1
|
Geck RC, Moresi NG, Anderson LM, Brewer R, Renz TR, Taylor MB, Dunham MJ. Experimental evolution of Saccharomyces cerevisiae for caffeine tolerance alters multidrug resistance and target of rapamycin signaling pathways. G3 (BETHESDA, MD.) 2024; 14:jkae148. [PMID: 38989875 PMCID: PMC11373655 DOI: 10.1093/g3journal/jkae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
Caffeine is a natural compound that inhibits the major cellular signaling regulator target of rapamycin (TOR), leading to widespread effects including growth inhibition. Saccharomyces cerevisiae yeast can adapt to tolerate high concentrations of caffeine in coffee and cacao fermentations and in experimental systems. While many factors affecting caffeine tolerance and TOR signaling have been identified, further characterization of their interactions and regulation remain to be studied. We used experimental evolution of S. cerevisiae to study the genetic contributions to caffeine tolerance in yeast, through a collaboration between high school students evolving yeast populations coupled with further research exploration in university labs. We identified multiple evolved yeast populations with mutations in PDR1 and PDR5, which contribute to multidrug resistance, and showed that gain-of-function mutations in multidrug resistance family transcription factors Pdr1, Pdr3, and Yrr1 differentially contribute to caffeine tolerance. We also identified loss-of-function mutations in TOR effectors Sit4, Sky1, and Tip41 and showed that these mutations contribute to caffeine tolerance. These findings support the importance of both the multidrug resistance family and TOR signaling in caffeine tolerance and can inform future exploration of networks affected by caffeine and other TOR inhibitors in model systems and industrial applications.
Collapse
Affiliation(s)
- Renee C Geck
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Naomi G Moresi
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Leah M Anderson
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | | | | | | | - Maitreya J Dunham
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2
|
Geck RC, Moresi NG, Anderson LM, Brewer R, Renz TR, Taylor MB, Dunham MJ. Experimental evolution of S. cerevisiae for caffeine tolerance alters multidrug resistance and TOR signaling pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.28.591555. [PMID: 38746122 PMCID: PMC11092465 DOI: 10.1101/2024.04.28.591555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Caffeine is a natural compound that inhibits the major cellular signaling regulator TOR, leading to widespread effects including growth inhibition. S. cerevisiae yeast can adapt to tolerate high concentrations of caffeine in coffee and cacao fermentations and in experimental systems. While many factors affecting caffeine tolerance and TOR signaling have been identified, further characterization of their interactions and regulation remain to be studied. We used experimental evolution of S. cerevisiae to study the genetic contributions to caffeine tolerance in yeast, through a collaboration between high school students evolving yeast populations coupled with further research exploration in university labs. We identified multiple evolved yeast populations with mutations in PDR1 and PDR5, which contribute to multidrug resistance, and showed that gain-of-function mutations in multidrug resistance family transcription factors PDR1, PDR3, and YRR1 differentially contribute to caffeine tolerance. We also identified loss-of-function mutations in TOR effectors SIT4, SKY1, and TIP41, and show that these mutations contribute to caffeine tolerance. These findings support the importance of both the multidrug resistance family and TOR signaling in caffeine tolerance, and can inform future exploration of networks affected by caffeine and other TOR inhibitors in model systems and industrial applications.
Collapse
Affiliation(s)
- Renee C Geck
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Naomi G Moresi
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Leah M Anderson
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | | | | | - M Bryce Taylor
- Program in Biology, Loras College, Dubuque, IA 52001, USA
| | - Maitreya J Dunham
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
3
|
Parmar S, Gonzalez SJ, Heckel JM, Mukherjee S, McClellan M, Clarke DJ, Johansson M, Tank D, Geisness A, Wood DK, Gardner MK. Robust microtubule dynamics facilitate low-tension kinetochore detachment in metaphase. J Cell Biol 2023; 222:e202202085. [PMID: 37166419 PMCID: PMC10182774 DOI: 10.1083/jcb.202202085] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/07/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023] Open
Abstract
During mitosis, sister chromatids are stretched apart at their centromeres via their attachment to oppositely oriented kinetochore microtubules. This stretching generates inwardly directed tension across the separated sister centromeres. The cell leverages this tension signal to detect and then correct potential errors in chromosome segregation, via a mechanical tension signaling pathway that detaches improperly attached kinetochores from their microtubules. However, the sequence of events leading up to these detachment events remains unknown. In this study, we used microfluidics to sustain and observe low-tension budding yeast metaphase spindles over multiple hours, allowing us to elucidate the tension history prior to a detachment event. We found that, under conditions in which kinetochore phosphorylation weakens low-tension kinetochore-microtubule connections, the mechanical forces produced via the dynamic growth and shortening of microtubules is required to efficiently facilitate detachment events. Our findings underscore the critical role of robust kinetochore microtubule dynamics in ensuring the fidelity of chromosome segregation during mitosis.
Collapse
Affiliation(s)
- Sneha Parmar
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Samuel J. Gonzalez
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Julia M. Heckel
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Soumya Mukherjee
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Mark McClellan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Duncan J. Clarke
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Marnie Johansson
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Damien Tank
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Athena Geisness
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Melissa K. Gardner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
4
|
Mol GS, Aruldhas D, Joe IH. Chemical reactivity, molecular electrostatic potential and in-silico analysis on benzimidazole fungicide benomyl. Heliyon 2022; 8:e11417. [DOI: 10.1016/j.heliyon.2022.e11417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/27/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022] Open
|
5
|
Jiang S, Chen Y, Han S, Lv L, Li L. Next-Generation Sequencing Applications for the Study of Fungal Pathogens. Microorganisms 2022; 10:microorganisms10101882. [PMID: 36296159 PMCID: PMC9609632 DOI: 10.3390/microorganisms10101882] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Next-generation sequencing (NGS) has become a widely used technology in biological research. NGS applications for clinical pathogen detection have become vital technologies. It is increasingly common to perform fast, accurate, and specific detection of clinical specimens using NGS. Pathogenic fungi with high virulence and drug resistance cause life-threatening clinical infections. NGS has had a significant biotechnological impact on detecting bacteria and viruses but is not equally applicable to fungi. There is a particularly urgent clinical need to use NGS to help identify fungi causing infections and prevent negative impacts. This review summarizes current research on NGS applications for fungi and offers a visual method of fungal detection. With the development of NGS and solutions for overcoming sequencing limitations, we suggest clinicians test specimens as soon as possible when encountering infections of unknown cause, suspected infections in vital organs, or rapidly progressive disease.
Collapse
Affiliation(s)
- Shiman Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
- Correspondence: ; Tel.: +86-0571-8723-6458
| |
Collapse
|
6
|
Ottilie S, Luth MR, Hellemann E, Goldgof GM, Vigil E, Kumar P, Cheung AL, Song M, Godinez-Macias KP, Carolino K, Yang J, Lopez G, Abraham M, Tarsio M, LeBlanc E, Whitesell L, Schenken J, Gunawan F, Patel R, Smith J, Love MS, Williams RM, McNamara CW, Gerwick WH, Ideker T, Suzuki Y, Wirth DF, Lukens AK, Kane PM, Cowen LE, Durrant JD, Winzeler EA. Adaptive laboratory evolution in S. cerevisiae highlights role of transcription factors in fungal xenobiotic resistance. Commun Biol 2022; 5:128. [PMID: 35149760 PMCID: PMC8837787 DOI: 10.1038/s42003-022-03076-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/21/2022] [Indexed: 12/24/2022] Open
Abstract
In vitro evolution and whole genome analysis were used to comprehensively identify the genetic determinants of chemical resistance in Saccharomyces cerevisiae. Sequence analysis identified many genes contributing to the resistance phenotype as well as numerous amino acids in potential targets that may play a role in compound binding. Our work shows that compound-target pairs can be conserved across multiple species. The set of 25 most frequently mutated genes was enriched for transcription factors, and for almost 25 percent of the compounds, resistance was mediated by one of 100 independently derived, gain-of-function SNVs found in a 170 amino acid domain in the two Zn2C6 transcription factors YRR1 and YRM1 (p < 1 × 10−100). This remarkable enrichment for transcription factors as drug resistance genes highlights their important role in the evolution of antifungal xenobiotic resistance and underscores the challenge to develop antifungal treatments that maintain potency. Ottilie et al. employ an experimental evolution approach to investigate the role of transcription factors in yeast chemical resistance. Most emergent mutations in resistant strains were enriched in transcription factor coding genes, highlighting their importance in drug resistance.
Collapse
Affiliation(s)
- Sabine Ottilie
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Madeline R Luth
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Erich Hellemann
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Gregory M Goldgof
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Eddy Vigil
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Prianka Kumar
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Andrea L Cheung
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Miranda Song
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Karla P Godinez-Macias
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Krypton Carolino
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Jennifer Yang
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Gisel Lopez
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Matthew Abraham
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Maureen Tarsio
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, NY, 13210, USA
| | - Emmanuelle LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Jake Schenken
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Felicia Gunawan
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Reysha Patel
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Joshua Smith
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Melissa S Love
- Calibr, a division of The Scripps Research Institutes, La Jolla, CA, 92037, USA
| | - Roy M Williams
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA.,Aspen Neuroscience, San Diego, CA, 92121, USA
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institutes, La Jolla, CA, 92037, USA
| | - William H Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, La Jolla, CA, 92037, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yo Suzuki
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, La Jolla, CA, 92037, USA
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Amanda K Lukens
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, NY, 13210, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA.
| |
Collapse
|
7
|
Schuyler SC, Chen HY. Using Budding Yeast to Identify Molecules That Block Cancer Cell 'Mitotic Slippage' Only in the Presence of Mitotic Poisons. Int J Mol Sci 2021; 22:ijms22157985. [PMID: 34360748 PMCID: PMC8347345 DOI: 10.3390/ijms22157985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022] Open
Abstract
Research on the budding yeast Saccharomyces cerevisiae has yielded fundamental discoveries on highly conserved biological pathways and yeast remains the best-studied eukaryotic cell in the world. Studies on the mitotic cell cycle and the discovery of cell cycle checkpoints in budding yeast has led to a detailed, although incomplete, understanding of eukaryotic cell cycle progression. In multicellular eukaryotic organisms, uncontrolled aberrant cell division is the defining feature of cancer. Some of the most successful classes of anti-cancer chemotherapeutic agents are mitotic poisons. Mitotic poisons are thought to function by inducing a mitotic spindle checkpoint-dependent cell cycle arrest, via the assembly of the highly conserved mitotic checkpoint complex (MCC), leading to apoptosis. Even in the presence of mitotic poisons, some cancer cells continue cell division via 'mitotic slippage', which may correlate with a cancer becoming refractory to mitotic poison chemotherapeutic treatments. In this review, knowledge about budding yeast cell cycle control is explored to suggest novel potential drug targets, namely, specific regions in the highly conserved anaphase-promoting complex/cyclosome (APC/C) subunits Apc1 and/or Apc5, and in a specific N-terminal region in the APC/C co-factor cell division cycle 20 (Cdc20), which may yield molecules which block 'mitotic slippage' only in the presence of mitotic poisons.
Collapse
Affiliation(s)
- Scott C. Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan;
- Division of Head and Neck Surgery, Department of Otolaryngology, Chang Gung Memorial Hospital, Kwei-Shan, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-03-211-8800 (ext. 3596)
| | - Hsin-Yu Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan;
| |
Collapse
|
8
|
Spectroscopic investigation, fungicidal activity and molecular dynamics simulation on benzimidazol-2-yl carbamate derivatives. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2018.08.092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
9
|
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: 0.4] [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
|
10
|
Shanmugam G, Jeon J. Computer-Aided Drug Discovery in Plant Pathology. THE PLANT PATHOLOGY JOURNAL 2017; 33:529-542. [PMID: 29238276 PMCID: PMC5720600 DOI: 10.5423/ppj.rw.04.2017.0084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 05/31/2023]
Abstract
Control of plant diseases is largely dependent on use of agrochemicals. However, there are widening gaps between our knowledge on plant diseases gained from genetic/mechanistic studies and rapid translation of the knowledge into target-oriented development of effective agrochemicals. Here we propose that the time is ripe for computer-aided drug discovery/design (CADD) in molecular plant pathology. CADD has played a pivotal role in development of medically important molecules over the last three decades. Now, explosive increase in information on genome sequences and three dimensional structures of biological molecules, in combination with advances in computational and informational technologies, opens up exciting possibilities for application of CADD in discovery and development of agrochemicals. In this review, we outline two categories of the drug discovery strategies: structure- and ligand-based CADD, and relevant computational approaches that are being employed in modern drug discovery. In order to help readers to dive into CADD, we explain concepts of homology modelling, molecular docking, virtual screening, and de novo ligand design in structure-based CADD, and pharmacophore modelling, ligand-based virtual screening, quantitative structure activity relationship modelling and de novo ligand design for ligand-based CADD. We also provide the important resources available to carry out CADD. Finally, we present a case study showing how CADD approach can be implemented in reality for identification of potent chemical compounds against the important plant pathogens, Pseudomonas syringae and Colletotrichum gloeosporioides.
Collapse
Affiliation(s)
| | - Junhyun Jeon
- Corresponding author. Phone) +82-53-810-3030, FAX) +82-53-810-4769, E-mail)
| |
Collapse
|
11
|
Domínguez Á, Muñoz E, López MC, Cordero M, Martínez JP, Viñas M. Transcriptomics as a tool to discover new antibacterial targets. Biotechnol Lett 2017; 39:819-828. [PMID: 28289911 DOI: 10.1007/s10529-017-2319-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
The emergence of antibiotic-resistant pathogens, multiple drug-resistance, and extremely drug-resistant strains demonstrates the need for improved strategies to discover new drug-based compounds. The development of transcriptomics, proteomics, and metabolomics has provided new tools for global studies of living organisms. However, the compendium of expression profiles produced by these methods has introduced new scientific challenges into antimicrobial research. In this review, we discuss the practical value of transcriptomic techniques as well as their difficulties and pitfalls. We advocate the construction of new databases of transcriptomic data, using standardized formats in addition to standardized models of bacterial and yeast similar to those used in systems biology. The inclusion of proteomic and metabolomic data is also essential, as the resulting networks can provide a landscape to rationally predict and exploit new drug targets and to understand drug synergies.
Collapse
Affiliation(s)
- Ángel Domínguez
- Department of Microbiology and Genetics, Universidad de Salamanca, Plaza de los Drs. de la Reina s/n, 37007, Salamanca, Spain.
| | - Elisa Muñoz
- Department of Cell Biology & Pathology, Universidad de Salamanca, Salamanca, Spain
| | - M Carmen López
- Department of Microbiology and Genetics, Universidad de Salamanca, Plaza de los Drs. de la Reina s/n, 37007, Salamanca, Spain
| | - Miguel Cordero
- Department of Medicine, Universidad de Salamanca, Salamanca, Spain
| | - José Pedro Martínez
- Department of Microbiology & Ecology, Universitat de Valencia/Estudi General (UVEG), Valencia, Spain
| | - Miguel Viñas
- Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Guerrero-Serrano G, Castanedo L, Cristóbal-Mondragón GR, Montalvo-Arredondo J, Riego-Ruíz L, DeLuna A, De Las Peñas A, Castaño I, Calera MR, Sánchez-Olea R. Npa3/ScGpn1 carboxy-terminal tail is dispensable for cell viability and RNA polymerase II nuclear targeting but critical for microtubule stability and function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:451-462. [PMID: 27965115 DOI: 10.1016/j.bbamcr.2016.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 12/03/2016] [Accepted: 12/09/2016] [Indexed: 10/20/2022]
Abstract
Genetic deletion of the essential GTPase Gpn1 or replacement of the endogenous gene by partial loss of function mutants in yeast is associated with multiple cellular phenotypes, including in all cases a marked cytoplasmic retention of RNA polymerase II (RNAPII). Global inhibition of RNAPII-mediated transcription due to malfunction of Gpn1 precludes the identification and study of other cellular function(s) for this GTPase. In contrast to the single Gpn protein present in Archaea, eukaryotic Gpn1 possesses an extension of approximately 100 amino acids at the C-terminal end of the GTPase domain. To determine the importance of this C-terminal extension in Saccharomyces cerevisiae Gpn1, we generated yeast strains expressing either C-terminal truncated (gpn1ΔC) or full-length ScGpn1. We found that ScGpn1ΔC was retained in the cell nucleus, an event physiologically relevant as gpn1ΔC cells contained a higher nuclear fraction of the RNAPII CTD phosphatase Rtr1. gpn1ΔC cells displayed an increased size, a delay in mitosis exit, and an increased sensitivity to the microtubule polymerization inhibitor benomyl at the cell proliferation level and two cellular events that depend on microtubule function: RNAPII nuclear targeting and vacuole integrity. These phenotypes were not caused by inhibition of RNAPII, as in gpn1ΔC cells RNAPII nuclear targeting and transcriptional activity were unaffected. These data, combined with our description here of a genetic interaction between GPN1 and BIK1, a microtubule plus-end tracking protein with a mitotic function, strongly suggest that the ScGpn1 C-terminal tail plays a critical role in microtubule dynamics and mitotic progression in an RNAPII-independent manner.
Collapse
Affiliation(s)
- Gehenna Guerrero-Serrano
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Manuel Nava 6, Zona Univesitaria, C.P. 78290, San Luis Potosí, San Luis Potosí, Mexico
| | - Leonardo Castanedo
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Manuel Nava 6, Zona Univesitaria, C.P. 78290, San Luis Potosí, San Luis Potosí, Mexico
| | - Gema R Cristóbal-Mondragón
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Manuel Nava 6, Zona Univesitaria, C.P. 78290, San Luis Potosí, San Luis Potosí, Mexico
| | - Javier Montalvo-Arredondo
- IPICYT, División de Biología Molecular, Camino a la Presa San José 2055, C.P. 78216, San Luis Potosí, San Luis Potosí, Mexico
| | - Lina Riego-Ruíz
- IPICYT, División de Biología Molecular, Camino a la Presa San José 2055, C.P. 78216, San Luis Potosí, San Luis Potosí, Mexico
| | - Alexander DeLuna
- LANGEBIO: Laboratorio Nacional de Genomica para la Biodiversidad, Km 9.6 Libramiento Norte Carretera León, C.P. 36821 Irapuato, Guanajuato, Mexico
| | - Alejandro De Las Peñas
- IPICYT, División de Biología Molecular, Camino a la Presa San José 2055, C.P. 78216, San Luis Potosí, San Luis Potosí, Mexico
| | - Irene Castaño
- IPICYT, División de Biología Molecular, Camino a la Presa San José 2055, C.P. 78216, San Luis Potosí, San Luis Potosí, Mexico
| | - Mónica R Calera
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Manuel Nava 6, Zona Univesitaria, C.P. 78290, San Luis Potosí, San Luis Potosí, Mexico.
| | - Roberto Sánchez-Olea
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Manuel Nava 6, Zona Univesitaria, C.P. 78290, San Luis Potosí, San Luis Potosí, Mexico.
| |
Collapse
|
13
|
Genomic sequencing-based mutational enrichment analysis identifies motility genes in a genetically intractable gut microbe. Proc Natl Acad Sci U S A 2016; 113:14127-14132. [PMID: 27911803 DOI: 10.1073/pnas.1612753113] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A major roadblock to understanding how microbes in the gastrointestinal tract colonize and influence the physiology of their hosts is our inability to genetically manipulate new bacterial species and experimentally assess the function of their genes. We describe the application of population-based genomic sequencing after chemical mutagenesis to map bacterial genes responsible for motility in Exiguobacterium acetylicum, a representative intestinal Firmicutes bacterium that is intractable to molecular genetic manipulation. We derived strong associations between mutations in 57 E. acetylicum genes and impaired motility. Surprisingly, less than half of these genes were annotated as motility-related based on sequence homologies. We confirmed the genetic link between individual mutations and loss of motility for several of these genes by performing a large-scale analysis of spontaneous suppressor mutations. In the process, we reannotated genes belonging to a broad family of diguanylate cyclases and phosphodiesterases to highlight their specific role in motility and assigned functions to uncharacterized genes. Furthermore, we generated isogenic strains that allowed us to establish that Exiguobacterium motility is important for the colonization of its vertebrate host. These results indicate that genetic dissection of a complex trait, functional annotation of new genes, and the generation of mutant strains to define the role of genes in complex environments can be accomplished in bacteria without the development of species-specific molecular genetic tools.
Collapse
|
14
|
Trends and Challenges in Pesticide Resistance Detection. TRENDS IN PLANT SCIENCE 2016; 21:834-853. [PMID: 27475253 DOI: 10.1016/j.tplants.2016.06.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/15/2016] [Accepted: 06/18/2016] [Indexed: 06/06/2023]
Abstract
Pesticide resistance is a crucial factor to be considered when developing strategies for the minimal use of pesticides while maintaining pesticide efficacy. This goal requires monitoring the emergence and development of resistance to pesticides in crop pests. To this end, various methods for resistance diagnosis have been developed for different groups of pests. This review provides an overview of biological, biochemical, and molecular methods that are currently used to detect and quantify pesticide resistance. The agronomic, technical, and economic advantages and drawbacks of each method are considered. Emerging technologies are also described, with their associated challenges and their potential for the detection of resistance mechanisms likely to be selected by current and future plant protection methods.
Collapse
|
15
|
Wong LH, Sinha S, Bergeron JR, Mellor JC, Giaever G, Flaherty P, Nislow C. Reverse Chemical Genetics: Comprehensive Fitness Profiling Reveals the Spectrum of Drug Target Interactions. PLoS Genet 2016; 12:e1006275. [PMID: 27588687 PMCID: PMC5010250 DOI: 10.1371/journal.pgen.1006275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/03/2016] [Indexed: 01/22/2023] Open
Abstract
The emergence and prevalence of drug resistance demands streamlined strategies to identify drug resistant variants in a fast, systematic and cost-effective way. Methods commonly used to understand and predict drug resistance rely on limited clinical studies from patients who are refractory to drugs or on laborious evolution experiments with poor coverage of the gene variants. Here, we report an integrative functional variomics methodology combining deep sequencing and a Bayesian statistical model to provide a comprehensive list of drug resistance alleles from complex variant populations. Dihydrofolate reductase, the target of methotrexate chemotherapy drug, was used as a model to identify functional mutant alleles correlated with methotrexate resistance. This systematic approach identified previously reported resistance mutations, as well as novel point mutations that were validated in vivo. Use of this systematic strategy as a routine diagnostics tool widens the scope of successful drug research and development. One of the most profound outcomes of fast, reliable genome sequencing is the ability to tailor drug therapy to an individual’s genotype. This ‘personalized’ or ‘precision medicine’ is the realization of a decades-long effort to maximize drug effect and limit unwanted side effects. An undesirable consequence of such targeted therapies, however, is the emergence of drug resistance. This outcome is the result of an evolutionary process where mutations in the drug target render the drug perturbation allow such mutant cells to proliferate. Because of the unbiased, and stochastic nature of the emergence of drug resistance, it is impossible to predict. We developed a test where hundreds of thousands of mutant cells are exposed to a drug simultaneously and those cells that modulate resistance survive. This method is innovative because it partners a high-throughput experimental protocol with a tailored statistical model to identify all mutations that modulate resistance. Finally, we used synthetic biology to re-create these mutations and demonstrate that they were, in fact, bona fide drug-resistant variants. These mutations were further extended and confirmed to also be resistant in the human orthologue. This combined biological-computational approach allows one to identify drug’s degree of resistance to both guide treatments and future drug discovery.
Collapse
Affiliation(s)
- Lai H. Wong
- Department of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Sunita Sinha
- Department of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Julien R. Bergeron
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | | | - Guri Giaever
- Department of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Patrick Flaherty
- Department of Mathematics and Statistics, University of Massachusetts, Amherst, Massachusetts, United States of America
- * E-mail: (PF); (CN)
| | - Corey Nislow
- Department of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
- * E-mail: (PF); (CN)
| |
Collapse
|
16
|
Goldgof GM, Durrant JD, Ottilie S, Vigil E, Allen KE, Gunawan F, Kostylev M, Henderson KA, Yang J, Schenken J, LaMonte GM, Manary MJ, Murao A, Nachon M, Murray R, Prescott M, McNamara CW, Slayman CW, Amaro RE, Suzuki Y, Winzeler EA. Comparative chemical genomics reveal that the spiroindolone antimalarial KAE609 (Cipargamin) is a P-type ATPase inhibitor. Sci Rep 2016; 6:27806. [PMID: 27291296 PMCID: PMC4904242 DOI: 10.1038/srep27806] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/20/2016] [Indexed: 11/30/2022] Open
Abstract
The spiroindolones, a new class of antimalarial medicines discovered in a cellular screen, are rendered less active by mutations in a parasite P-type ATPase, PfATP4. We show here that S. cerevisiae also acquires mutations in a gene encoding a P-type ATPase (ScPMA1) after exposure to spiroindolones and that these mutations are sufficient for resistance. KAE609 resistance mutations in ScPMA1 do not confer resistance to unrelated antimicrobials, but do confer cross sensitivity to the alkyl-lysophospholipid edelfosine, which is known to displace ScPma1p from the plasma membrane. Using an in vitro cell-free assay, we demonstrate that KAE609 directly inhibits ScPma1p ATPase activity. KAE609 also increases cytoplasmic hydrogen ion concentrations in yeast cells. Computer docking into a ScPma1p homology model identifies a binding mode that supports genetic resistance determinants and in vitro experimental structure-activity relationships in both P. falciparum and S. cerevisiae. This model also suggests a shared binding site with the dihydroisoquinolones antimalarials. Our data support a model in which KAE609 exerts its antimalarial activity by directly interfering with P-type ATPase activity.
Collapse
Affiliation(s)
- Gregory M. Goldgof
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | - Jacob D. Durrant
- Department of Chemistry & Biochemistry and the National
Biomedical Computation Resource, University of California, San
Diego, La Jolla, California, USA
| | - Sabine Ottilie
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Edgar Vigil
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Kenneth E. Allen
- Department of Genetics, Yale University School of
Medicine, New Haven, Connecticut, USA
| | - Felicia Gunawan
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Maxim Kostylev
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | | | - Jennifer Yang
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Jake Schenken
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Gregory M. LaMonte
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Micah J. Manary
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Ayako Murao
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | - Marie Nachon
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Rebecca Murray
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Maximo Prescott
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Case W. McNamara
- Genomics Institute of the Novartis Research Foundation,
San Diego, California, USA
| | - Carolyn W. Slayman
- Department of Genetics, Yale University School of
Medicine, New Haven, Connecticut, USA
| | - Rommie E. Amaro
- Department of Chemistry & Biochemistry and the National
Biomedical Computation Resource, University of California, San
Diego, La Jolla, California, USA
| | - Yo Suzuki
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | - Elizabeth A. Winzeler
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| |
Collapse
|
17
|
Duncan R, Kourout M, Grigorenko E, Fisher C, Dong M. Advances in multiplex nucleic acid diagnostics for blood-borne pathogens: promises and pitfalls. Expert Rev Mol Diagn 2015; 16:83-95. [PMID: 26581018 DOI: 10.1586/14737159.2016.1112272] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The large number of blood-borne viruses, bacteria and parasites currently of concern, as well as many newly emerging pathogens, presents a daunting challenge to protection of the safety of blood for transfusion and diagnosing infectious diseases. Focusing on nucleic acid diagnostic tests, multiplex devices are coming into use with many more in various developmental stages that promise to offer solutions to the clinical need. The characteristics, advantages and disadvantages of platforms in clinical use and at the research and development stage are examined here. The presence of multiple assays and associated reagents operating simultaneously on one platform, implementation in traditional clinical laboratories and regulatory review will present special challenges. Fortunately, clinical laboratories have made dramatic technical progress in the last two decades and regulatory agencies have publicly expressed support for development of multiplex devices.
Collapse
Affiliation(s)
- Robert Duncan
- a Center for Biologics Evaluation and Research , US FDA , Silver Spring , MD , USA
| | - Moussa Kourout
- a Center for Biologics Evaluation and Research , US FDA , Silver Spring , MD , USA
| | | | - Carolyn Fisher
- a Center for Biologics Evaluation and Research , US FDA , Silver Spring , MD , USA
| | - Ming Dong
- a Center for Biologics Evaluation and Research , US FDA , Silver Spring , MD , USA
| |
Collapse
|