1
|
Zhao Q, Wang L, Luo Y. Recent advances in natural products exploitation in Streptomyces via synthetic biology. Eng Life Sci 2019; 19:452-462. [PMID: 32625022 DOI: 10.1002/elsc.201800137] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 02/08/2019] [Accepted: 03/01/2019] [Indexed: 02/05/2023] Open
Abstract
Natural products of microbial origin have proven to be the wellspring of clinically useful compounds for human therapeutics. Streptomyces species are predominant sources of bioactive compounds, most of which serve as potential drug candidates. While the exploitation of natural products has been severely reduced over the past two decades, the growing crisis of evolution and dissemination of drug resistant pathogens have again attracted great interest in this field. The emerging synthetic biology has been heralded as a new bioengineering platform to discover novel bioactive compounds and expand bioactive natural products diversity and production. Herein, we review recent advances in the natural products exploitation of Streptomyces with the applications of synthetic biology from three major aspects, including recently developed synthetic biology tools, natural products biosynthetic pathway engineering strategies as well as chassis host modifications.
Collapse
Affiliation(s)
- Qiyuan Zhao
- Department of Gastroenterology Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center of Biotherapy Chengdu P. R. China
| | - Liping Wang
- Department of Gastroenterology Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center of Biotherapy Chengdu P. R. China
| | - Yunzi Luo
- Department of Gastroenterology Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center of Biotherapy Chengdu P. R. China
| |
Collapse
|
2
|
Wu H, Chu Z, Zhang W, Zhang C, Ni J, Fang H, Chen Y, Wang Y, Zhang L, Zhang B. Transcriptome-guided target identification of the TetR-like regulator SACE_5754 and engineered overproduction of erythromycin in Saccharopolyspora erythraea. J Biol Eng 2019; 13:11. [PMID: 30697347 PMCID: PMC6346578 DOI: 10.1186/s13036-018-0135-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 12/17/2018] [Indexed: 11/13/2022] Open
Abstract
Background Erythromycin A (Er-A) produced by the actinomycete Saccharopolyspora erythraea is an important antibiotic extensively used in human medicine. Dissecting of transcriptional regulators and their target genes associated with erythromycin biosynthesis is crucial to obtain erythromycin overproducer strains through engineering of relevant regulatory elements in S. erythraea. Results Here, we identified a TetR family transcriptional regulator (TFR), SACE_5754, negatively controlling erythromycin production. SACE_5754 indirectly repressed the transcription of ery cluster and cannot regulate itself and its adjacent gene SACE_5753. RNA-seq coupled with EMSAs and qRT-PCR was performed to identify the targets of SACE_5754, and confirmed that transcription of SACE_0388 (encoding a pyruvate, water diknase), SACE_3599 (encoding an antibiotic resistance macrolide glycosyltransferase) and SACE_6149 (encoding a FAD-binding monooxygenase) were directly repressed by SACE_5754. A consensus palindromic sequence TYMAGG-n2/n4/n11-KKTKRA (Y: C/T, M: A/C, K: T/G, R: A/G) was proved to be essential for SACE_5754 binding using DNase I footprinting and EMSAs. During the three target genes of SACE_5754, SACE_0388 and SACE_6149 exhibited the positive effect on erythromycin production. Overexpression of either SACE_0388 or SACE_6149 in ∆SACE_5754 further increased the Er-A production. By engineering the industrial strain S. erythraea WB with deletion of SACE_5754 combined with overexpression of either SACE_0388 or SACE_6149, Er-A production in WB∆SACE_5754/pIB139–0388 and WB∆SACE_5754/pIB139–6149 was successively increased by 42 and 30% compared to WB. Co-overexpression of SACE_0388 and SACE_6149 in WB∆SACE_5754 resulted in enhanced Er-A production by 64% relative to WB. In a 5-L fermenter, WB∆SACE_5754/pIB139–0388-6149 produced 4998 mg/L Er-A, a 48% increase over WB. Conclusion We have identified a TFR, SACE_5754, as a negative regulator of erythromycin biosynthesis, and engineering of SACE_5754 and its target genes, SACE_0388 and SACE_6149, resulted in enhanced erythromycin production in both wild-type and industrial S. erythraea strains. The strategy demonstrated here may be valuable to facilitate the manipulation of transcriptional regulators and their targets for production improvement of antibiotics in industrial actinomycetes. Electronic supplementary material The online version of this article (10.1186/s13036-018-0135-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hang Wu
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Zuling Chu
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Wanxiang Zhang
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Chi Zhang
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Jingshu Ni
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Heshi Fang
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Yuhong Chen
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Yansheng Wang
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| | - Lixin Zhang
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China.,2State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Buchang Zhang
- 1School of Life Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601 China
| |
Collapse
|
3
|
Wei J, He L, Niu G. Regulation of antibiotic biosynthesis in actinomycetes: Perspectives and challenges. Synth Syst Biotechnol 2018; 3:229-235. [PMID: 30417136 PMCID: PMC6215055 DOI: 10.1016/j.synbio.2018.10.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 02/08/2023] Open
Abstract
Actinomycetes are the main sources of antibiotics. The onset and level of production of each antibiotic is subject to complex control by multi-level regulators. These regulators exert their functions at hierarchical levels. At the lower level, cluster-situated regulators (CSRs) directly control the transcription of neighboring genes within the gene cluster. Higher-level pleiotropic and global regulators exert their functions mainly through modulating the transcription of CSRs. Advances in understanding of the regulation of antibiotic biosynthesis in actinomycetes have inspired us to engineer these regulators for strain improvement and antibiotic discovery.
Collapse
Affiliation(s)
- Junhong Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China
| | - Lang He
- Biotechnology Research Center, Southwest University, Chongqing, 400715, China.,Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
| | - Guoqing Niu
- Biotechnology Research Center, Southwest University, Chongqing, 400715, China.,Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
4
|
Kallifidas D, Jiang G, Ding Y, Luesch H. Rational engineering of Streptomyces albus J1074 for the overexpression of secondary metabolite gene clusters. Microb Cell Fact 2018; 17:25. [PMID: 29454348 PMCID: PMC5816538 DOI: 10.1186/s12934-018-0874-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/09/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genome sequencing revealed that Streptomyces sp. can dedicate up to ~ 10% of their genomes for the biosynthesis of bioactive secondary metabolites. However, the majority of these biosynthetic gene clusters are only weakly expressed or not at all. Indeed, the biosynthesis of natural products is highly regulated through integrating multiple nutritional and environmental signals perceived by pleiotropic and pathway-specific transcriptional regulators. Although pathway-specific refactoring has been a proved, productive approach for the activation of individual gene clusters, the construction of a global super host strain by targeting pleiotropic-specific genes for the expression of multiple diverse gene clusters is an attractive approach. RESULTS Streptomyces albus J1074 is a gifted heterologous host. To further improve its secondary metabolite expression capability, we rationally engineered the host by targeting genes affecting NADPH availability, precursor flux, cell growth and biosynthetic gene transcriptional activation. These studies led to the activation of the native paulomycin pathway in engineered S. albus strains and importantly the upregulated expression of the heterologous actinorhodin gene cluster. CONCLUSIONS Rational engineering of Streptomyces albus J1074 yielded a series of mutants with improved capabilities for native and heterologous expression of secondary metabolite gene clusters.
Collapse
Affiliation(s)
- Dimitris Kallifidas
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610 USA
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610 USA
| | - Guangde Jiang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610 USA
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610 USA
| | - Yousong Ding
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610 USA
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610 USA
| | - Hendrik Luesch
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610 USA
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610 USA
| |
Collapse
|
5
|
Ceapă CD, Vázquez-Hernández M, Rodríguez-Luna SD, Cruz Vázquez AP, Jiménez Suárez V, Rodríguez-Sanoja R, Alvarez-Buylla ER, Sánchez S. Genome mining of Streptomyces scabrisporus NF3 reveals symbiotic features including genes related to plant interactions. PLoS One 2018; 13:e0192618. [PMID: 29447216 PMCID: PMC5813959 DOI: 10.1371/journal.pone.0192618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/27/2018] [Indexed: 12/17/2022] Open
Abstract
Endophytic bacteria are wide-spread and associated with plant physiological benefits, yet their genomes and secondary metabolites remain largely unidentified. In this study, we explored the genome of the endophyte Streptomyces scabrisporus NF3 for discovery of potential novel molecules as well as genes and metabolites involved in host interactions. The complete genomes of seven Streptomyces and three other more distantly related bacteria were used to define the functional landscape of this unique microbe. The S. scabrisporus NF3 genome is larger than the average Streptomyces genome and not structured for an obligate endosymbiotic lifestyle; this and the fact that can grow in R2YE media implies that it could include a soil-living stage. The genome displays an enrichment of genes associated with amino acid production, protein secretion, secondary metabolite and antioxidants production and xenobiotic degradation, indicating that S. scabrisporus NF3 could contribute to the metabolic enrichment of soil microbial communities and of its hosts. Importantly, besides its metabolic advantages, the genome showed evidence for differential functional specificity and diversification of plant interaction molecules, including genes for the production of plant hormones, stress resistance molecules, chitinases, antibiotics and siderophores. Given the diversity of S. scabrisporus mechanisms for host upkeep, we propose that these strategies were necessary for its adaptation to plant hosts and to face changes in environmental conditions.
Collapse
Affiliation(s)
- Corina Diana Ceapă
- Departmento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Melissa Vázquez-Hernández
- Departmento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Stefany Daniela Rodríguez-Luna
- Departmento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Angélica Patricia Cruz Vázquez
- Departmento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- Instituto Tecnológico de Tuxtla Gutiérrez,Tuxtla, Gutiérrez, Chiapas, México
| | - Verónica Jiménez Suárez
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Romina Rodríguez-Sanoja
- Departmento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Elena R. Alvarez-Buylla
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Sergio Sánchez
- Departmento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| |
Collapse
|
6
|
Farha MA, Brown ED. Strategies for target identification of antimicrobial natural products. Nat Prod Rep 2016; 33:668-80. [DOI: 10.1039/c5np00127g] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite a pervasive decline in natural product research at many pharmaceutical companies over the last two decades, natural products have undeniably been a prolific and unsurpassed source for new lead antibacterial compounds.
Collapse
Affiliation(s)
- Maya A. Farha
- M.G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry and Biomedical Sciences
- McMaster University
- Hamilton
- Canada
| | - Eric D. Brown
- M.G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry and Biomedical Sciences
- McMaster University
- Hamilton
- Canada
| |
Collapse
|
7
|
Genetic manipulation of secondary metabolite biosynthesis for improved production in Streptomyces and other actinomycetes. J Ind Microbiol Biotechnol 2015; 43:343-70. [PMID: 26364200 DOI: 10.1007/s10295-015-1682-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 08/28/2015] [Indexed: 12/18/2022]
Abstract
Actinomycetes continue to be important sources for the discovery of secondary metabolites for applications in human medicine, animal health, and crop protection. With the maturation of actinomycete genome mining as a robust approach to identify new and novel cryptic secondary metabolite gene clusters, it is critical to continue developing methods to activate and enhance secondary metabolite biosynthesis for discovery, development, and large-scale manufacturing. This review covers recent reports on promising new approaches and further validations or technical improvements of existing approaches to strain improvement applicable to a wide range of Streptomyces species and other actinomycetes.
Collapse
|
8
|
Bekker V, Dodd A, Brady D, Rumbold K. Tools for metabolic engineering in Streptomyces. Bioengineered 2015; 5:293-9. [PMID: 25482230 DOI: 10.4161/bioe.29935] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
During the last few decades, Streptomycetes have shown to be a very important and adaptable group of bacteria for the production of various beneficial secondary metabolites. These secondary metabolites have been of great interest in academia and the pharmaceutical industries. To date, a vast variety of techniques and tools for metabolic engineering of relevant structural biosynthetic gene clusters have been developed. The main aim of this review is to summarize and discuss the published literature on tools for metabolic engineering of Streptomyces over the last decade. These strategies involve precursor engineering, structural and regulatory gene engineering, and the up or downregulation of genes, as well as genome shuffling and the use of genome scale metabolic models, which can reconstruct bacterial metabolic pathways to predict phenotypic changes and hence rationalize engineering strategies. These tools are continuously being developed to simplify the engineering strategies for this vital group of bacteria.
Collapse
Affiliation(s)
- Valerie Bekker
- a School of Molecular and Cell Biology; University of the Witwatersrand; Johannesburg, South Africa
| | | | | | | |
Collapse
|
9
|
Suryadevara CM, Gedeon PC, Sanchez-Perez L, Verla T, Alvarez-Breckenridge C, Choi BD, Fecci PE, Sampson JH. Are BiTEs the "missing link" in cancer therapy? Oncoimmunology 2015; 4:e1008339. [PMID: 26155413 DOI: 10.1080/2162402x.2015.1008339] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 02/06/2023] Open
Abstract
Conventional treatment for cancer routinely includes surgical resection and some combination of chemotherapy and radiation. These approaches are frequently accompanied by unintended and highly toxic collateral damage to healthy tissues, which are offset by only marginal prognostic improvements in patients with advanced cancers. This unfortunate balance has driven the development of novel therapies that aim to target tumors both safely and efficiently. Over the past decade, mounting evidence has supported the therapeutic utility of T-cell-centered cancer immunotherapy, which, in its various iterations, has been shown capable of eliciting highly precise and robust antitumor responses both in animal models and human trials. The identification of tumor-specific targets has further fueled a growing interest in T-cell therapies given their potential to circumvent the non-specific nature of traditional treatments. Of the several strategies geared toward achieving T-cell recognition of tumor, bispecific antibodies (bsAbs) represent a novel class of biologics that have garnered enthusiasm in recent years due to their versatility, specificity, safety, cost, and ease of production. Bispecific T-cell Engagers (BiTEs) are a subclass of bsAbs that are specific for CD3 on one arm and a tumor antigen on the second. As such, BiTEs function by recruiting and activating polyclonal populations of T-cells at tumor sites, and do so without the need for co-stimulation or conventional MHC recognition. Blinatumomab, a well-characterized BiTE, has emerged as a promising recombinant bscCD19×CD3 construct that has demonstrated remarkable antitumor activity in patients with B-cell malignancies. This clinical success has resulted in the rapid extension of BiTE technology against a greater repertoire of tumor antigens and the recent US Food and Drug Administration's (FDA) accelerated approval of blinatumomab for the treatment of a rare form of acute lymphoblastic leukemia (ALL). In this review, we dissect the role of T-cell therapeutics in the new era of cancer immunotherapy, appraise the value of CAR T-cells in the context of solid tumors, and discuss why the BiTE platform may rescue several of the apparent deficits and shortcomings of competing immunotherapies to support its widespread clinical application.
Collapse
Key Words
- ACT, adoptive cell therapy
- AICD, activation induced cell death
- ALL, acute lymphoblastic leukemia
- APC, antigen presenting cell
- BiTE, bispecific T-cell engager
- BsAb, bispecific antibody
- CAR, chimeric antigen receptors
- CHO, chinese hamster ovary
- CML, chronic myeloid leukemia
- GBM, glioblastoma
- MAb, monoclonal antibody
- MHC, major histocompatibility complex
- OS, overall survival
- ScFv, single chain variable fragment
- T lymphocytes
- TAA, tumor associated antigens
- TCR, T-cell receptor
- TIL, tumor infiltrating lymphocytes
- TREG, regulatory T-cells
- TSA, tumor specific antigens
- VV, vaccinia virus
- bispecific antibodies
- immunotherapy
- malignancies
Collapse
Affiliation(s)
- Carter M Suryadevara
- Duke Brain Tumor Immunotherapy Program; Division of Neurosurgery; Department of Surgery; Duke University Medical Center ; Durham, NC, USA ; Department of Pathology; Duke University Medical Center ; Durham, NC, USA ; The Preston Robert Tisch Brain Tumor Center; Duke University Medical Center ; Durham, NC, USA
| | - Patrick C Gedeon
- Duke Brain Tumor Immunotherapy Program; Division of Neurosurgery; Department of Surgery; Duke University Medical Center ; Durham, NC, USA ; The Preston Robert Tisch Brain Tumor Center; Duke University Medical Center ; Durham, NC, USA ; Department of Biomedical Engineering; Duke University ; Durham, NC, USA
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program; Division of Neurosurgery; Department of Surgery; Duke University Medical Center ; Durham, NC, USA ; The Preston Robert Tisch Brain Tumor Center; Duke University Medical Center ; Durham, NC, USA
| | - Terence Verla
- Duke Brain Tumor Immunotherapy Program; Division of Neurosurgery; Department of Surgery; Duke University Medical Center ; Durham, NC, USA ; The Preston Robert Tisch Brain Tumor Center; Duke University Medical Center ; Durham, NC, USA
| | | | - Bryan D Choi
- Department of Neurosurgery; Massachusetts General Hospital and Harvard Medical School ; Boston, MA, USA
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program; Division of Neurosurgery; Department of Surgery; Duke University Medical Center ; Durham, NC, USA ; The Preston Robert Tisch Brain Tumor Center; Duke University Medical Center ; Durham, NC, USA
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program; Division of Neurosurgery; Department of Surgery; Duke University Medical Center ; Durham, NC, USA ; Department of Pathology; Duke University Medical Center ; Durham, NC, USA ; The Preston Robert Tisch Brain Tumor Center; Duke University Medical Center ; Durham, NC, USA ; Department of Biomedical Engineering; Duke University ; Durham, NC, USA
| |
Collapse
|
10
|
Harvey AL, Edrada-Ebel R, Quinn RJ. The re-emergence of natural products for drug discovery in the genomics era. Nat Rev Drug Discov 2015; 14:111-29. [PMID: 25614221 DOI: 10.1038/nrd4510] [Citation(s) in RCA: 1565] [Impact Index Per Article: 156.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Natural products have been a rich source of compounds for drug discovery. However, their use has diminished in the past two decades, in part because of technical barriers to screening natural products in high-throughput assays against molecular targets. Here, we review strategies for natural product screening that harness the recent technical advances that have reduced these barriers. We also assess the use of genomic and metabolomic approaches to augment traditional methods of studying natural products, and highlight recent examples of natural products in antimicrobial drug discovery and as inhibitors of protein-protein interactions. The growing appreciation of functional assays and phenotypic screens may further contribute to a revival of interest in natural products for drug discovery.
Collapse
Affiliation(s)
- Alan L Harvey
- 1] Research and Innovation Support, Dublin City University, Dublin 9, Ireland. [2] Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0NR, UK
| | - RuAngelie Edrada-Ebel
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0NR, UK
| | - Ronald J Quinn
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| |
Collapse
|