1
|
Brittin NJ, Aceti DJ, Braun DR, Anderson JM, Ericksen SS, Rajski SR, Currie CR, Andes DR, Bugni TS. Dereplication of Natural Product Antifungals via Liquid Chromatography-Tandem Mass Spectrometry and Chemical Genomics. Molecules 2024; 30:77. [PMID: 39795134 PMCID: PMC11721837 DOI: 10.3390/molecules30010077] [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: 11/14/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025] Open
Abstract
Recently expanded reports of multidrug-resistant fungal infections underscore the need to develop new and more efficient methods for antifungal drug discovery. A ubiquitous problem in natural product drug discovery campaigns is the rediscovery of known compounds or their relatives; accordingly, we have integrated Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) for structural dereplication and Yeast Chemical Genomics for bioprocess evaluation into a screening platform to identify such compounds early in the screening process. We identified 450 fractions inhibiting Candida albicans and the resistant strains of C. auris and C. glabrata among more than 40,000 natural product fractions. LC-MS/MS and chemical genomics were then used to identify those with known chemistry and mechanisms of action. The parallel deployment of these orthogonal methods improved the detection of unwanted compound classes over the methods applied individually.
Collapse
Affiliation(s)
- Nathaniel J. Brittin
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA; (N.J.B.); (D.R.B.); (J.M.A.); (S.R.R.)
- Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J. Aceti
- Small Molecule Screening Facility, UW Carbone Cancer Center, Madison, WI 53792, USA; (D.J.A.); (S.S.E.)
| | - Doug R. Braun
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA; (N.J.B.); (D.R.B.); (J.M.A.); (S.R.R.)
| | - Josephine M. Anderson
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA; (N.J.B.); (D.R.B.); (J.M.A.); (S.R.R.)
| | - Spencer S. Ericksen
- Small Molecule Screening Facility, UW Carbone Cancer Center, Madison, WI 53792, USA; (D.J.A.); (S.S.E.)
| | - Scott R. Rajski
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA; (N.J.B.); (D.R.B.); (J.M.A.); (S.R.R.)
| | - 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, ON L8S 4L8, Canada;
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David R. Andes
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA;
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
| | - Tim S. Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA; (N.J.B.); (D.R.B.); (J.M.A.); (S.R.R.)
- Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, WI 53705, USA
- Small Molecule Screening Facility, UW Carbone Cancer Center, Madison, WI 53792, USA; (D.J.A.); (S.S.E.)
| |
Collapse
|
2
|
Forster DT, Li SC, Yashiroda Y, Yoshimura M, Li Z, Isuhuaylas LAV, Itto-Nakama K, Yamanaka D, Ohya Y, Osada H, Wang B, Bader GD, Boone C. BIONIC: biological network integration using convolutions. Nat Methods 2022; 19:1250-1261. [PMID: 36192463 PMCID: PMC11236286 DOI: 10.1038/s41592-022-01616-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 08/16/2022] [Indexed: 01/21/2023]
Abstract
Biological networks constructed from varied data can be used to map cellular function, but each data type has limitations. Network integration promises to address these limitations by combining and automatically weighting input information to obtain a more accurate and comprehensive representation of the underlying biology. We developed a deep learning-based network integration algorithm that incorporates a graph convolutional network framework. Our method, BIONIC (Biological Network Integration using Convolutions), learns features that contain substantially more functional information compared to existing approaches. BIONIC has unsupervised and semisupervised learning modes, making use of available gene function annotations. BIONIC is scalable in both size and quantity of the input networks, making it feasible to integrate numerous networks on the scale of the human genome. To demonstrate the use of BIONIC in identifying new biology, we predicted and experimentally validated essential gene chemical-genetic interactions from nonessential gene profiles in yeast.
Collapse
Affiliation(s)
- Duncan T Forster
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada
| | - Sheena C Li
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Yoko Yashiroda
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Mami Yoshimura
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Zhijian Li
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Kaori Itto-Nakama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Daisuke Yamanaka
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Osada
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Bo Wang
- Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada.
- Peter Munk Cardiac Center, University Health Network, Toronto, Ontario, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.
- The Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan.
| |
Collapse
|
3
|
Hogan AM, Cardona ST. Gradients in gene essentiality reshape antibacterial research. FEMS Microbiol Rev 2022; 46:fuac005. [PMID: 35104846 PMCID: PMC9075587 DOI: 10.1093/femsre/fuac005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 02/03/2023] Open
Abstract
Essential genes encode the processes that are necessary for life. Until recently, commonly applied binary classifications left no space between essential and non-essential genes. In this review, we frame bacterial gene essentiality in the context of genetic networks. We explore how the quantitative properties of gene essentiality are influenced by the nature of the encoded process, environmental conditions and genetic background, including a strain's distinct evolutionary history. The covered topics have important consequences for antibacterials, which inhibit essential processes. We argue that the quantitative properties of essentiality can thus be used to prioritize antibacterial cellular targets and desired spectrum of activity in specific infection settings. We summarize our points with a case study on the core essential genome of the cystic fibrosis pathobiome and highlight avenues for targeted antibacterial development.
Collapse
Affiliation(s)
- Andrew M Hogan
- Department of Microbiology, University of Manitoba, 45 Chancellor's Circle, Winnipeg, Manitoba R3T 2N2, Canada
| | - Silvia T Cardona
- Department of Microbiology, University of Manitoba, 45 Chancellor's Circle, Winnipeg, Manitoba R3T 2N2, Canada
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Room 543 - 745 Bannatyne Avenue, Winnipeg, Manitoba, R3E 0J9, Canada
| |
Collapse
|
4
|
Ward HN, Aregger M, Gonatopoulos-Pournatzis T, Billmann M, Ohsumi TK, Brown KR, Blencowe BJ, Moffat J, Myers CL. Analysis of combinatorial CRISPR screens with the Orthrus scoring pipeline. Nat Protoc 2021; 16:4766-4798. [PMID: 34508259 PMCID: PMC9084619 DOI: 10.1038/s41596-021-00596-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 06/03/2021] [Indexed: 02/08/2023]
Abstract
The continued improvement of combinatorial CRISPR screening platforms necessitates the development of new computational pipelines for scoring combinatorial screening data. Unlike for single-guide RNA (sgRNA) pooled screening platforms, combinatorial scoring for multiplexed systems is confounded by guide design parameters such as the number of gRNAs per construct, the position of gRNAs along constructs, and additional features that may impact gRNA expression, processing or capture. In this protocol we describe Orthrus, an R package for processing, scoring and analyzing combinatorial CRISPR screening data that addresses these challenges. This protocol walks through the application of Orthrus to previously published combinatorial screening data from the CHyMErA experimental system, a platform we recently developed that pairs Cas9 with Cas12a gRNAs and enables programmed targeting of multiple genomic sites. We demonstrate Orthrus' features for screen quality assessment and two distinct scoring modes for dual guide RNAs (dgRNAs) that target the same gene twice or dgRNAs that target two different genes. Running Orthrus requires basic R programming experience, ~5-10 min of computational time and 15-60 min total.
Collapse
Affiliation(s)
- Henry N Ward
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Michael Aregger
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- RNA Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Thomas Gonatopoulos-Pournatzis
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- RNA Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Maximilian Billmann
- Department of Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | | | - Kevin R Brown
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin J Blencowe
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jason Moffat
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Chad L Myers
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, MN, USA.
- Department of Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Clionamines stimulate autophagy, inhibit Mycobacterium tuberculosis survival in macrophages, and target Pik1. Cell Chem Biol 2021; 29:870-882.e11. [PMID: 34520745 DOI: 10.1016/j.chembiol.2021.07.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/16/2021] [Accepted: 07/21/2021] [Indexed: 12/25/2022]
Abstract
The pathogen Mycobacterium tuberculosis (Mtb) evades the innate immune system by interfering with autophagy and phagosomal maturation in macrophages, and, as a result, small molecule stimulation of autophagy represents a host-directed therapeutics (HDTs) approach for treatment of tuberculosis (TB). Here we show the marine natural product clionamines activate autophagy and inhibit Mtb survival in macrophages. A yeast chemical-genetics approach identified Pik1 as target protein of the clionamines. Biotinylated clionamine B pulled down Pik1 from yeast cell lysates and a clionamine analog inhibited phosphatidyl 4-phosphate (PI4P) production in yeast Golgi membranes. Chemical-genetic profiles of clionamines and cationic amphiphilic drugs (CADs) are closely related, linking the clionamine mode of action to co-localization with PI4P in a vesicular compartment. Small interfering RNA (siRNA) knockdown of PI4KB, a human homolog of Pik1, inhibited the survival of Mtb in macrophages, identifying PI4KB as an unexploited molecular target for efforts to develop HDT drugs for treatment of TB.
Collapse
|
6
|
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
|
7
|
Richards R, Schwartz HR, Honeywell ME, Stewart MS, Cruz-Gordillo P, Joyce AJ, Landry BD, Lee MJ. Drug antagonism and single-agent dominance result from differences in death kinetics. Nat Chem Biol 2020; 16:791-800. [PMID: 32251407 PMCID: PMC7311243 DOI: 10.1038/s41589-020-0510-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Cancer treatment generally involves drugs used in combinations. Most previous work has focused on identifying and understanding synergistic drug-drug interactions; however, understanding antagonistic interactions remains an important and understudied issue. To enrich for antagonism and reveal common features of these combinations, we screened all pairwise combinations of drugs characterized as activators of regulated cell death. This network is strongly enriched for antagonism, particularly a form of antagonism that we call 'single-agent dominance'. Single-agent dominance refers to antagonisms in which a two-drug combination phenocopies one of the two agents. Dominance results from differences in cell death onset time, with dominant drugs acting earlier than their suppressed counterparts. We explored mechanisms by which parthanatotic agents dominate apoptotic agents, finding that dominance in this scenario is caused by mutually exclusive and conflicting use of Poly(ADP-ribose) polymerase 1 (PARP1). Taken together, our study reveals death kinetics as a predictive feature of antagonism, due to inhibitory crosstalk between cell death pathways.
Collapse
Affiliation(s)
- Ryan Richards
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Hannah R Schwartz
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Megan E Honeywell
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Mariah S Stewart
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Peter Cruz-Gordillo
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Anna J Joyce
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Benjamin D Landry
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael J Lee
- Program in Systems Biology (PSB), University of Massachusetts Medical School, Worcester, MA, USA.
- Program in Molecular Medicine (PMM), University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Molecular, Cell and Cancer Biology (MCCB), University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
8
|
Morgens DW, Chan C, Kane AJ, Weir NR, Li A, Dubreuil MM, Tsui CK, Hess GT, Lavertu A, Han K, Polyakov N, Zhou J, Handy EL, Alabi P, Dombroski A, Yao D, Altman RB, Sello JK, Denic V, Bassik MC. Retro-2 protects cells from ricin toxicity by inhibiting ASNA1-mediated ER targeting and insertion of tail-anchored proteins. eLife 2019; 8:48434. [PMID: 31674906 PMCID: PMC6858068 DOI: 10.7554/elife.48434] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
The small molecule Retro-2 prevents ricin toxicity through a poorly-defined mechanism of action (MOA), which involves halting retrograde vesicle transport to the endoplasmic reticulum (ER). CRISPRi genetic interaction analysis revealed Retro-2 activity resembles disruption of the transmembrane domain recognition complex (TRC) pathway, which mediates post-translational ER-targeting and insertion of tail-anchored (TA) proteins, including SNAREs required for retrograde transport. Cell-based and in vitro assays show that Retro-2 blocks delivery of newly-synthesized TA-proteins to the ER-targeting factor ASNA1 (TRC40). An ASNA1 point mutant identified using CRISPR-mediated mutagenesis abolishes both the cytoprotective effect of Retro-2 against ricin and its inhibitory effect on ASNA1-mediated ER-targeting. Together, our work explains how Retro-2 prevents retrograde trafficking of toxins by inhibiting TA-protein targeting, describes a general CRISPR strategy for predicting the MOA of small molecules, and paves the way for drugging the TRC pathway to treat broad classes of viruses known to be inhibited by Retro-2.
Collapse
Affiliation(s)
- David W Morgens
- Department of Genetics, Stanford University, Stanford, United States
| | - Charlene Chan
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Andrew J Kane
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Nicholas R Weir
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Amy Li
- Department of Genetics, Stanford University, Stanford, United States
| | | | - C Kimberly Tsui
- Department of Genetics, Stanford University, Stanford, United States
| | - Gaelen T Hess
- Department of Genetics, Stanford University, Stanford, United States
| | - Adam Lavertu
- Biomedical Informatics Training Program, Stanford University, Stanford, United States
| | - Kyuho Han
- Department of Genetics, Stanford University, Stanford, United States
| | - Nicole Polyakov
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Jing Zhou
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Emma L Handy
- Department of Chemistry, Brown University, Providence, United States
| | - Philip Alabi
- Department of Chemistry, Brown University, Providence, United States
| | - Amanda Dombroski
- Department of Chemistry, Brown University, Providence, United States
| | - David Yao
- Department of Genetics, Stanford University, Stanford, United States
| | - Russ B Altman
- Department of Genetics, Stanford University, Stanford, United States.,Bioengineering, Stanford University, Stanford, United States
| | - Jason K Sello
- Department of Chemistry, Brown University, Providence, United States
| | - Vladimir Denic
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Michael C Bassik
- Department of Genetics, Stanford University, Stanford, United States.,Program in Cancer Biology, Stanford University, Stanford, United States.,Stanford University Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford, United States
| |
Collapse
|
9
|
Zhou FL, Li SC, Zhu Y, Guo WJ, Shao LJ, Nelson J, Simpkins S, Yang DH, Liu Q, Yashiroda Y, Xu JB, Fan YY, Yue JM, Yoshida M, Xia T, Myers CL, Boone C, Wang MW. Integrating yeast chemical genomics and mammalian cell pathway analysis. Acta Pharmacol Sin 2019; 40:1245-1255. [PMID: 31138898 PMCID: PMC6786357 DOI: 10.1038/s41401-019-0231-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/14/2019] [Indexed: 12/27/2022]
Abstract
Chemical genomics has been applied extensively to evaluate small molecules that modulate biological processes in Saccharomyces cerevisiae. Here, we use yeast as a surrogate system for studying compounds that are active against metazoan targets. Large-scale chemical-genetic profiling of thousands of synthetic and natural compounds from the Chinese National Compound Library identified those with high-confidence bioprocess target predictions. To discover compounds that have the potential to function like therapeutic agents with known targets, we also analyzed a reference library of approved drugs. Previously uncharacterized compounds with chemical-genetic profiles resembling existing drugs that modulate autophagy and Wnt/β-catenin signal transduction were further examined in mammalian cells, and new modulators with specific modes of action were validated. This analysis exploits yeast as a general platform for predicting compound bioactivity in mammalian cells.
Collapse
Affiliation(s)
- Fu-Lai Zhou
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sheena C Li
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, 3510198, Japan
| | - Yue Zhu
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wan-Jing Guo
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li-Jun Shao
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Justin Nelson
- Bioinformatics and Computational Biology Program, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455, USA
| | - Scott Simpkins
- Bioinformatics and Computational Biology Program, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455, USA
| | - De-Hua Yang
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
| | - Qing Liu
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
| | - Yoko Yashiroda
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, 3510198, Japan
| | - Jin-Biao Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yao-Yue Fan
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jian-Min Yue
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Minoru Yoshida
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, 3510198, Japan
- Department of Biology, The University of Tokyo, Bunkyo-ku, Tokyo, 1138657, Japan
- Collaborative Research for Innovative Microbiology, The University of Tokyo, Bunkyo-ku, Tokyo, 1138657, Japan
| | - Tian Xia
- Department of Electronics and Information Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chad L Myers
- Bioinformatics and Computational Biology Program, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455, USA.
| | - Charles Boone
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, 3510198, Japan.
- Donnelly Centre and Department of Molecular Genetics, University of Toronto, Ontario, M5S 3E1, Canada.
| | - Ming-Wei Wang
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
10
|
Simpkins SW, Deshpande R, Nelson J, Li SC, Piotrowski JS, Ward HN, Yashiroda Y, Osada H, Yoshida M, Boone C, Myers CL. Using BEAN-counter to quantify genetic interactions from multiplexed barcode sequencing experiments. Nat Protoc 2019; 14:415-440. [PMID: 30635653 DOI: 10.1038/s41596-018-0099-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The construction of genome-wide mutant collections has enabled high-throughput, high-dimensional quantitative characterization of gene and chemical function, particularly via genetic and chemical-genetic interaction experiments. As the throughput of such experiments increases with improvements in sequencing technology and sample multiplexing, appropriate tools must be developed to handle the large volume of data produced. Here, we describe how to apply our approach to high-throughput, fitness-based profiling of pooled mutant yeast collections using the BEAN-counter software pipeline (Barcoded Experiment Analysis for Next-generation sequencing) for analysis. The software has also successfully processed data from Schizosaccharomyces pombe, Escherichia coli, and Zymomonas mobilis mutant collections. We provide general recommendations for the design of large-scale, multiplexed barcode sequencing experiments. The procedure outlined here was used to score interactions for ~4 million chemical-by-mutant combinations in our recently published chemical-genetic interaction screen of nearly 14,000 chemical compounds across seven diverse compound collections. Here we selected a representative subset of these data on which to demonstrate our analysis pipeline. BEAN-counter is open source, written in Python, and freely available for academic use. Users should be proficient at the command line; advanced users who wish to analyze larger datasets with hundreds or more conditions should also be familiar with concepts in analysis of high-throughput biological data. BEAN-counter encapsulates the knowledge we have accumulated from, and successfully applied to, our multiplexed, pooled barcode sequencing experiments. This protocol will be useful to those interested in generating their own high-dimensional, quantitative characterizations of gene or chemical function in a high-throughput manner.
Collapse
Affiliation(s)
- Scott W Simpkins
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Raamesh Deshpande
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Justin Nelson
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Sheena C Li
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Jeff S Piotrowski
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan.,Yumanity Therapeutics, Cambridge, MA, USA
| | - Henry Neil Ward
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Yoko Yashiroda
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, 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.,Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Chad L Myers
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Twin Cities, Minneapolis, MN, USA. .,Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
11
|
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
|