1
|
Brittin NJ, Anderson JM, Braun DR, Rajski SR, Currie CR, Bugni TS. Machine Learning-Based Bioactivity Classification of Natural Products Using LC-MS/MS Metabolomics. JOURNAL OF NATURAL PRODUCTS 2025; 88:361-372. [PMID: 39919314 DOI: 10.1021/acs.jnatprod.4c01123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
The rediscovery of known drug classes represents a major challenge in natural products drug discovery. Compound rediscovery inhibits the ability of researchers to explore novel natural products and wastes significant amounts of time and resources. This study introduces a novel machine learning framework that can effectively characterize the bioactivity of natural products by leveraging liquid chromatography tandem mass spectrometry and untargeted metabolomics analysis. This accelerates natural product drug discovery by addressing the challenge of dereplicating previously discovered bioactive compounds. Utilizing the SIRIUS 5 metabolomics software suite and in-silico-generated fragmentation spectra, we have trained a ML model capable of predicting a compound's drug class. This approach enables the rapid identification of bioactive scaffolds from LC-MS/MS data, even without reference experimental spectra. The model was trained on a diverse set of molecular fingerprints generated by SIRIUS 5 to effectively classify compounds based on their core pharmacophores. Our model robustly classified 21 diverse bioactive drug classes, achieving accuracies greater than 93% on experimental spectra. This study underscores the potential of ML combined with MFPs to dereplicate bioactive natural products based on pharmacophore, streamlining the discovery process and expediting improved methods of isolating novel antibacterial and antifungal agents.
Collapse
Affiliation(s)
- Nathaniel J Brittin
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Josephine M Anderson
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Doug R Braun
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Scott R Rajski
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Cameron R Currie
- Department of Biochemistry and Biomedical Sciences, M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Small Molecule Screening Facility, UW Carbone Cancer Center, Madison, Wisconsin 53792, United States
- Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
2
|
Romano KP, Bagnall J, Warrier T, Sullivan J, Ferrara K, Orzechowski M, Nguyen P, Raines K, Livny J, Shoresh N, Hung D. Perturbation-Specific Transcriptional Mapping for unbiased target elucidation of antibiotics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590978. [PMID: 38712067 PMCID: PMC11071498 DOI: 10.1101/2024.04.25.590978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The rising prevalence of antibiotic resistance threatens human health. While more sophisticated strategies for antibiotic discovery are being developed, target elucidation of new chemical entities remains challenging. In the post-genomic era, expression profiling can play an important role in mechanism-of-action (MOA) prediction by reporting on the cellular response to perturbation. However, the broad application of transcriptomics has yet to fulfill its promise of transforming target elucidation due to challenges in identifying the most relevant, direct responses to target inhibition. We developed an unbiased strategy for MOA prediction, called Perturbation-Specific Transcriptional Mapping (PerSpecTM), in which large-throughput expression profiling of wildtype or hypomorphic mutants, depleted for essential targets, enables a computational strategy to address this challenge. We applied PerSpecTM to perform reference-based MOA prediction based on the principle that similar perturbations, whether chemical or genetic, will elicit similar transcriptional responses. Using this approach, we elucidated the MOAs of three new molecules with activity against Pseudomonas aeruginosa by comparing their expression profiles to those of a reference set of antimicrobial compounds with known MOAs. We also show that transcriptional responses to small molecule inhibition resemble those resulting from genetic depletion of essential targets by CRISPRi by PerSpecTM, demonstrating proof-of-concept that correlations between expression profiles of small molecule and genetic perturbations can facilitate MOA prediction when no chemical entities exist to serve as a reference. Empowered by PerSpecTM, this work lays the foundation for an unbiased, readily scalable, systematic reference-based strategy for MOA elucidation that could transform antibiotic discovery efforts.
Collapse
|
3
|
Robbins N, Cowen LE. Antifungal discovery. Curr Opin Microbiol 2022; 69:102198. [PMID: 36037637 PMCID: PMC10726697 DOI: 10.1016/j.mib.2022.102198] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022]
Abstract
Fungi have a profound impact on human health, leading to billions of infections and millions of deaths worldwide each year. Exacerbating the public health burden is the continued emergence of drug-resistant fungal pathogens coupled with a dearth of treatment options to combat serious infections. Despite this health threat, scientific advances in chemistry, genetics, and biochemistry methodologies have enabled novel antifungal compounds to be discovered. Here, we describe current approaches for the discovery and characterization of novel antifungals, focusing on the identification of novel chemical matter and elucidation of the cellular target of bioactive compounds, followed by a review of the most promising emerging therapies in the antifungal-development pipeline.
Collapse
Affiliation(s)
- Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
4
|
Abstract
The last several decades have witnessed a surge in drug-resistant fungal infections that pose a serious threat to human health. While there is a limited arsenal of drugs that can be used to treat systemic infections, scientific advances have provided renewed optimism for the discovery of novel antifungals. The development of chemical-genomic assays using Saccharomyces cerevisiae has provided powerful methods to identify the mechanism of action of molecules in a living cell. Advances in molecular biology techniques have enabled complementary assays to be developed in fungal pathogens, including Candida albicans and Cryptococcus neoformans. These approaches enable the identification of target genes for drug candidates, as well as genes involved in buffering drug target pathways. Here, we examine yeast chemical-genomic assays and highlight how such resources can be utilized to predict the mechanisms of action of compounds, to study virulence attributes of diverse fungal pathogens, and to bolster the antifungal pipeline.
Collapse
Affiliation(s)
- Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada;
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada;
| |
Collapse
|
5
|
Fu C, Zhang X, Veri AO, Iyer KR, Lash E, Xue A, Yan H, Revie NM, Wong C, Lin ZY, Polvi EJ, Liston SD, VanderSluis B, Hou J, Yashiroda Y, Gingras AC, Boone C, O’Meara TR, O’Meara MJ, Noble S, Robbins N, Myers CL, Cowen LE. Leveraging machine learning essentiality predictions and chemogenomic interactions to identify antifungal targets. Nat Commun 2021; 12:6497. [PMID: 34764269 PMCID: PMC8586148 DOI: 10.1038/s41467-021-26850-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/22/2021] [Indexed: 02/08/2023] Open
Abstract
Fungal pathogens pose a global threat to human health, with Candida albicans among the leading killers. Systematic analysis of essential genes provides a powerful strategy to discover potential antifungal targets. Here, we build a machine learning model to generate genome-wide gene essentiality predictions for C. albicans and expand the largest functional genomics resource in this pathogen (the GRACE collection) by 866 genes. Using this model and chemogenomic analyses, we define the function of three uncharacterized essential genes with roles in kinetochore function, mitochondrial integrity, and translation, and identify the glutaminyl-tRNA synthetase Gln4 as the target of N-pyrimidinyl-β-thiophenylacrylamide (NP-BTA), an antifungal compound.
Collapse
Affiliation(s)
- Ci Fu
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Xiang Zhang
- grid.17635.360000000419368657Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Amanda O. Veri
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Kali R. Iyer
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Emma Lash
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Alice Xue
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Huijuan Yan
- grid.266102.10000 0001 2297 6811Department of Microbiology and Immunology, UCSF School of Medicine, San Francisco, CA 94143 USA
| | - Nicole M. Revie
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Cassandra Wong
- grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Zhen-Yuan Lin
- grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Elizabeth J. Polvi
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Sean D. Liston
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Benjamin VanderSluis
- grid.17635.360000000419368657Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Jing Hou
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada ,grid.17063.330000 0001 2157 2938Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Yoko Yashiroda
- grid.509461.fRIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198 Japan
| | - Anne-Claude Gingras
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Charles Boone
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada ,grid.17063.330000 0001 2157 2938Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1 Canada ,grid.509461.fRIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198 Japan
| | - Teresa R. O’Meara
- grid.214458.e0000000086837370Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
| | - Matthew J. O’Meara
- grid.214458.e0000000086837370Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109 USA
| | - Suzanne Noble
- grid.266102.10000 0001 2297 6811Department of Microbiology and Immunology, UCSF School of Medicine, San Francisco, CA 94143 USA
| | - Nicole Robbins
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Chad L. Myers
- grid.17635.360000000419368657Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Leah E. Cowen
- grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| |
Collapse
|
6
|
Safizadeh H, Simpkins SW, Nelson J, Li SC, Piotrowski JS, Yoshimura M, Yashiroda Y, Hirano H, Osada H, Yoshida M, Boone C, Myers CL. Improving Measures of Chemical Structural Similarity Using Machine Learning on Chemical-Genetic Interactions. J Chem Inf Model 2021; 61:4156-4172. [PMID: 34318674 PMCID: PMC8479812 DOI: 10.1021/acs.jcim.0c00993] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
A common strategy
for identifying molecules likely to possess a
desired biological activity is to search large databases of compounds
for high structural similarity to a query molecule that demonstrates
this activity, under the assumption that structural similarity is
predictive of similar biological activity. However, efforts to systematically
benchmark the diverse array of available molecular fingerprints and
similarity coefficients have been limited by a lack of large-scale
datasets that reflect biological similarities of compounds. To elucidate
the relative performance of these alternatives, we systematically
benchmarked 11 different molecular fingerprint encodings, each combined
with 13 different similarity coefficients, using a large set of chemical–genetic
interaction data from the yeast Saccharomyces cerevisiae as a systematic proxy for biological activity. We found that the
performance of different molecular fingerprints and similarity coefficients
varied substantially and that the all-shortest path fingerprints paired
with the Braun-Blanquet similarity coefficient provided superior performance
that was robust across several compound collections. We further proposed
a machine learning pipeline based on support vector machines that
offered a fivefold improvement relative to the best unsupervised approach.
Our results generally suggest that using high-dimensional chemical–genetic
data as a basis for refining molecular fingerprints can be a powerful
approach for improving prediction of biological functions from chemical
structures.
Collapse
Affiliation(s)
- Hamid Safizadeh
- Department of Electrical and Computer Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States.,Department of Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Scott W Simpkins
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Justin Nelson
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Sheena C Li
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada.,RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Jeff S Piotrowski
- RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Mami Yoshimura
- RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Yoko Yashiroda
- RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Hiroyuki Hirano
- RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Hiroyuki Osada
- RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Minoru Yoshida
- RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan.,Department of Biotechnology and Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Bunkyo City, Tokyo 113-8654, Japan
| | - Charles Boone
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada.,RIKEN Center for Sustainable Resource Science (CSRS), Wako, Saitama 351-0198, Japan
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States.,Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
7
|
Zhang F, Zhao M, Braun DR, Ericksen SS, Piotrowski JS, Nelson J, Peng J, Ananiev GE, Chanana S, Barns K, Fossen J, Sanchez H, Chevrette MG, Guzei IA, Zhao C, Guo L, Tang W, Currie CR, Rajski SR, Audhya A, Andes DR, Bugni TS. A marine microbiome antifungal targets urgent-threat drug-resistant fungi. Science 2020; 370:974-978. [PMID: 33214279 PMCID: PMC7756952 DOI: 10.1126/science.abd6919] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/29/2022]
Abstract
New antifungal drugs are urgently needed to address the emergence and transcontinental spread of fungal infectious diseases, such as pandrug-resistant Candida auris. Leveraging the microbiomes of marine animals and cutting-edge metabolomics and genomic tools, we identified encouraging lead antifungal molecules with in vivo efficacy. The most promising lead, turbinmicin, displays potent in vitro and mouse-model efficacy toward multiple-drug-resistant fungal pathogens, exhibits a wide safety index, and functions through a fungal-specific mode of action, targeting Sec14 of the vesicular trafficking pathway. The efficacy, safety, and mode of action distinct from other antifungal drugs make turbinmicin a highly promising antifungal drug lead to help address devastating global fungal pathogens such as C. auris.
Collapse
Affiliation(s)
- Fan Zhang
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Miao Zhao
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Doug R Braun
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Spencer S Ericksen
- Small Molecule Screening Facility, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | | | | | - Jian Peng
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Gene E Ananiev
- Small Molecule Screening Facility, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Shaurya Chanana
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Kenneth Barns
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Jen Fossen
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Hiram Sanchez
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Marc G Chevrette
- Department of Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery and Department of Plant Pathology, University of Wisconsin-Madison, Madison, WI, USA
| | - Ilia A Guzei
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Changgui Zhao
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Le Guo
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Weiping Tang
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Cameron R Currie
- Department of Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott R Rajski
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Anjon Audhya
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - David R Andes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
8
|
Duran-Frigola M, Pauls E, Guitart-Pla O, Bertoni M, Alcalde V, Amat D, Juan-Blanco T, Aloy P. Extending the small-molecule similarity principle to all levels of biology with the Chemical Checker. Nat Biotechnol 2020; 38:1087-1096. [PMID: 32440005 PMCID: PMC7616951 DOI: 10.1038/s41587-020-0502-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 03/27/2020] [Indexed: 02/07/2023]
Abstract
Small molecules are usually compared by their chemical structure, but there is no unified analytic framework for representing and comparing their biological activity. We present the Chemical Checker (CC), which provides processed, harmonized and integrated bioactivity data on ~800,000 small molecules. The CC divides data into five levels of increasing complexity, from the chemical properties of compounds to their clinical outcomes. In between, it includes targets, off-targets, networks and cell-level information, such as omics data, growth inhibition and morphology. Bioactivity data are expressed in a vector format, extending the concept of chemical similarity to similarity between bioactivity signatures. We show how CC signatures can aid drug discovery tasks, including target identification and library characterization. We also demonstrate the discovery of compounds that reverse and mimic biological signatures of disease models and genetic perturbations in cases that could not be addressed using chemical information alone. Overall, the CC signatures facilitate the conversion of bioactivity data to a format that is readily amenable to machine learning methods.
Collapse
Affiliation(s)
- Miquel Duran-Frigola
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Eduardo Pauls
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oriol Guitart-Pla
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Martino Bertoni
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Víctor Alcalde
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - David Amat
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Teresa Juan-Blanco
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
9
|
Xue A, Robbins N, Cowen LE. Advances in fungal chemical genomics for the discovery of new antifungal agents. Ann N Y Acad Sci 2020; 1496:5-22. [PMID: 32860238 DOI: 10.1111/nyas.14484] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/09/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022]
Abstract
Invasive fungal infections have escalated from a rare curiosity to a major cause of human mortality around the globe. This is in part due to a scarcity in the number of antifungal drugs available to combat mycotic disease, making the discovery of novel bioactive compounds and determining their mode of action of utmost importance. The development and application of chemical genomic assays using the model yeast Saccharomyces cerevisiae has provided powerful methods to identify the mechanism of action of diverse molecules in a living cell. Furthermore, complementary assays are continually being developed in fungal pathogens, most notably Candida albicans and Cryptococcus neoformans, to elucidate compound mechanism of action directly in the pathogen of interest. Collectively, the suite of chemical genetic assays that have been developed in multiple fungal species enables the identification of candidate drug target genes, as well as genes involved in buffering drug target pathways, and genes involved in general cellular responses to small molecules. In this review, we examine current yeast chemical genomic assays and highlight how such resources provide powerful tools that can be utilized to bolster the antifungal pipeline.
Collapse
Affiliation(s)
- Alice Xue
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Schramm VL, Meek TD. Enhanced Antibiotic Discovery by PROSPECTing. Biochemistry 2019; 58:3475-3476. [PMID: 31397555 DOI: 10.1021/acs.biochem.9b00616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Vern L Schramm
- Department of Biochemistry , Albert Einstein College of Medicine , 1300 Morris Park Avenue , Bronx , New York 10461 , United States
| | - Thomas D Meek
- Department of Biochemistry and Biophysics , Texas A&M University , College Station , Texas 77843 , United States
| |
Collapse
|
11
|
Large-scale chemical-genetics yields new M. tuberculosis inhibitor classes. Nature 2019; 571:72-78. [PMID: 31217586 DOI: 10.1038/s41586-019-1315-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/21/2019] [Indexed: 01/07/2023]
Abstract
New antibiotics are needed to combat rising levels of resistance, with new Mycobacterium tuberculosis (Mtb) drugs having the highest priority. However, conventional whole-cell and biochemical antibiotic screens have failed. Here we develop a strategy termed PROSPECT (primary screening of strains to prioritize expanded chemistry and targets), in which we screen compounds against pools of strains depleted of essential bacterial targets. We engineered strains that target 474 essential Mtb genes and screened pools of 100-150 strains against activity-enriched and unbiased compound libraries, probing more than 8.5 million chemical-genetic interactions. Primary screens identified over tenfold more hits than screening wild-type Mtb alone, with chemical-genetic interactions providing immediate, direct target insights. We identified over 40 compounds that target DNA gyrase, the cell wall, tryptophan, folate biosynthesis and RNA polymerase, as well as inhibitors that target EfpA. Chemical optimization yielded EfpA inhibitors with potent wild-type activity, thus demonstrating the ability of PROSPECT to yield inhibitors against targets that would have eluded conventional drug discovery.
Collapse
|
12
|
From Yeast to Humans: Leveraging New Approaches in Yeast to Accelerate Discovery of Therapeutic Targets for Synucleinopathies. Methods Mol Biol 2019; 2049:419-444. [PMID: 31602625 DOI: 10.1007/978-1-4939-9736-7_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neurodegenerative diseases (ND) represent a growing, global health crisis, one that lacks any disease-modifying therapeutic strategy. This critical need for new therapies must be met with an exhaustive approach to exploit all tools available. A yeast (Saccharomyces cerevisiae) model of α-synuclein toxicity-the protein causally linked to Parkinson's disease and other synucleinopathies-offers a powerful approach that takes advantage of the unique offerings of this system: tractable genetics, robust high-throughput screening strategies, unparalleled data repositories, powerful computational tools, and extensive evolutionary conservation of fundamental biological pathways. These attributes have enabled genetic and small molecule screens that have revealed toxic phenotypes and drug targets that translate directly to patient-derived iPSC neurons. Extending these insights, recent advances in genetic network analyses have generated the first "humanized" α-synuclein network, which has identified druggable proteins and led to validation of the toxic phenotypes in patient-derived cells. Unbiased phenotypic small molecule screens can identify compounds targeting critical proteins within α-synuclein networks. While identification of direct drug targets for phenotypic screen hits represents a bottleneck, high-throughput chemical genetic methods provide a means to uncover cellular targets and pathways for large numbers of compounds in parallel. Taken together, the yeast α-synuclein model and associated tools can reveal insights into underlying cellular pathologies, lead molecules and their cognate targets, and strategies to translate mechanisms of toxicity and cytoprotection into complex neuronal systems.
Collapse
|
13
|
Simpkins SW, Nelson J, Deshpande R, Li SC, Piotrowski JS, Wilson EH, Gebre AA, Safizadeh H, Okamoto R, Yoshimura M, Costanzo M, Yashiroda Y, Ohya Y, Osada H, Yoshida M, Boone C, Myers CL. Predicting bioprocess targets of chemical compounds through integration of chemical-genetic and genetic interactions. PLoS Comput Biol 2018; 14:e1006532. [PMID: 30376562 PMCID: PMC6226211 DOI: 10.1371/journal.pcbi.1006532] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 11/09/2018] [Accepted: 09/26/2018] [Indexed: 02/01/2023] Open
Abstract
Chemical-genetic interactions–observed when the treatment of mutant cells with chemical compounds reveals unexpected phenotypes–contain rich functional information linking compounds to their cellular modes of action. To systematically identify these interactions, an array of mutants is challenged with a compound and monitored for fitness defects, generating a chemical-genetic interaction profile that provides a quantitative, unbiased description of the cellular function(s) perturbed by the compound. Genetic interactions, obtained from genome-wide double-mutant screens, provide a key for interpreting the functional information contained in chemical-genetic interaction profiles. Despite the utility of this approach, integrative analyses of genetic and chemical-genetic interaction networks have not been systematically evaluated. We developed a method, called CG-TARGET (Chemical Genetic Translation via A Reference Genetic nETwork), that integrates large-scale chemical-genetic interaction screening data with a genetic interaction network to predict the biological processes perturbed by compounds. In a recent publication, we applied CG-TARGET to a screen of nearly 14,000 chemical compounds in Saccharomyces cerevisiae, integrating this dataset with the global S. cerevisiae genetic interaction network to prioritize over 1500 compounds with high-confidence biological process predictions for further study. We present here a formal description and rigorous benchmarking of the CG-TARGET method, showing that, compared to alternative enrichment-based approaches, it achieves similar or better accuracy while substantially improving the ability to control the false discovery rate of biological process predictions. Additional investigation of the compatibility of chemical-genetic and genetic interaction profiles revealed that one-third of observed chemical-genetic interactions contributed to the highest-confidence biological process predictions and that negative chemical-genetic interactions overwhelmingly formed the basis of these predictions. We also present experimental validations of CG-TARGET-predicted tubulin polymerization and cell cycle progression inhibitors. Our approach successfully demonstrates the use of genetic interaction networks in the high-throughput functional annotation of compounds to biological processes. Understanding how chemical compounds affect biological systems is of paramount importance as pharmaceutical companies strive to develop life-saving medicines, governments seek to regulate the safety of consumer products and agrichemicals, and basic scientists continue to study the fundamental inner workings of biological organisms. One powerful approach to characterize the effects of chemical compounds in living cells is chemical-genetic interaction screening. Using this approach, a collection of cells–each with a different defined genetic perturbation–is tested for sensitivity or resistance to the presence of a compound, resulting in a quantitative profile describing the functional effects of that compound on the cells. The work presented here describes our efforts to integrate compounds’ chemical-genetic interaction profiles with reference genetic interaction profiles containing information on gene function to predict the cellular processes perturbed by the compounds. We focused on specifically developing a method that could scale to perform these functional predictions for large collections of thousands of screened compounds and robustly control the false discovery rate. With chemical-genetic and genetic interaction screens now underway in multiple species including human cells, the method described here can be generally applied to enable the characterization of compounds’ effects across the tree of life.
Collapse
Affiliation(s)
- Scott W. Simpkins
- University of Minnesota-Twin Cities, Bioinformatics and Computational Biology Graduate Program, Minneapolis, Minnesota, United States of America
| | - Justin Nelson
- University of Minnesota-Twin Cities, Bioinformatics and Computational Biology Graduate Program, Minneapolis, Minnesota, United States of America
| | - Raamesh Deshpande
- University of Minnesota-Twin Cities, Department of Computer Science and Engineering, Minneapolis, Minnesota, United States of America
| | - Sheena C. Li
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | | | - Erin H. Wilson
- University of Minnesota-Twin Cities, Department of Computer Science and Engineering, Minneapolis, Minnesota, United States of America
| | - Abraham A. Gebre
- University of Tokyo, Department of Integrated Biosciences, Graduate School of Frontier Sciences, Kashiwa, Chiba, Japan
| | - Hamid Safizadeh
- University of Minnesota-Twin Cities, Department of Computer Science and Engineering, Minneapolis, Minnesota, United States of America
- University of Minnesota, Department of Electrical and Computer Engineering, Minneapolis, Minnesota, United States of America
| | - Reika Okamoto
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Mami Yoshimura
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Michael Costanzo
- University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Yoko Yashiroda
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Yoshikazu Ohya
- University of Tokyo, Department of Integrated Biosciences, Graduate School of Frontier Sciences, Kashiwa, Chiba, Japan
| | - Hiroyuki Osada
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Minoru Yoshida
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Charles Boone
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
- University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
- * E-mail: (CB); (CLM)
| | - Chad L. Myers
- University of Minnesota-Twin Cities, Bioinformatics and Computational Biology Graduate Program, Minneapolis, Minnesota, United States of America
- University of Minnesota-Twin Cities, Department of Computer Science and Engineering, Minneapolis, Minnesota, United States of America
- * E-mail: (CB); (CLM)
| |
Collapse
|
14
|
Patrick KL, Wojcechowskyj JA, Bell SL, Riba MN, Jing T, Talmage S, Xu P, Cabello AL, Xu J, Shales M, Jimenez-Morales D, Ficht TA, de Figueiredo P, Samuel JE, Li P, Krogan NJ, Watson RO. Quantitative Yeast Genetic Interaction Profiling of Bacterial Effector Proteins Uncovers a Role for the Human Retromer in Salmonella Infection. Cell Syst 2018; 7:323-338.e6. [PMID: 30077634 PMCID: PMC6160342 DOI: 10.1016/j.cels.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/21/2018] [Accepted: 06/21/2018] [Indexed: 11/26/2022]
Abstract
Intracellular bacterial pathogens secrete a repertoire of effector proteins into host cells that are required to hijack cellular pathways and cause disease. Despite decades of research, the molecular functions of most bacterial effectors remain unclear. To address this gap, we generated quantitative genetic interaction profiles between 36 validated and putative effectors from three evolutionarily divergent human bacterial pathogens and 4,190 yeast deletion strains. Correlating effector-generated profiles with those of yeast mutants, we recapitulated known biology for several effectors with remarkable specificity and predicted previously unknown functions for others. Biochemical and functional validation in human cells revealed a role for an uncharacterized component of the Salmonella SPI-2 translocon, SseC, in regulating maintenance of the Salmonella vacuole through interactions with components of the host retromer complex. These results exhibit the power of genetic interaction profiling to discover and dissect complex biology at the host-pathogen interface.
Collapse
Affiliation(s)
- Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Jason A Wojcechowskyj
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA; J. David Gladstone Institute, San Francisco, CA 94158, USA
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Morgan N Riba
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Tao Jing
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Sara Talmage
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Pengbiao Xu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Ana L Cabello
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA; Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, TX 77843, USA
| | - Jiewei Xu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA
| | - Michael Shales
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA
| | - David Jimenez-Morales
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA; J. David Gladstone Institute, San Francisco, CA 94158, USA
| | - Thomas A Ficht
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA; Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, TX 77843, USA
| | - James E Samuel
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA; J. David Gladstone Institute, San Francisco, CA 94158, USA.
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA.
| |
Collapse
|