s41570-023-00564-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2023] [Indexed: 01/05/2024]
Abstract
Filamentous fungi produce a very wide range of complex and often bioactive metabolites, demonstrating their inherent ability as hosts of complex biosynthetic pathways. Recent advances in molecular sciences related to fungi have afforded the development of new tools that allow the rational total biosynthesis of highly complex specialized metabolites in a single process. Increasingly, these pathways can also be engineered to produce new metabolites. Engineering can be at the level of gene deletion, gene addition, formation of mixed pathways, engineering of scaffold synthases and engineering of tailoring enzymes. Combination of these approaches with hosts that can metabolize low-value waste streams opens the prospect of one-step syntheses from garbage.
Collapse
Affiliation(s)
- Russell J Cox
- Institute for Organic Chemistry and BMWZ, Leibniz University of Hannover, Hannover, Germany.
| |
Collapse
|
7
|
Li Y, Lin P, Lu X, Yan H, Wei H, Liu C, Liu X, Yang Y, Molnár I, Bai Z. Plasmid Copy Number Engineering Accelerates Fungal Polyketide Discovery upon Unnatural Polyketide Biosynthesis. ACS Synth Biol 2023; 12:2226-2235. [PMID: 37463503 DOI: 10.1021/acssynbio.3c00178] [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: 07/20/2023]
Abstract
Saccharomyces cerevisiae has been extensively used as a convenient synthetic biology chassis to reconstitute fungal polyketide biosynthetic pathways. Despite progress in refactoring these pathways for expression and optimization of the yeast production host by metabolic engineering, product yields often remain unsatisfactory. Such problems are especially acute when synthetic biological production is used for bioprospecting via genome mining or when chimeric fungal polyketide synthases (PKSs) are employed to produce novel bioactive compounds. In this work, we demonstrate that empirically balancing the expression levels of the two collaborating PKS subunits that afford benzenediol lactone (BDL)-type fungal polyketides is a facile strategy to improve the product yields. This is accomplished by systematically and independently altering the copy numbers of the two plasmids that express these PKS subunits. We applied this plasmid copy number engineering strategy to two orphan PKSs from genome mining where the yields of the presumed BDL products in S. cerevisiae were far too low for product isolation. This optimization resulted in product yield improvements of up to 10-fold, allowing for the successful isolation and structure elucidation of new BDL analogues. Heterocombinations of these PKS subunits from genome mining with those from previously identified BDL pathways led to the combinatorial biosynthesis of several additional novel BDL-type polyketides.
Collapse
Affiliation(s)
- Ye Li
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Pingxin Lin
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Xuan Lu
- School of Life Science and Biotechnology, Dalian University, Dalian 116622, China
| | - Hao Yan
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Huan Wei
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Chunli Liu
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Xiuxia Liu
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Yankun Yang
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - István Molnár
- Southwest Center for Natural Products Research, University of Arizona, Tucson, Arizona 85706, United States
- VTT Technical Research Centre of Finland Ltd., Espoo 02044, Finland
| | - Zhonghu Bai
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
8
|
Danaeifar M, Mazlomi MA. Combinatorial biosynthesis: playing chess with the metabolism. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2023; 25:171-190. [PMID: 35435779 DOI: 10.1080/10286020.2022.2065265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023]
Abstract
Secondary metabolites are a group of natural products that produced by bacteria, fungi and plants. Many applications of these compounds from medicine to industry have been discovered. However, some changes in their structure and biosynthesis mechanism are necessary for their properties to be more suitable and also for their production to be profitable. The main and most useful method to achieve this goal is combinatorial biosynthesis. This technique uses the multi-unit essence of the secondary metabolites biosynthetic enzymes to make changes in their order, structure and also the organism that produces them.
Collapse
Affiliation(s)
- Mohsen Danaeifar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Mohammad Ali Mazlomi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| |
Collapse
|
9
|
Liu Q, Zhang D, Gao S, Cai X, Yao M, Xu Y, Gong Y, Zheng K, Mao Y, Yang L, Yang D, Molnár I, Yang X. Didepside Formation by the Nonreducing Polyketide Synthase Preu6 of Preussia isomera Requires Interaction of Starter Acyl Transferase and Thioesterase Domains. Angew Chem Int Ed Engl 2023; 62:e202214379. [PMID: 36484777 DOI: 10.1002/anie.202214379] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/13/2022]
Abstract
Orsellinic acid (OA) derivatives are produced by filamentous fungi using nonreducing polyketide synthases (nrPKSs). The chain-releasing thioesterase (TE) domains of such nrPKSs were proposed to also catalyze dimerization to yield didepsides, such as lecanoric acid. Here, we use combinatorial domain exchanges, domain dissections and reconstitutions to reveal that the TE domain of the lecanoric acid synthase Preu6 of Preussia isomera must collaborate with the starter acyl transferase (SAT) domain from the same nrPKS. We show that artificial SAT-TE fusion proteins are highly effective catalysts and reprogram the ketide homologation chassis to form didepsides. We also demonstrate that dissected SAT and TE domains of Preu6 physically interact, and SAT and TE domains of OA-synthesizing nrPKSs may co-evolve. Our work highlights an unexpected domain-domain interaction in nrPKSs that must be considered for the combinatorial biosynthesis of unnatural didepsides, depsidones, and diphenyl ethers.
Collapse
Affiliation(s)
- Qingpei Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Dan Zhang
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Shuaibiao Gao
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Xianhua Cai
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Ming Yao
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Yao Xu
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Yifu Gong
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Ke Zheng
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Yigui Mao
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| | - Liyan Yang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, No.98 Daling Road, Nanning, 530007, P. R. China
| | - Dengfeng Yang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, No.98 Daling Road, Nanning, 530007, P. R. China
| | - István Molnár
- VTT Technical Research Centre of Finland, Division of Industrial Biotechnology and Food Solutions, Tietotie 2, Espoo, 02150, Finland
| | - Xiaolong Yang
- School of Pharmaceutical Sciences, South-Central Minzu University, 182 Minyuan Road, Hongshan District, Wuhan, 430074, P. R. China
| |
Collapse
|
10
|
Yan H, Fu Z, Lin P, Gu Y, Cao J, Li Y. Inhibition of human glioblastoma multiforme cells by 10,11-dehydrocurvularin through the MMP-2 and PI3K/AKT signaling pathways. Eur J Pharmacol 2022; 936:175348. [DOI: 10.1016/j.ejphar.2022.175348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 10/15/2022] [Accepted: 10/20/2022] [Indexed: 11/25/2022]
|
11
|
Skellam E. Biosynthesis of fungal polyketides by collaborating and trans-acting enzymes. Nat Prod Rep 2022; 39:754-783. [PMID: 34842268 DOI: 10.1039/d1np00056j] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Covering: 1999 up to 2021Fungal polyketides encompass a range of structurally diverse molecules with a wide variety of biological activities. The giant multifunctional enzymes that synthesize polyketide backbones remain enigmatic, as do many of the tailoring enzymes involved in functional modifications. Recent advances in elucidating biosynthetic gene clusters (BGCs) have revealed numerous examples of fungal polyketide synthases that require the action of collaborating enzymes to synthesize the carbon backbone. This review will discuss collaborating and trans-acting enzymes involved in loading, extending, and releasing polyketide intermediates from fungal polyketide synthases, and additional modifications introduced by trans-acting enzymes demonstrating the complexity encountered when investigating natural product biosynthesis in fungi.
Collapse
Affiliation(s)
- Elizabeth Skellam
- Department of Chemistry, BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX 76203, USA.
| |
Collapse
|
12
|
Kuttikrishnan S, Prabhu KS, Al Sharie AH, Al Zu'bi YO, Alali FQ, Oberlies NH, Ahmad A, El-Elimat T, Uddin S. Natural resorcylic acid lactones: A chemical biology approach for anticancer activity. Drug Discov Today 2021; 27:547-557. [PMID: 34655796 DOI: 10.1016/j.drudis.2021.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/25/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022]
Abstract
Resorcylic acid lactones (RALs) are fungal polyketides that consist of a β-resorcylic acid residue (2,4-dihydroxybenzoic acid) embedded in a macrolactone ring. RALs exhibit a broad range of biological activities, including anticancer activities. Following discovery of the selective Hsp90 inhibition activity of radicicol, the kinase inhibition activity of hypothemycin, monocillin II, 5Z-7-oxo-zeaenol, and L-783,277 RALs, and the nuclear factor kappa B (NF-κB) inhibition activity of the RAL zearalenone, have attracted great attention as potential therapeutics for cancer treatment. In this minireview, we focus on natural RALs that possess cytotoxic activities [IC50 values < 10 μM (or 4-5 μg/ml)], discussing their structures, isolation, occurrence, biological activities, and anticancer molecular mechanisms.
Collapse
Affiliation(s)
- Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed H Al Sharie
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Yazan O Al Zu'bi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Feras Q Alali
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; QU Health, Qatar University, Doha, Qatar
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, United States
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Tamam El-Elimat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory of Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
13
|
Okorafor IC, Chen M, Tang Y. High-Titer Production of Olivetolic Acid and Analogs in Engineered Fungal Host Using a Nonplant Biosynthetic Pathway. ACS Synth Biol 2021; 10:2159-2166. [PMID: 34415146 DOI: 10.1021/acssynbio.1c00309] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The microbial synthesis of cannabinoids and related molecules requires access to the intermediate olivetolic acid (OA). Whereas plant enzymes have been explored for E. coli and yeast biosynthesis, moderate yields and shunt product formation are major hurdles. Here, based on the chemical logic to form 2,4-dihydroxybenzoate-containing natural products, we discovered a set of fungal tandem polyketide synthases that can produce OA and the related octanoyl-primed derivative sphaerophorolcarboxylic acid in high titers using the model organism Aspergillus nidulans. This new set of enzymes will enable new synthetic biology strategies to access microbial cannabinoids.
Collapse
|
14
|
Tippelt A, Nett M. Saccharomyces cerevisiae as host for the recombinant production of polyketides and nonribosomal peptides. Microb Cell Fact 2021; 20:161. [PMID: 34412657 PMCID: PMC8374128 DOI: 10.1186/s12934-021-01650-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/05/2021] [Indexed: 01/30/2023] Open
Abstract
As a robust, fast growing and genetically tractable organism, the budding yeast Saccharomyces cerevisiae is one of the most widely used hosts in biotechnology. Its applications range from the manufacturing of vaccines and hormones to bulk chemicals and biofuels. In recent years, major efforts have been undertaken to expand this portfolio to include structurally complex natural products, such as polyketides and nonribosomally synthesized peptides. These compounds often have useful pharmacological properties, which make them valuable drugs for the treatment of infectious diseases, cancer, or autoimmune disorders. In nature, polyketides and nonribosomal peptides are generated by consecutive condensation reactions of short chain acyl-CoAs or amino acids, respectively, with the substrates and reaction intermediates being bound to large, multidomain enzymes. For the reconstitution of these multistep catalytic processes, the enzymatic assembly lines need to be functionally expressed and the required substrates must be supplied in reasonable quantities. Furthermore, the production hosts need to be protected from the toxicity of the biosynthetic products. In this review, we will summarize and evaluate the status quo regarding the heterologous production of polyketides and nonribosomal peptides in S. cerevisiae. Based on a comprehensive literature analysis, prerequisites for a successful pathway reconstitution could be deduced, as well as recurring bottlenecks in this microbial host.
Collapse
Affiliation(s)
- Anna Tippelt
- Department of Biochemical and Chemical Engineering, Laboratory of Technical Biology, TU Dortmund University, Emil-Figge-Strasse 66, 44227, Dortmund, Germany
| | - Markus Nett
- Department of Biochemical and Chemical Engineering, Laboratory of Technical Biology, TU Dortmund University, Emil-Figge-Strasse 66, 44227, Dortmund, Germany.
| |
Collapse
|
15
|
Xie L, Xiao D, Wang X, Wang C, Bai J, Yue Q, Yue H, Li Y, Molnár I, Xu Y, Zhang L. Combinatorial Biosynthesis of Sulfated Benzenediol Lactones with a Phenolic Sulfotransferase from Fusarium graminearum PH-1. mSphere 2020; 5:e00949-20. [PMID: 33239367 PMCID: PMC7690957 DOI: 10.1128/msphere.00949-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/04/2020] [Indexed: 11/20/2022] Open
Abstract
Total biosynthesis or whole-cell biocatalytic production of sulfated small molecules relies on the discovery and implementation of appropriate sulfotransferase enzymes. Although fungi are prominent biocatalysts and have been used to sulfate drug-like phenolics, no gene encoding a sulfotransferase enzyme has been functionally characterized from these organisms. Here, we identify a phenolic sulfotransferase, FgSULT1, by genome mining from the plant-pathogenic fungus Fusarium graminearum PH-1. We expressed FgSULT1 in a Saccharomyces cerevisiae chassis to modify a broad range of benzenediol lactones and their nonmacrocyclic congeners, together with an anthraquinone, with the resulting unnatural natural product (uNP) sulfates displaying increased solubility. FgSULT1 shares low similarity with known animal and plant sulfotransferases. Instead, it forms a sulfotransferase family with putative bacterial and fungal enzymes for phase II detoxification of xenobiotics and allelochemicals. Among fungi, putative FgSULT1 homologues are encoded in the genomes of Fusarium spp. and a few other genera in nonsyntenic regions, some of which may be related to catabolic sulfur recycling. Computational structure modeling combined with site-directed mutagenesis revealed that FgSULT1 retains the key catalytic residues and the typical fold of characterized animal and plant sulfotransferases. Our work opens the way for the discovery of hitherto unknown fungal sulfotransferases and provides a synthetic biological and enzymatic platform that can be adapted to produce bioactive sulfates, together with sulfate ester standards and probes for masked mycotoxins, precarcinogenic toxins, and xenobiotics.IMPORTANCE Sulfation is an expedient strategy to increase the solubility, bioavailability, and bioactivity of nutraceuticals and clinically important drugs. However, chemical or biological synthesis of sulfoconjugates is challenging. Genome mining, heterologous expression, homology structural modeling, and site-directed mutagenesis identified FgSULT1 of Fusarium graminearum PH-1 as a cytosolic sulfotransferase with the typical fold and active site architecture of characterized animal and plant sulfotransferases, despite low sequence similarity. FgSULT1 homologues are sparse in fungi but form a distinct clade with bacterial sulfotransferases. This study extends the functionally characterized sulfotransferase superfamily to the kingdom Fungi and demonstrates total biosynthetic and biocatalytic synthetic biological platforms to produce unnatural natural product (uNP) sulfoconjugates. Such uNP sulfates may be utilized for drug discovery in human and veterinary medicine and crop protection. Our synthetic biological methods may also be adapted to generate masked mycotoxin standards for food safety and environmental monitoring applications and to expose precarcinogenic xenobiotics.
Collapse
Affiliation(s)
- Linan Xie
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Dongliang Xiao
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Xiaojing Wang
- Southwest Center for Natural Products Research, University of Arizona, Tucson, Arizona, USA
- Microbial Pharmacology Laboratory, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Chen Wang
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Jing Bai
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
- School of Chemistry, Biology and Material Engineering, Suzhou University of Science and Technology, Suzhou City, Jiangsu Province, People's Republic of China
| | - Qun Yue
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Haitao Yue
- Department of Biology and Biotechnology, Xinjiang University, Urumqi, People's Republic of China
| | - Ye Li
- Southwest Center for Natural Products Research, University of Arizona, Tucson, Arizona, USA
- National Engineering Lab for Cereal Fermentation Technology, Jiangnan University, Wuxi, People's Republic of China
| | - István Molnár
- Southwest Center for Natural Products Research, University of Arizona, Tucson, Arizona, USA
| | - Yuquan Xu
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Liwen Zhang
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| |
Collapse
|
16
|
Wang C, Wang X, Zhang L, Yue Q, Liu Q, Xu YM, Gunatilaka AAL, Wei X, Xu Y, Molnár I. Intrinsic and Extrinsic Programming of Product Chain Length and Release Mode in Fungal Collaborating Iterative Polyketide Synthases. J Am Chem Soc 2020; 142:17093-17104. [PMID: 32833442 DOI: 10.1021/jacs.0c07050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Combinatorial biosynthesis with fungal polyketide synthases (PKSs) promises to produce unprecedented bioactive "unnatural" natural products (uNPs) for drug discovery. Genome mining of the dothideomycete Rhytidhysteron rufulum uncovered a collaborating highly reducing PKS (hrPKS)-nonreducing PKS (nrPKS) pair. These enzymes produce trace amounts of rare S-type benzenediol macrolactone congeners with a phenylacetate core in a heterologous host. However, subunit shuffling and domain swaps with voucher enzymes demonstrated that all PKS domains are highly productive. This contradiction led us to reveal novel programming layers exerted by the starter unit acyltransferase (SAT) and the thioesterase (TE) domains on the PKS system. First, macrocyclic vs linear product formation is dictated by the intrinsic biosynthetic program of the TE domain. Next, the chain length of the hrPKS product is strongly influenced in trans by the off-loading preferences of the nrPKS SAT domain. Last, TE domains are size-selective filters that facilitate or obstruct product formation from certain priming units. Thus, the intrinsic programs of the SAT and TE domains are both part of the extrinsic program of the hrPKS subunit and modulate the observable metaprogram of the whole PKS system. Reconstruction of SAT and TE phylogenies suggests that these domains travel different evolutionary trajectories, with the resulting divergence creating potential conflicts in the PKS metaprogram. Such conflicts often emerge in chimeric PKSs created by combinatorial biosynthesis, reducing biosynthetic efficiency or even incapacitating the system. Understanding the points of failure for such engineered biocatalysts is pivotal to advance the biosynthetic production of uNPs.
Collapse
Affiliation(s)
- Chen Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South Street, Beijing 100081, P. R. China.,Southwest Center for Natural Products Research, University of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United States
| | - Xiaojing Wang
- Microbial Pharmacology Laboratory, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai 201318, P. R. China
| | - Liwen Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - Qun Yue
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - Qingpei Liu
- Southwest Center for Natural Products Research, University of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United States.,School of Pharmaceutical Sciences, South-Central University for Nationalities, 182 Minyuan Road, Hongshan District, Wuhan 430074, P. R. China
| | - Ya-Ming Xu
- Southwest Center for Natural Products Research, University of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United States
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, University of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United States
| | - Xiaoyi Wei
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization/Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, P. R. China
| | - Yuquan Xu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - István Molnár
- Southwest Center for Natural Products Research, University of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United States
| |
Collapse
|
17
|
Koch AA, Schmidt JJ, Lowell AN, Hansen DA, Coburn KM, Chemler JA, Sherman DH. Probing Selectivity and Creating Structural Diversity Through Hybrid Polyketide Synthases. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Aaron A. Koch
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
| | - Jennifer J. Schmidt
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
| | - Andrew N. Lowell
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
- Current address: Department of Chemistry Virginia Tech Blacksburg VA 24061 USA
| | - Douglas A. Hansen
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
| | - Katherine M. Coburn
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
| | - Joseph A. Chemler
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
| | - David H. Sherman
- Life Sciences Institute The University of Michigan (USA) 210 Washtenaw Avenue Ann Arbor MI 48109-2216 USA
- Departments of Medicinal Chemistry, Chemistry, Microbiology & Immunology The University of Michigan USA
| |
Collapse
|
18
|
Yuan S, Gopal JV, Ren S, Chen L, Liu L, Gao Z. Anticancer fungal natural products: Mechanisms of action and biosynthesis. Eur J Med Chem 2020; 202:112502. [PMID: 32652407 DOI: 10.1016/j.ejmech.2020.112502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
Abstract
Many fungal metabolites show promising anticancer properties both in vitro and in animal models, and some synthetic analogs of those metabolites have progressed into clinical trials. However, currently, there are still no fungi-derived agents approved as anticancer drugs. Two potential reasons could be envisioned: 1) lacking a clear understanding of their anticancer mechanism of action, 2) unable to supply enough materials to support the preclinical and clinic developments. In this review, we will summarize recent efforts on elucidating the anticancer mechanisms and biosynthetic pathways of several promising anticancer fungal natural products.
Collapse
Affiliation(s)
- Siwen Yuan
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jannu Vinay Gopal
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shuya Ren
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Litong Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China
| | - Zhizeng Gao
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China.
| |
Collapse
|
19
|
Koch AA, Schmidt JJ, Lowell AN, Hansen DA, Coburn KM, Chemler JA, Sherman DH. Probing Selectivity and Creating Structural Diversity Through Hybrid Polyketide Synthases. Angew Chem Int Ed Engl 2020; 59:13575-13580. [PMID: 32357274 DOI: 10.1002/anie.202004991] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Indexed: 11/09/2022]
Abstract
Engineering polyketide synthases (PKS) to produce new metabolites requires an understanding of catalytic points of failure during substrate processing. Growing evidence indicates the thioesterase (TE) domain as a significant bottleneck within engineered PKS systems. We created a series of hybrid PKS modules bearing exchanged TE domains from heterologous pathways and challenged them with both native and non-native polyketide substrates. Reactions pairing wildtype PKS modules with non-native substrates primarily resulted in poor conversions to anticipated macrolactones. Likewise, product formation with native substrates and hybrid PKS modules bearing non-cognate TE domains was severely reduced. In contrast, non-native substrates were converted by most hybrid modules containing a substrate compatible TE, directly implicating this domain as the major catalytic gatekeeper and highlighting its value as a target for protein engineering to improve analog production in PKS pathways.
Collapse
Affiliation(s)
- Aaron A Koch
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA
| | - Jennifer J Schmidt
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA
| | - Andrew N Lowell
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA.,Current address: Department of Chemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Douglas A Hansen
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA
| | - Katherine M Coburn
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA
| | - Joseph A Chemler
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA
| | - David H Sherman
- Life Sciences Institute, The University of Michigan (USA), 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA.,Departments of Medicinal Chemistry, Chemistry, Microbiology & Immunology, The University of Michigan, USA
| |
Collapse
|
20
|
Zhou J, Gao Y, Chang JL, Yu HY, Chen J, Zhou M, Meng XG, Ruan HL. Resorcylic Acid Lactones from an Ilyonectria sp. JOURNAL OF NATURAL PRODUCTS 2020; 83:1505-1514. [PMID: 32323537 DOI: 10.1021/acs.jnatprod.9b01167] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Twelve new resorcylic acid lactones (RALs) including three new 16-membered RALs (1a, 1b and 2), eight new 14-membered RALs (3-10), and one new 12-membered RAL (11), along with five known 14-membered RALs (12-16), were identified from the fermentation of the soil-derived fungus Ilyonectria sp. sb65. Their structures were established by detailed analyses of 1D and 2D NMR, HRESIMS, and X-ray diffraction crystallography. All new compounds were evaluated for their cytotoxic effects against three human cancer cell lines, along with their potential as TRAIL sensitizers in TRAIL-resistant A549 human lung adenocarcinoma cells and their in vitro immunosuppressive effects against ConA-induced T-cell and LPS-induced B-cell proliferation.
Collapse
Affiliation(s)
- Jia Zhou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Wuhan 430030, People's Republic of China
| | - Ying Gao
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Wuhan 430030, People's Republic of China
| | - Jin-Ling Chang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Wuhan 430030, People's Republic of China
| | - Heng-Yi Yu
- Department of Pharmacy, Tongji Hospital Affiliated Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Juan Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Wuhan 430030, People's Republic of China
| | - Ming Zhou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Wuhan 430030, People's Republic of China
| | - Xiang-Gao Meng
- College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Han-Li Ruan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Wuhan 430030, People's Republic of China
| |
Collapse
|
21
|
Synthetic biology based construction of biological activity-related library of fungal decalin-containing diterpenoid pyrones. Nat Commun 2020; 11:1830. [PMID: 32286350 PMCID: PMC7156458 DOI: 10.1038/s41467-020-15664-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/19/2020] [Indexed: 12/12/2022] Open
Abstract
A synthetic biology method based on heterologous biosynthesis coupled with genome mining is a promising approach for increasing the opportunities to rationally access natural product with novel structures and biological activities through total biosynthesis and combinatorial biosynthesis. Here, we demonstrate the advantage of the synthetic biology method to explore biological activity-related chemical space through the comprehensive heterologous biosynthesis of fungal decalin-containing diterpenoid pyrones (DDPs). Genome mining reveals putative DDP biosynthetic gene clusters distributed in five fungal genera. In addition, we design extended DDP pathways by combinatorial biosynthesis. In total, ten DDP pathways, including five native pathways, four extended pathways and one shunt pathway, are heterologously reconstituted in a genetically tractable heterologous host, Aspergillus oryzae, resulting in the production of 22 DDPs, including 15 new analogues. We also demonstrate the advantage of expanding the diversity of DDPs to probe various bioactive molecules through a wide range of biological evaluations. Combining genome mining and heterologous expression in a genetically tractable host can lead to bioactive natural products discovery and production. Here, the authors employ this strategy for new decalin-containing diterpenoid pyrenes production by expressing native, extended, and shunt pathways in Aspergillus oryzae.
Collapse
|
22
|
Zhang JJ, Tang X, Moore BS. Genetic platforms for heterologous expression of microbial natural products. Nat Prod Rep 2019; 36:1313-1332. [PMID: 31197291 PMCID: PMC6750982 DOI: 10.1039/c9np00025a] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Covering: 2005 up to 2019Natural products are of paramount importance in human medicine. Not only are most antibacterial and anticancer drugs derived directly from or inspired by natural products, many other branches of medicine, such as immunology, neurology, and cardiology, have similarly benefited from natural product-based drugs. Typically, the genetic material required to synthesize a microbial specialized product is arranged in a multigene biosynthetic gene cluster (BGC), which codes for proteins associated with molecule construction, regulation, and transport. The ability to connect natural product compounds to BGCs and vice versa, along with ever-increasing knowledge of biosynthetic machineries, has spawned the field of genomics-guided natural product genome mining for the rational discovery of new chemical entities. One significant challenge in the field of natural product genome mining is how to rapidly link orphan biosynthetic genes to their associated chemical products. This review highlights state-of-the-art genetic platforms to identify, interrogate, and engineer BGCs from diverse microbial sources, which can be broken into three stages: (1) cloning and isolation of genomic loci, (2) heterologous expression in a host organism, and (3) genetic manipulation of cloned pathways. In the future, we envision natural product genome mining will be rapidly accelerated by de novo DNA synthesis and refactoring of whole biosynthetic pathways in combination with systematic heterologous expression methodologies.
Collapse
Affiliation(s)
- Jia Jia Zhang
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California at San Diego, La Jolla, California, USA.
| | - Xiaoyu Tang
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California at San Diego, La Jolla, California, USA.
| | - Bradley S Moore
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California at San Diego, La Jolla, California, USA. and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, California, USA
| |
Collapse
|
23
|
Xie L, Zhang L, Bai J, Yue Q, Zhang M, Li J, Wang C, Xu Y. Methylglucosylation of Phenolic Compounds by Fungal Glycosyltransferase-Methyltransferase Functional Modules. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8573-8580. [PMID: 31293156 DOI: 10.1021/acs.jafc.9b02819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Glycosylation endows both natural and synthetic small molecules with modulated physicochemical and biological properties. Plant and bacterial glycosyltransferases capable of decorating various privileged scaffolds have been extensively studied, but those from kingdom Fungi still remain underexploited. Here, we use a combination of genome mining and heterologous expression techniques to identify four novel glycosyltransferase-methyltransferase (GT-MT) functional modules from Hypocreales fungi. These GT-MT modules display decent substrate promiscuity and regiospecificity, methylglucosylating a panel of natural products such as flavonoids, stilbenoids, anthraquinones, and benzenediol lactones. Native GT-MT modules can be split up and regrouped into hybrid modules with similar or even improved efficacy as compared with native pairs. Methylglucosylation of kaempferol considerably improves its insecticidal activity against the larvae of oriental armyworm Mythimna separata (Walker). Our work provides a set of efficient biocatalysts for the combinatorial biosynthesis of small molecule glycosides that may have significant importance to the pharmaceutical, agricultural, and food industries.
Collapse
Affiliation(s)
- Linan Xie
- Biotechnology Research Institute , Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street , Beijing 100081 , P. R. China
| | - Liwen Zhang
- Biotechnology Research Institute , Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street , Beijing 100081 , P. R. China
| | - Jing Bai
- Biotechnology Research Institute , Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street , Beijing 100081 , P. R. China
| | - Qun Yue
- Biotechnology Research Institute , Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street , Beijing 100081 , P. R. China
| | - Min Zhang
- School of Agricultural Sciences , Zhengzhou University , Kexue Avenue 100 , Zhengzhou 450001 , P. R. China
| | - Jiancheng Li
- Institute of Plant Protection , Hebei Academy of Agriculture and Forestry Sciences , 437 Dongguan Street , Baoding 071000 , P. R. China
| | - Chen Wang
- Biotechnology Research Institute , Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street , Beijing 100081 , P. R. China
| | - Yuquan Xu
- Biotechnology Research Institute , Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street , Beijing 100081 , P. R. China
- Agricultural Genomics Institute at Shenzhen , Chinese Academy of Agricultural Sciences , 7 Pengfei Road , Shenzhen 518124 , P. R. China
| |
Collapse
|
24
|
Wang X, Wang C, Duan L, Zhang L, Liu H, Xu YM, Liu Q, Mao T, Zhang W, Chen M, Lin M, Gunatilaka AAL, Xu Y, Molnár I. Rational Reprogramming of O-Methylation Regioselectivity for Combinatorial Biosynthetic Tailoring of Benzenediol Lactone Scaffolds. J Am Chem Soc 2019; 141:4355-4364. [PMID: 30767524 PMCID: PMC6416077 DOI: 10.1021/jacs.8b12967] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Indexed: 11/28/2022]
Abstract
O-Methylation modulates the pharmacokinetic and pharmacodynamic (PK/PD) properties of small-molecule natural products, affecting their bioavailability, stability, and binding to targets. Diversity-oriented combinatorial biosynthesis of new chemical entities for drug discovery and optimization of known bioactive scaffolds during drug development both demand efficient O-methyltransferase (OMT) biocatalysts with considerable substrate promiscuity and tunable regioselectivity that can be deployed in a scalable and sustainable manner. Here we demonstrate efficient total biosynthetic and biocatalytic platforms that use a pair of fungal OMTs with orthogonal regiospecificity to produce unnatural O-methylated benzenediol lactone polyketides. We show that rational, structure-guided active-site cavity engineering can reprogram the regioselectivity of these enzymes. We also characterize the interplay of engineered regioselectivity with substrate plasticity. These findings will guide combinatorial biosynthetic tailoring of unnatural products toward the generation of diverse chemical matter for drug discovery and the PK/PD optimization of bioactive scaffolds for drug development.
Collapse
Affiliation(s)
- Xiaojing Wang
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
- State
Key Laboratory of Plant Physiology and Biochemistry, Department of
Plant Sciences, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Chen Wang
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
| | - Lixin Duan
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
- Guangzhou
University of Chinese Medicine, 232 Waihuan East Road, Guangzhou University
City, Panyu District, Guangzhou 510006, P.R. China
| | - Liwen Zhang
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
| | - Hang Liu
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
| | - Ya-ming Xu
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
| | - Qingpei Liu
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
- Key
Laboratory of Environment Correlative Dietology, College of Food Science
and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Tonglin Mao
- State
Key Laboratory of Plant Physiology and Biochemistry, Department of
Plant Sciences, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Wei Zhang
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
| | - Ming Chen
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
| | - Min Lin
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
| | - A. A. Leslie Gunatilaka
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
| | - Yuquan Xu
- Biotechnology
Research Institute, Chinese Academy of Agricultural
Sciences, 12 Zhongguancun South Street, Beijing 100081, P.R. China
| | - István Molnár
- Southwest
Center for Natural Products Research, University
of Arizona, 250 East Valencia Road, Tucson, Arizona 85706, United
States
| |
Collapse
|
25
|
Strategies for Engineering Natural Product Biosynthesis in Fungi. Trends Biotechnol 2018; 37:416-427. [PMID: 30316556 DOI: 10.1016/j.tibtech.2018.09.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/02/2018] [Accepted: 09/12/2018] [Indexed: 01/22/2023]
Abstract
Fungi are a prolific source of bioactive compounds, some of which have been developed as essential medicines and life-enhancing drugs. Genome sequencing has revealed that fungi have the potential to produce considerably more natural products (NPs) than are typically observed in the laboratory. Recently, there have been significant advances in the identification, understanding, and engineering of fungal biosynthetic gene clusters (BGCs). This review briefly describes examples of the engineering of fungal NP biosynthesis at the global, pathway, and enzyme level using in vivo and in vitro approaches and refers to the range and scale of heterologous expression systems available, developments in combinatorial biosynthesis, progress in understanding how fungal BGCs are regulated, and the applications of these novel biosynthetic enzymes as biocatalysts.
Collapse
|
26
|
Synthetic biology toolkits and applications in Saccharomyces cerevisiae. Biotechnol Adv 2018; 36:1870-1881. [PMID: 30031049 DOI: 10.1016/j.biotechadv.2018.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022]
Abstract
Synthetic biologists construct biological components and systems to look into biological phenomena and drive a myriad of practical applications that aim to tackle current global challenges in energy, healthcare and the environment. While most tools have been established in bacteria, particularly Escherichia coli, recent years have seen parallel developments in the model yeast strain Saccharomyces cerevisiae, one of the most well-understood eukaryotic biological system. Here, we outline the latest advances in yeast synthetic biology tools based on a framework of abstraction hierarchies of parts, circuits and genomes. In brief, the creation and characterization of biological parts are explored at the transcriptional, translational and post-translational levels. Using characterized parts as building block units, the designing of functional circuits is elaborated with examples. In addition, the status and potential applications of synthetic genomes as a genome level platform for biological system construction are also discussed. In addition to the development of a toolkit, we describe how those tools have been applied in the areas of drug production and screening, study of disease mechanisms, pollutant sensing and bioremediation. Finally, we provide a future outlook of yeast as a workhorse of eukaryotic genetics and a chosen chassis in this field.
Collapse
|
27
|
Xie L, Zhang L, Wang C, Wang X, Xu YM, Yu H, Wu P, Li S, Han L, Gunatilaka AAL, Wei X, Lin M, Molnár I, Xu Y. Methylglucosylation of aromatic amino and phenolic moieties of drug-like biosynthons by combinatorial biosynthesis. Proc Natl Acad Sci U S A 2018; 115:E4980-E4989. [PMID: 29760061 PMCID: PMC5984488 DOI: 10.1073/pnas.1716046115] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glycosylation is a prominent strategy to optimize the pharmacokinetic and pharmacodynamic properties of drug-like small-molecule scaffolds by modulating their solubility, stability, bioavailability, and bioactivity. Glycosyltransferases applicable for "sugarcoating" various small-molecule acceptors have been isolated and characterized from plants and bacteria, but remained cryptic from filamentous fungi until recently, despite the frequent use of some fungi for whole-cell biocatalytic glycosylations. Here, we use bioinformatic and genomic tools combined with heterologous expression to identify a glycosyltransferase-methyltransferase (GT-MT) gene pair that encodes a methylglucosylation functional module in the ascomycetous fungus Beauveria bassiana The GT is the founding member of a family nonorthologous to characterized fungal enzymes. Using combinatorial biosynthetic and biocatalytic platforms, we reveal that this GT is a promiscuous enzyme that efficiently modifies a broad range of drug-like substrates, including polyketides, anthraquinones, flavonoids, and naphthalenes. It yields both O- and N-glucosides with remarkable regio- and stereospecificity, a spectrum not demonstrated for other characterized fungal enzymes. These glucosides are faithfully processed by the dedicated MT to afford 4-O-methylglucosides. The resulting "unnatural products" show increased solubility, while representative polyketide methylglucosides also display increased stability against glycoside hydrolysis. Upon methylglucosidation, specific polyketides were found to attain cancer cell line-specific antiproliferative or matrix attachment inhibitory activities. These findings will guide genome mining for fungal GTs with novel substrate and product specificities, and empower the efficient combinatorial biosynthesis of a broad range of natural and unnatural glycosides in total biosynthetic or biocatalytic formats.
Collapse
Affiliation(s)
- Linan Xie
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China
| | - Liwen Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China
| | - Chen Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China
- Natural Products Center, University of Arizona, Tucson, AZ 85706
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, 510650 Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, 510650 Guangzhou, People's Republic of China
| | - Xiaojing Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China
- Natural Products Center, University of Arizona, Tucson, AZ 85706
| | - Ya-Ming Xu
- Natural Products Center, University of Arizona, Tucson, AZ 85706
| | - Hefen Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical University, 100069 Beijing, People's Republic of China
| | - Ping Wu
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, 510650 Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, 510650 Guangzhou, People's Republic of China
| | - Shenglan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical University, 100069 Beijing, People's Republic of China
| | - Lida Han
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China
| | | | - Xiaoyi Wei
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, 510650 Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, 510650 Guangzhou, People's Republic of China
| | - Min Lin
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China;
| | - István Molnár
- Natural Products Center, University of Arizona, Tucson, AZ 85706;
| | - Yuquan Xu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, 100081 Beijing, People's Republic of China;
| |
Collapse
|
28
|
Schor R, Cox R. Classic fungal natural products in the genomic age: the molecular legacy of Harold Raistrick. Nat Prod Rep 2018. [PMID: 29537034 DOI: 10.1039/c8np00021b] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Covering: 1893 to 2017Harold Raistrick was involved in the discovery of many of the most important classes of fungal metabolites during the 20th century. This review focusses on how these discoveries led to developments in isotopic labelling, biomimetic chemistry and the discovery, analysis and exploitation of biosynthetic gene clusters for major classes of fungal metabolites including: alternariol; geodin and metabolites of the emodin pathway; maleidrides; citrinin and the azaphilones; dehydrocurvularin; mycophenolic acid; and the tropolones. Key recent advances in the molecular understanding of these important pathways, including the discovery of biosynthetic gene clusters, the investigation of the molecular and chemical aspects of key biosynthetic steps, and the reengineering of key components of the pathways are reviewed and compared. Finally, discussion of key relationships between metabolites and pathways and the most important recent advances and opportunities for future research directions are given.
Collapse
Affiliation(s)
- Raissa Schor
- Institut für Organische Chemie, BMWZ, Leibniz Universität Hannover, Germany.
| | - Russell Cox
- Institut für Organische Chemie, BMWZ, Leibniz Universität Hannover, Germany.
| |
Collapse
|
29
|
Guzmán-Trampe S, Ceapa CD, Manzo-Ruiz M, Sánchez S. Synthetic biology era: Improving antibiotic’s world. Biochem Pharmacol 2017; 134:99-113. [DOI: 10.1016/j.bcp.2017.01.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
|
30
|
Liu YN, Zhang TJ, Lu XX, Ma BL, Ren A, Shi L, Jiang AL, Yu HS, Zhao MW. Membrane fluidity is involved in the regulation of heat stress induced secondary metabolism in Ganoderma lucidum. Environ Microbiol 2017; 19:1653-1668. [PMID: 28198137 DOI: 10.1111/1462-2920.13693] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/06/2017] [Indexed: 01/17/2023]
Abstract
Ganoderma lucidum has become a potential model system for evaluating how environmental factors regulate the secondary metabolism of basidiomycetes. Heat stress (HS) is one of the most important environmental factors. It was previously reported that HS could induce the biosynthesis of ganoderic acids (GA). In this study, we found that HS increased GA biosynthesis and also significantly increased cell membrane fluidity. Furthermore, our results showed that addition of the membrane rigidifier dimethylsulfoxide (DMSO) could revert the increased GA biosynthesis elicited by HS. These results indicate that an increase in membrane fluidity is associated with HS-induced GA biosynthesis. Further evidence showed that the GA content was decreased in D9des-silenced strains and could be reverted to WT levels by addition of the membrane fluidizer benzyl alcohol (BA). In contrast, GA content was increased in D9des-overexpression strains and could be reverted to WT levels by the addition of DMSO. Furthermore, both membrane fluidity and GA biosynthesis induced by HS could be reverted by DMSO in WT and D9des-silenced strains. To the best of our knowledge, this is the first report demonstrating that membrane fluidity is involved in the regulation of heat stress induced secondary metabolism in filamentous fungi.
Collapse
Affiliation(s)
- Yong-Nan Liu
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Tian-Jun Zhang
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Xiao-Xiao Lu
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Bao-Liang Ma
- Department of Physics, Science of College, Nanjing Agricultural University, Nanjing, 210095, P.R China
| | - Ang Ren
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Liang Shi
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Ai-Liang Jiang
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Han-Shou Yu
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| | - Ming-Wen Zhao
- Key Laboratory of Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P.R. China
| |
Collapse
|
31
|
Sun L, Wang S, Zhang S, Shao L, Zhang Q, Skidmore C, Chang CWT, Yu D, Zhan J. Characterization of Three Tailoring Enzymes in Dutomycin Biosynthesis and Generation of a Potent Antibacterial Analogue. ACS Chem Biol 2016; 11:1992-2001. [PMID: 27195476 DOI: 10.1021/acschembio.6b00245] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The anthracycline natural product dutomycin and its precursor POK-MD1 were isolated from Streptomyces minoensis NRRL B-5482. The dutomycin biosynthetic gene cluster was identified by genome sequencing and disruption of the ketosynthase gene. Two polyketide synthase (PKS) systems are present in the gene cluster, including a type II PKS and a rare highly reducing iterative type I PKS. The type I PKS DutG repeatedly uses its active sites to create a nine-carbon triketide chain that is subsequently transferred to the α-l-axenose moiety of POK-MD1 at 4″-OH to yield dutomycin. Using a heterologous recombination approach, we disrupted a putative methyltransferase gene (dutMT1) and two glycosyltransferase genes (dutGT1 and dutGT2). Analysis of the metabolites of these mutants revealed the functions of these genes and yielded three dutomycin analogues SW140, SW91, and SW75. The major product SW91 in Streptomyces minoensis NRRL B-5482-ΔDutMT1 was identified as 12-desmethyl-dutomycin, suggesting that DutMT1 is the dedicated 12-methyltransferase. This was confirmed by the in vitro enzymatic assay. DutGT1 and DutGT2 were found to be responsible for the introduction of β-d-amicetose and α-l-axenose, respectively. Dutomycin and SW91 showed strong antibacterial activity against Staphylococcus aureus and methicillin-resistant S. aureus, whereas POK-MD1 and SW75 had no obvious inhibition, which revealed the essential role of the C-4″ triketide chain in antibacterial activity. The minimal inhibitory concentration of SW91 against the two strains was 0.125 μg mL(-1), lower than that of dutomycin (0.25 μg mL(-1)), indicating that the antibacterial activity of dutomycin can be improved through biosynthetic structural modification.
Collapse
Affiliation(s)
- Lei Sun
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
| | - Siyuan Wang
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
| | - Shuwei Zhang
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
| | - Lei Shao
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
| | - Qian Zhang
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Chad Skidmore
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
| | - Cheng-Wei Tom Chang
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Dayu Yu
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
- Department
of Applied Chemistry and Biological Engineering, College of Chemical
Engineering, Northeast Dianli University, Jilin, Jilin 132012, China
| | - Jixun Zhan
- Department
of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, Utah 84322-4105, United States
- TCM and Ethnomedicine Innovation & Development Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
32
|
Abstract
Polyketide biosynthesis engages a series of well-timed biosynthetic operations to generate elaborate natural products from simple building blocks. Mimicry of these processes has offered practical means for total synthesis and provided a foundation for reaction discovery. We now report an unusual intramolecular trans-amidation reaction discovered while preparing stabilized probes for the study of actinorhodin biosynthesis. This rapid cyclization event offers insight into the natural cyclization process inherent to the biosynthesis of type II polyketide antibiotics.
Collapse
|
33
|
Khater S, Anand S, Mohanty D. In silico methods for linking genes and secondary metabolites: The way forward. Synth Syst Biotechnol 2016; 1:80-88. [PMID: 29062931 PMCID: PMC5640692 DOI: 10.1016/j.synbio.2016.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/18/2016] [Accepted: 03/01/2016] [Indexed: 11/26/2022] Open
Abstract
In silico methods for linking genomic space to chemical space have played a crucial role in genomics driven discovery of new natural products as well as biosynthesis of altered natural products by engineering of biosynthetic pathways. Here we give an overview of available computational tools and then briefly describe a novel computational framework, namely retro-biosynthetic enumeration of biosynthetic reactions, which can add to the repertoire of computational tools available for connecting natural products to their biosynthetic gene clusters. Most of the currently available bioinformatics tools for analysis of secondary metabolite biosynthetic gene clusters utilize the “Genes to Metabolites” approach. In contrast to the “Genes to Metabolites” approach, the “Metabolites to Genes” or retro-biosynthetic approach would involve enumerating the various biochemical transformations or enzymatic reactions which would generate the given chemical moiety starting from a set of precursor molecules and identifying enzymatic domains which can potentially catalyze the enumerated biochemical transformations. In this article, we first give a brief overview of the presently available in silico tools and approaches for analysis of secondary metabolite biosynthetic pathways. We also discuss our preliminary work on development of algorithms for retro-biosynthetic enumeration of biochemical transformations to formulate a novel computational method for identifying genes associated with biosynthesis of a given polyketide or nonribosomal peptide.
Collapse
Affiliation(s)
- Shradha Khater
- Bioinformatics Center, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Swadha Anand
- Bioinformatics Center, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Debasisa Mohanty
- Bioinformatics Center, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
34
|
Bond C, Tang Y, Li L. Saccharomyces cerevisiae as a tool for mining, studying and engineering fungal polyketide synthases. Fungal Genet Biol 2016; 89:52-61. [PMID: 26850128 PMCID: PMC4789138 DOI: 10.1016/j.fgb.2016.01.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/01/2016] [Accepted: 01/09/2016] [Indexed: 12/17/2022]
Abstract
Small molecule secondary metabolites produced by organisms such as plants, bacteria, and fungi form a fascinating and important group of natural products, many of which have shown promise as medicines. Fungi in particular have been important sources of natural product polyketide pharmaceuticals. While the structural complexity of these polyketides makes them interesting and useful bioactive compounds, these same features also make them difficult and expensive to prepare and scale-up using synthetic methods. Currently, nearly all commercial polyketides are prepared through fermentation or semi-synthesis. However, elucidation and engineering of polyketide pathways in the native filamentous fungi hosts are often hampered due to a lack of established genetic tools and of understanding of the regulation of fungal secondary metabolisms. Saccharomyces cerevisiae has many advantages beneficial to the study and development of polyketide pathways from filamentous fungi due to its extensive genetic toolbox and well-studied metabolism. This review highlights the benefits S. cerevisiae provides as a tool for mining, studying, and engineering fungal polyketide synthases (PKSs), as well as notable insights this versatile tool has given us into the mechanisms and products of fungal PKSs.
Collapse
Affiliation(s)
- Carly Bond
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, United States
| | - Yi Tang
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, United States; Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, United States.
| | - Li Li
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, United States; Engineering Research Center of Industrial Microbiology (Ministry of Education), College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, China; State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
35
|
Sun L, Wang S, Zhang S, Yu D, Qin Y, Huang H, Wang W, Zhan J. Identification of a type III polyketide synthase involved in the biosynthesis of spirolaxine. Appl Microbiol Biotechnol 2016; 100:7103-13. [DOI: 10.1007/s00253-016-7444-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/29/2016] [Accepted: 03/06/2016] [Indexed: 11/30/2022]
|
36
|
King JR, Edgar S, Qiao K, Stephanopoulos G. Accessing Nature's diversity through metabolic engineering and synthetic biology. F1000Res 2016; 5. [PMID: 27081481 PMCID: PMC4813638 DOI: 10.12688/f1000research.7311.1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
In this perspective, we highlight recent examples and trends in metabolic engineering and synthetic biology that demonstrate the synthetic potential of enzyme and pathway engineering for natural product discovery. In doing so, we introduce natural paradigms of secondary metabolism whereby simple carbon substrates are combined into complex molecules through “scaffold diversification”, and subsequent “derivatization” of these scaffolds is used to synthesize distinct complex natural products. We provide examples in which modern pathway engineering efforts including combinatorial biosynthesis and biological retrosynthesis can be coupled to directed enzyme evolution and rational enzyme engineering to allow access to the “privileged” chemical space of natural products in industry-proven microbes. Finally, we forecast the potential to produce natural product-like discovery platforms in biological systems that are amenable to single-step discovery, validation, and synthesis for streamlined discovery and production of biologically active agents.
Collapse
Affiliation(s)
- Jason R King
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Steven Edgar
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kangjian Qiao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
37
|
Zhang L, Zhou Z, Guo Q, Fokkens L, Miskei M, Pócsi I, Zhang W, Chen M, Wang L, Sun Y, Donzelli BGG, Gibson DM, Nelson DR, Luo JG, Rep M, Liu H, Yang S, Wang J, Krasnoff SB, Xu Y, Molnár I, Lin M. Insights into Adaptations to a Near-Obligate Nematode Endoparasitic Lifestyle from the Finished Genome of Drechmeria coniospora. Sci Rep 2016; 6:23122. [PMID: 26975455 PMCID: PMC4792172 DOI: 10.1038/srep23122] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/29/2016] [Indexed: 12/18/2022] Open
Abstract
Nematophagous fungi employ three distinct predatory strategies: nematode trapping, parasitism of females and eggs, and endoparasitism. While endoparasites play key roles in controlling nematode populations in nature, their application for integrated pest management is hindered by the limited understanding of their biology. We present a comparative analysis of a high quality finished genome assembly of Drechmeria coniospora, a model endoparasitic nematophagous fungus, integrated with a transcriptomic study. Adaptation of D. coniospora to its almost completely obligate endoparasitic lifestyle led to the simplification of many orthologous gene families involved in the saprophytic trophic mode, while maintaining orthologs of most known fungal pathogen-host interaction proteins, stress response circuits and putative effectors of the small secreted protein type. The need to adhere to and penetrate the host cuticle led to a selective radiation of surface proteins and hydrolytic enzymes. Although the endoparasite has a simplified secondary metabolome, it produces a novel peptaibiotic family that shows antibacterial, antifungal and nematicidal activities. Our analyses emphasize the basic malleability of the D. coniospora genome: loss of genes advantageous for the saprophytic lifestyle; modulation of elements that its cohort species utilize for entomopathogenesis; and expansion of protein families necessary for the nematode endoparasitic lifestyle.
Collapse
Affiliation(s)
- Liwen Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhengfu Zhou
- Key Laboratory of Agricultural Genomics (Beijing), Ministry of Agriculture, China
| | - Qiannan Guo
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Like Fokkens
- Molecular Plant Pathology, University of Amsterdam, Amsterdam, the Netherlands
| | - Márton Miskei
- Department of Biotechnology and Microbiology, Faculty of Science and Technology, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - István Pócsi
- Department of Biotechnology and Microbiology, Faculty of Science and Technology, University of Debrecen, Hungary
| | - Wei Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ming Chen
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lei Wang
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Yamin Sun
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Bruno G. G. Donzelli
- Plant Pathology & Plant-Microbe Biology, Cornell University, Ithaca, New York, USA
| | - Donna M. Gibson
- USDA-ARS, Robert W. Holley Center for Agriculture and Health, Ithaca, New York, USA
| | - David R. Nelson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jian-Guang Luo
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Martijn Rep
- Molecular Plant Pathology, University of Amsterdam, Amsterdam, the Netherlands
| | - Hang Liu
- Key Laboratory of Agricultural Genomics (Beijing), Ministry of Agriculture, China
| | - Shengnan Yang
- Key Laboratory of Agricultural Genomics (Beijing), Ministry of Agriculture, China
| | - Jing Wang
- Key Laboratory of Agricultural Genomics (Beijing), Ministry of Agriculture, China
| | - Stuart B. Krasnoff
- USDA-ARS, Robert W. Holley Center for Agriculture and Health, Ithaca, New York, USA
| | - Yuquan Xu
- Key Laboratory of Agricultural Genomics (Beijing), Ministry of Agriculture, China
| | - István Molnár
- Natural Products Center, School of Natural Resources and the Environment, University of Arizona, Tucson, Arizona, USA
| | - Min Lin
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
38
|
Bai J, Lu Y, Xu YM, Zhang W, Chen M, Lin M, Gunatilaka AAL, Xu Y, Molnár I. Diversity-Oriented Combinatorial Biosynthesis of Hybrid Polyketide Scaffolds from Azaphilone and Benzenediol Lactone Biosynthons. Org Lett 2016; 18:1262-5. [PMID: 26934205 DOI: 10.1021/acs.orglett.6b00110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two disparate polyketide families, the benzenediol lactones and the azaphilones, are produced by fungi using iterative polyketide synthase (iPKS) enzymes consisting of collaborating partner subunits. Exploitation of this common biosynthetic logic using iPKS subunit shuffling allowed the diversity-oriented combinatorial biosynthesis of unprecedented polyketide scaffolds new to nature, bearing structural motifs from both of these orthogonal natural product families. Starter unit acyltransferase domain replacements proved necessary but not sufficient to guarantee communication between iPKS subunits.
Collapse
Affiliation(s)
- Jing Bai
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street, Beijing 100081, P. R. China.,Natural Products Center, School of Natural Resources and the Environment, The University of Arizona , 250 East Valencia Road, Tucson, Arizona 85706, United States
| | - Yuanyuan Lu
- Natural Products Center, School of Natural Resources and the Environment, The University of Arizona , 250 East Valencia Road, Tucson, Arizona 85706, United States.,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University , 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Ya-ming Xu
- Natural Products Center, School of Natural Resources and the Environment, The University of Arizona , 250 East Valencia Road, Tucson, Arizona 85706, United States
| | - Wei Zhang
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - Ming Chen
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - Min Lin
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - A A Leslie Gunatilaka
- Natural Products Center, School of Natural Resources and the Environment, The University of Arizona , 250 East Valencia Road, Tucson, Arizona 85706, United States
| | - Yuquan Xu
- Biotechnology Research Institute, The Chinese Academy of Agricultural Sciences , 12 Zhongguancun South Street, Beijing 100081, P. R. China
| | - István Molnár
- Natural Products Center, School of Natural Resources and the Environment, The University of Arizona , 250 East Valencia Road, Tucson, Arizona 85706, United States
| |
Collapse
|
39
|
Abstract
Polyketides are a diverse group of natural products that form the basis of many important drugs. The engineering of the polyketide synthase (PKS) enzymes responsible for the formation of these compounds has long been considered to have great potential for producing new bioactive molecules. Recent advances in this field have contributed to the understanding of this powerful and complex enzymatic machinery, particularly with regard to domain activity and engineering, unique building block formation and incorporation, and programming rules and limitations. New developments in tools for
in vitro biochemical analysis, full-length megasynthase structural studies, and
in vivo heterologous expression will continue to improve our fundamental understanding of polyketide synthesis as well as our ability to engineer the production of polyketides.
Collapse
Affiliation(s)
- Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Joyce Liu
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| |
Collapse
|
40
|
Rational biosynthetic approaches for the production of new-to-nature compounds in fungi. Fungal Genet Biol 2016; 89:89-101. [PMID: 26872866 DOI: 10.1016/j.fgb.2016.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 02/04/2016] [Accepted: 02/04/2016] [Indexed: 01/06/2023]
Abstract
Filamentous fungi have the ability to produce a wide range of secondary metabolites some of which are potent toxins whereas others are exploited as food additives or drugs. Fungal natural products still play an important role in the discovery of new chemical entities for potential use as pharmaceuticals. However, in most cases they cannot be directly used as drugs due to toxic side effects or suboptimal pharmacokinetics. To improve drug-like properties, including bioactivity and stability or to produce better precursors for semi-synthetic routes, one needs to generate non-natural derivatives from known fungal secondary metabolites. In this minireview, we describe past and recent biosynthetic approaches for the diversification of fungal natural products, covering examples from precursor-directed biosynthesis, mutasynthesis, metabolic engineering and biocombinatorial synthesis. To illustrate the current state-of-the-art, challenges and pitfalls, we lay particular emphasis on the class of fungal cyclodepsipeptides which have been studied longtime for product diversification and which are of pharmaceutical relevance as drugs.
Collapse
|
41
|
Meepagala KM, Johnson RD, Duke SO. Curvularin and Dehydrocurvularin as Phytotoxic Constituents from <i>Curvularia intermedia</i> Infecting <i>Pandanus amaryllifolius</i>. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/jacen.2016.51002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
42
|
Horsman ME, Hari TPA, Boddy CN. Polyketide synthase and non-ribosomal peptide synthetase thioesterase selectivity: logic gate or a victim of fate? Nat Prod Rep 2016; 33:183-202. [DOI: 10.1039/c4np00148f] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Thioesterases (TEs) are product offloading enzymes from FAS, PKS, and NRPS complexes. We review the diversity, structure, and mechanism of PKS and NRPS TEs and analyze TE loading and release steps as possible logic gates with a view to predicting TE function in new pathways.
Collapse
Affiliation(s)
- Mark E. Horsman
- Department of chemistry
- Centre for Catalysis Research and Innovation
- University of Ottawa
- Canada
| | - Taylor P. A. Hari
- Department of chemistry
- Centre for Catalysis Research and Innovation
- University of Ottawa
- Canada
| | - Christopher N. Boddy
- Department of chemistry
- Centre for Catalysis Research and Innovation
- University of Ottawa
- Canada
| |
Collapse
|
43
|
Abstract
Synthetic biology (SB) is an emerging discipline, which is slowly reorienting the field of drug discovery. For thousands of years, living organisms such as plants were the major source of human medicines. The difficulty in resynthesizing natural products, however, often turned pharmaceutical industries away from this rich source for human medicine. More recently, progress on transformation through genetic manipulation of biosynthetic units in microorganisms has opened the possibility of in-depth exploration of the large chemical space of natural products derivatives. Success of SB in drug synthesis culminated with the bioproduction of artemisinin by microorganisms, a tour de force in protein and metabolic engineering. Today, synthetic cells are not only used as biofactories but also used as cell-based screening platforms for both target-based and phenotypic-based approaches. Engineered genetic circuits in synthetic cells are also used to decipher disease mechanisms or drug mechanism of actions and to study cell-cell communication within bacteria consortia. This review presents latest developments of SB in the field of drug discovery, including some challenging issues such as drug resistance and drug toxicity.
Collapse
Affiliation(s)
| | - Pablo Carbonell
- Faculty of Life Sciences, SYNBIOCHEM Centre, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
- Department of Experimental and Health Sciences (DCEXS), Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
44
|
Affiliation(s)
- Sarah E. O'Connor
- The John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom;
| |
Collapse
|
45
|
|
46
|
Control of litchi downy blight by zeamines produced by Dickeya zeae. Sci Rep 2015; 5:15719. [PMID: 26499339 PMCID: PMC4620559 DOI: 10.1038/srep15719] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 10/02/2015] [Indexed: 11/25/2022] Open
Abstract
Zeamines (ZMS), a class of polyamine-polyketide-nonribosomal peptide produced by bacterial isolate Dickeya zeae, were shown recently to be potent antibiotics against some bacterial pathogens. In this study, the results indicated that ZMS showed antifungal activity against Peronophythora litchii and other fungal pathogens. The activity of ZMS against the oomycete pathogen P. litchi, which causes the devastating litchi downy blight, was further investigated under in vitro and in vivo conditions. ZMS displayed potent inhibitory activity against the mycelial growth and sporangia germination of P. litchii. At a concentration of 2 μg/mL, about 99% of the sporangia germination was inhibited. Scanning electron microscopy and transmission electron microscopy analyses showed that treatment with ZMS could cause substantial damages to the oomycete endomembrane system. Furthermore, treatment of litchi fruits with ZMS solution significantly (P < 0.05) reduced the fruits decay and peel browning caused by P. litchii infection during storage at 28 °C. Taken together, our results provide useful clues on the antifungal mechanisms of ZMS, and highlight the promising potentials of ZMS as a fungicide, which in particular, may be useful for prevention and control of litchi fruits decay and browning caused by P. litchii infection during storage and transportation.
Collapse
|
47
|
Use of a biosynthetic intermediate to explore the chemical diversity of pseudo-natural fungal polyketides. Nat Chem 2015; 7:737-43. [DOI: 10.1038/nchem.2308] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/23/2015] [Indexed: 01/20/2023]
|
48
|
Sun H, Liu Z, Zhao H, Ang EL. Recent advances in combinatorial biosynthesis for drug discovery. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:823-33. [PMID: 25709407 PMCID: PMC4334309 DOI: 10.2147/dddt.s63023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Because of extraordinary structural diversity and broad biological activities, natural products have played a significant role in drug discovery. These therapeutically important secondary metabolites are assembled and modified by dedicated biosynthetic pathways in their host living organisms. Traditionally, chemists have attempted to synthesize natural product analogs that are important sources of new drugs. However, the extraordinary structural complexity of natural products sometimes makes it challenging for traditional chemical synthesis, which usually involves multiple steps, harsh conditions, toxic organic solvents, and byproduct wastes. In contrast, combinatorial biosynthesis exploits substrate promiscuity and employs engineered enzymes and pathways to produce novel “unnatural” natural products, substantially expanding the structural diversity of natural products with potential pharmaceutical value. Thus, combinatorial biosynthesis provides an environmentally friendly way to produce natural product analogs. Efficient expression of the combinatorial biosynthetic pathway in genetically tractable heterologous hosts can increase the titer of the compound, eventually resulting in less expensive drugs. In this review, we will discuss three major strategies for combinatorial biosynthesis: 1) precursor-directed biosynthesis; 2) enzyme-level modification, which includes swapping of the entire domains, modules and subunits, site-specific mutagenesis, and directed evolution; 3) pathway-level recombination. Recent examples of combinatorial biosynthesis employing these strategies will also be highlighted in this review.
Collapse
Affiliation(s)
- Huihua Sun
- Metabolic Engineering Research Laboratory, Institute of Chemical and Engineering Sciences, Agency for Science, Technology and Research, Singapore
| | - Zihe Liu
- Metabolic Engineering Research Laboratory, Institute of Chemical and Engineering Sciences, Agency for Science, Technology and Research, Singapore
| | - Huimin Zhao
- Metabolic Engineering Research Laboratory, Institute of Chemical and Engineering Sciences, Agency for Science, Technology and Research, Singapore ; Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ee Lui Ang
- Metabolic Engineering Research Laboratory, Institute of Chemical and Engineering Sciences, Agency for Science, Technology and Research, Singapore
| |
Collapse
|
49
|
Cacho RA, Tang Y, Chooi YH. Next-generation sequencing approach for connecting secondary metabolites to biosynthetic gene clusters in fungi. Front Microbiol 2015; 5:774. [PMID: 25642215 PMCID: PMC4294208 DOI: 10.3389/fmicb.2014.00774] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/17/2014] [Indexed: 12/20/2022] Open
Abstract
Genomics has revolutionized the research on fungal secondary metabolite (SM) biosynthesis. To elucidate the molecular and enzymatic mechanisms underlying the biosynthesis of a specific SM compound, the important first step is often to find the genes that responsible for its synthesis. The accessibility to fungal genome sequences allows the bypass of the cumbersome traditional library construction and screening approach. The advance in next-generation sequencing (NGS) technologies have further improved the speed and reduced the cost of microbial genome sequencing in the past few years, which has accelerated the research in this field. Here, we will present an example work flow for identifying the gene cluster encoding the biosynthesis of SMs of interest using an NGS approach. We will also review the different strategies that can be employed to pinpoint the targeted gene clusters rapidly by giving several examples stemming from our work.
Collapse
Affiliation(s)
- Ralph A Cacho
- Chemical and Biomolecular Engineering Department, University of California Los Angeles, Los Angeles, CA, USA
| | - Yi Tang
- Chemical and Biomolecular Engineering Department, University of California Los Angeles, Los Angeles, CA, USA ; Chemistry and Biochemistry Department, University of California Los Angeles, Los Angeles, CA, USA
| | - Yit-Heng Chooi
- Plant Sciences Division, Research School of Biology, The Australian National University Canberra, ACT, Australia
| |
Collapse
|
50
|
Fidan O, Zhan J. Recent advances in engineering yeast for pharmaceutical protein production. RSC Adv 2015. [DOI: 10.1039/c5ra13003d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Currently available systems and synthetic biology tools can be applied to yeast engineering for improved biopharmaceutical protein production.
Collapse
Affiliation(s)
- Ozkan Fidan
- Department of Biological Engineering
- Utah State University
- Logan
- USA
| | - Jixun Zhan
- Department of Biological Engineering
- Utah State University
- Logan
- USA
| |
Collapse
|