1
|
Friday SN, Koellner LA, Brown SW, Calbat CN, Curran CF, Dietz JD, Koenig‐Dummer A, Friday PD, Parker JA, Simmons NA, Urmey FA, West AN, Zagler SG, Viola RE, Halkides CJ. Development of Efficient Covalent Inactivators of a Fungal Aspartate Semialdehyde Dehydrogenase. Drug Dev Res 2025; 86:e70095. [PMID: 40341627 PMCID: PMC12060211 DOI: 10.1002/ddr.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/21/2025] [Accepted: 04/12/2025] [Indexed: 05/10/2025]
Abstract
Aspartate semialdehyde dehydrogenase (ASADH) catalyzes the second step in the fungal pathway towards the synthesis of threonine, isoleucine, and methionine, and it has been identified as a viable target for antifungal drug development. Our previous work produced a group of vinyl sulfones that function as irreversible covalent inactivators of this enzyme. We have now expanded this initial set to produce vinyl sulfones with higher kinetic efficiency as covalent inactivators of ASADH purified from the pathogenic fungal species Candida albicans. The catalytic efficiency of these inactivators has also been compared to related classes of irreversible inactivators, vinyl sulfonamides, acrylamides, and sulfonyl ketones.
Collapse
Affiliation(s)
- Samantha N. Friday
- Department of Chemistry and BiochemistryUniversity of ToledoToledoOhioUSA
| | - Liezel A. Koellner
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Spencer W. Brown
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Christopher N. Calbat
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Catherine F. Curran
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Jordan D. Dietz
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Andreas Koenig‐Dummer
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Paul D. Friday
- Department of Chemistry and BiochemistryUniversity of ToledoToledoOhioUSA
| | - James A. Parker
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Noah A. Simmons
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Finnean A. Urmey
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Alexis N. West
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Sebastian G. Zagler
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| | - Ronald E. Viola
- Department of Chemistry and BiochemistryUniversity of ToledoToledoOhioUSA
| | - Christopher J. Halkides
- Department of Chemistry and BiochemistryUniversity of North Carolina WilmingtonWilmingtonNorth CarolinaUSA
| |
Collapse
|
2
|
Yan ZZ, Hu HW, Xiong C, Peleg AY, Chen QL, Sáez-Sandino T, Maestre F, Delgado-Baquerizo M, Singh BK. Environmental microbiome, human fungal pathogens, and antimicrobial resistance. Trends Microbiol 2025; 33:112-129. [PMID: 39304419 DOI: 10.1016/j.tim.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Traditionally, antifungal resistance (AFR) has received much less attention compared with bacterial resistance to antibiotics. However, global changes, pandemics, and emerging new fungal infections have highlighted global health consequences of AFR. The recent report of the World Health Organisation (WHO) has identified fungal priority pathogens, and recognised AFR among the greatest global health threats. This is particularly important given the significant increase in fungal infections linked to climate change and pandemics. Environmental factors play critical roles in AFR and fungal infections, as many clinically relevant fungal pathogens and AFR originate from the environment (mainly soil). In addition, the environment serves as a potential rich source for the discovery of new antifungal agents, including mycoviruses and bacterial probiotics, which hold promise for effective therapies. In this article, we summarise the environmental pathways of AFR development and spread among high priority fungal pathogens, and propose potential mechanisms of AFR development and spread. We identify a research priority list to address key knowledge gaps in our understanding of environmental AFR. Further, we propose an integrated roadmap for predictive risk management of AFR that is critical for effective surveillance and forecasting of public health outcomes under current and future climatic conditions.
Collapse
Affiliation(s)
- Zhen-Zhen Yan
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia
| | - Hang-Wei Hu
- School of Agriculture, Food, and Ecosystem Science, Faculty of Science, The University of Melbourne, Victoria, Australia
| | - Chao Xiong
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia
| | - Anton Y Peleg
- Department of Infectious Disease, The Alfred Hospital and Central Clinical School, Monash University, Victoria, Australia; Department of Microbiology, Monash University, Melbourne, Australia; Centre to Impact Antimicrobial Resistance, Monash University, Melbourne, Australia
| | - Qing-Lin Chen
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Science, Xiamen, China; University of Chinese Academy of Sciences, Beijing, China
| | - Tadeo Sáez-Sandino
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia
| | - Fernando Maestre
- Environmental Sciences and Engineering, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Manuel Delgado-Baquerizo
- Laboratorio de Biodiversidad y Funcionamiento Ecosistémico, Instituto de Recursos Naturales y Agrobiología de Sevilla (IRNAS), CSIC, Sevilla, Spain
| | - Brajesh K Singh
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia.
| |
Collapse
|
3
|
Carolus H, Sofras D, Boccarella G, Jacobs S, Biriukov V, Goossens L, Chen A, Vantyghem I, Verbeeck T, Pierson S, Lobo Romero C, Steenackers H, Lagrou K, van den Berg P, Berman J, Gabaldón T, Van Dijck P. Collateral sensitivity counteracts the evolution of antifungal drug resistance in Candida auris. Nat Microbiol 2024; 9:2954-2969. [PMID: 39472696 DOI: 10.1038/s41564-024-01811-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/15/2024] [Indexed: 11/02/2024]
Abstract
Antifungal drug resistance represents a serious global health threat, necessitating new treatment strategies. Here we investigated collateral sensitivity (CS), in which resistance to one drug increases sensitivity to another, and cross-resistance (XR), in which one drug resistance mechanism reduces susceptibility to multiple drugs, since CS and XR dynamics can guide treatment design to impede resistance development, but have not been systematically explored in pathogenic fungi. We used experimental evolution and mathematical modelling of Candida auris population dynamics during cyclic and combined drug exposures and found that especially CS-based drug cycling can effectively prevent the emergence of drug resistance. In addition, we found that a CS-based treatment switch can actively select against or eradicate resistant sub-populations, highlighting the potential to consider CS in therapeutic decision-making upon resistance detection. Furthermore, we show that some CS trends are robust among different strains and resistance mechanisms. Overall, these findings provide a promising direction for improved antifungal treatment approaches.
Collapse
Affiliation(s)
- Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium.
| | - Dimitrios Sofras
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Giorgio Boccarella
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
- Evolutionary Modelling Group, Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Stef Jacobs
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Vladislav Biriukov
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Louise Goossens
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Alicia Chen
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ina Vantyghem
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Tibo Verbeeck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Siebe Pierson
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Celia Lobo Romero
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Hans Steenackers
- Centre for Microbial and Plant Genetics, Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| | - Pieter van den Berg
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
- Evolutionary Modelling Group, Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium.
- KU Leuven One Health Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Schmidlin K, Apodaca S, Newell D, Sastokas A, Kinsler G, Geiler-Samerotte K. Distinguishing mutants that resist drugs via different mechanisms by examining fitness tradeoffs. eLife 2024; 13:RP94144. [PMID: 39255191 PMCID: PMC11386965 DOI: 10.7554/elife.94144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
There is growing interest in designing multidrug therapies that leverage tradeoffs to combat resistance. Tradeoffs are common in evolution and occur when, for example, resistance to one drug results in sensitivity to another. Major questions remain about the extent to which tradeoffs are reliable, specifically, whether the mutants that provide resistance to a given drug all suffer similar tradeoffs. This question is difficult because the drug-resistant mutants observed in the clinic, and even those evolved in controlled laboratory settings, are often biased towards those that provide large fitness benefits. Thus, the mutations (and mechanisms) that provide drug resistance may be more diverse than current data suggests. Here, we perform evolution experiments utilizing lineage-tracking to capture a fuller spectrum of mutations that give yeast cells a fitness advantage in fluconazole, a common antifungal drug. We then quantify fitness tradeoffs for each of 774 evolved mutants across 12 environments, finding these mutants group into classes with characteristically different tradeoffs. Their unique tradeoffs may imply that each group of mutants affects fitness through different underlying mechanisms. Some of the groupings we find are surprising. For example, we find some mutants that resist single drugs do not resist their combination, while others do. And some mutants to the same gene have different tradeoffs than others. These findings, on one hand, demonstrate the difficulty in relying on consistent or intuitive tradeoffs when designing multidrug treatments. On the other hand, by demonstrating that hundreds of adaptive mutations can be reduced to a few groups with characteristic tradeoffs, our findings may yet empower multidrug strategies that leverage tradeoffs to combat resistance. More generally speaking, by grouping mutants that likely affect fitness through similar underlying mechanisms, our work guides efforts to map the phenotypic effects of mutation.
Collapse
Affiliation(s)
- Kara Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Sam Apodaca
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Daphne Newell
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Alexander Sastokas
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Grant Kinsler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, United States
| | - Kerry Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| |
Collapse
|
5
|
Murante D, Hogan DA. Drivers of diversification in fungal pathogen populations. PLoS Pathog 2024; 20:e1012430. [PMID: 39264909 PMCID: PMC11392411 DOI: 10.1371/journal.ppat.1012430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Abstract
To manage and treat chronic fungal diseases effectively, we require an improved understanding of their complexity. There is an increasing appreciation that chronic infection populations are often heterogeneous due to diversification and drift, even within a single microbial species. Genetically diverse populations can contribute to persistence and resistance to treatment by maintaining cells with different phenotypes capable of thriving in these dynamic environments. In chronic infections, fungal pathogens undergo prolonged challenges that can drive trait selection to convergent adapted states through restricted access to critical nutrients, assault by immune effectors, competition with other species, and antifungal drugs. This review first highlights the various genetic and epigenetic mechanisms that promote diversity in pathogenic fungal populations and provide an additional barrier to assessing the actual heterogeneity of fungal infections. We then review existing studies of evolution and genetic heterogeneity in fungal populations from lung infections associated with the genetic disease cystic fibrosis. We conclude with a discussion of open research questions that, once answered, may aid in diagnosing and treating chronic fungal infections.
Collapse
Affiliation(s)
- Daniel Murante
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Deborah Ann Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
6
|
Anderson MZ, Dietz SM. Evolution and strain diversity advance exploration of Candida albicans biology. mSphere 2024; 9:e0064123. [PMID: 39012122 PMCID: PMC11351040 DOI: 10.1128/msphere.00641-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
Fungi were some of the earliest organismal systems used to explore mutational processes and its phenotypic consequences on members of a species. Yeasts that cause significant human disease were quickly incorporated into these investigations to define the genetic and phenotypic drivers of virulence. Among Candida species, Candida albicans has emerged as a model for studying genomic processes of evolution because of its clinical relevance, relatively small genome, and ability to tolerate complex chromosomal changes. Here, we describe major recent findings that used evolution of strains from defined genetic backgrounds to delineate mutational and adaptative processes and include how nascent exploration into naturally occurring variation is contributing to these conceptual frameworks. Ultimately, efforts to discern adaptive mechanisms used by C. albicans will continue to divulge new biology and can better inform treatment regimens for the increasing prevalence of fungal disease.
Collapse
Affiliation(s)
- Matthew Z. Anderson
- Department of Medical Genetics, Laboratory of Genetics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Center for Genomic Science Innovation, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Siobhan M. Dietz
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| |
Collapse
|
7
|
Cárdenas Parra LY, Rojas Rodríguez AE, Pérez Cárdenas JE, Pérez-Agudelo JM. Molecular Evaluation of the mRNA Expression of the ERG11, ERG3, CgCDR1, and CgSNQ2 Genes Linked to Fluconazole Resistance in Candida glabrata in a Colombian Population. J Fungi (Basel) 2024; 10:509. [PMID: 39057394 PMCID: PMC11277825 DOI: 10.3390/jof10070509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION The study of Candida glabrata genes associated with fluconazole resistance, from a molecular perspective, increases the understanding of the phenomenon with a view to its clinical applicability. OBJECTIVE We sought to establish the predictive molecular profile of fluconazole resistance in Candida glabrata by analyzing the ERG11, ERG3, CgCDR1, and CgSNQ2 genes. METHOD Expression was quantified using RT-qPCR. Metrics were obtained through molecular docking and Fisher discriminant functions. Additionally, a predictive classification was made against the susceptibility of C. glabrata to fluconazole. RESULTS The relative expression of the ERG3, CgCDR1, and CgSNQ2 genes was higher in the fluconazole-resistant strains than in the fluconazole-susceptible, dose-dependent strains. The gene with the highest relative expression in the fluconazole-exposed strains was CgCDR1, and in both the resistant and susceptible, dose-dependent strains exposed to fluconazole, this was also the case. The molecular docking model generated a median number of contacts between fluconazole and ERG11 that was lower than the median number of contacts between fluconazole and ERG3, -CgCDR1, and -CgSNQ2. The predicted classification through the multivariate model for fluconazole susceptibility achieved an accuracy of 73.5%. CONCLUSION The resistant strains had significant expression levels of genes encoding efflux pumps and the ERG3 gene. Molecular analysis makes the identification of a low affinity between fluconazole and its pharmacological target possible, which may explain the lower intrinsic susceptibility of the fungus to fluconazole.
Collapse
Affiliation(s)
- Leidy Yurany Cárdenas Parra
- Facultad de Ciencias para la Salud, Universidad de Caldas, Manizales 170004, Colombia; (L.Y.C.P.); (J.E.P.C.); (J.M.P.-A.)
- Facultad de Ciencias de la Salud, Universidad Católica de Manizales, Manizales 170001, Colombia
| | | | - Jorge Enrique Pérez Cárdenas
- Facultad de Ciencias para la Salud, Universidad de Caldas, Manizales 170004, Colombia; (L.Y.C.P.); (J.E.P.C.); (J.M.P.-A.)
| | - Juan Manuel Pérez-Agudelo
- Facultad de Ciencias para la Salud, Universidad de Caldas, Manizales 170004, Colombia; (L.Y.C.P.); (J.E.P.C.); (J.M.P.-A.)
| |
Collapse
|
8
|
Das S, Singh S, Tawde Y, Dutta TK, Rudramurthy SM, Kaur H, Shaw T, Ghosh A. Comparative fitness trade-offs associated with azole resistance in Candida auris clinical isolates. Heliyon 2024; 10:e32386. [PMID: 38988564 PMCID: PMC11233892 DOI: 10.1016/j.heliyon.2024.e32386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Multidrug-resistant yeast Candida auris is a serious threat to public health with documented survival in various hospital niches. The dynamics of this survival benefit and its trade off with drug resistance are still unknown for this pathogen. In this study we investigate the oxidative stress response (OSR) in fluconazole-resistant C. auris and compare its relative fitness with fluconazole-susceptible strains. A total of 351 C. auris clinical isolates (61 fluconazole-susceptible and 290 fluconazole-resistant) were screened for stress tolerance by spot assay and 95.08 % fluconazole-susceptible isolates were hyper-resistant to oxidative stress while majority (94.5 %) fluconazole-resistant isolates had lower oxidative tolerance. Expression of Hog1 and Cta1 gene transcript levels and cellular catalase levels were significantly higher in fluconazole-susceptible isolates and a corresponding higher intracellular reactive oxygen species level (iROS) was accumulated in the fluconazole-resistant isolates. Biofilm formation and cell viability under oxidative stress revealed higher biofilm formation and better viability in fluconazole-susceptible isolates. Fluconazole-resistant isolates had higher basal cell wall chitin. On comparison of virulence, the % cytotoxicity in A549 cell line was higher in fluconazole-susceptible isolates and the median survival of the infected larvae in G. mellonella infection model was higher in fluconazole-resistant (5; IQR:4.5-5 days) vs. fluconazole-susceptible C. auris (2; IQR:1.5-2.5 days). All organisms evolve with changes in their environmental conditions, to ensure an optimal balance between proliferation and survival. Development of tolerance to a certain kind of stress example antifungal exposure in yeast can leads to a compensatory decrease in tolerance for other stresses. This study provides useful insights into the comparative fitness and antifungal susceptibility trade off in C. auris. We report a negative association between H2O2 tolerance and fluconazole susceptibility. Using in-vitro cell cytotoxicity and in-vivo survival assays we also demonstrate the higher virulence potential of fluconazole-susceptible C. auris isolates corroborating the negative correlation between susceptibility and pathogen survival or virulence. These findings could also be translated to clinical practice by investigating the possibility of using molecules targeting stress response and fitness regulating pathways for management of this serious infection.
Collapse
Affiliation(s)
- Sourav Das
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shreya Singh
- Department of Microbiology. Dr. B. R. Ambedkar State Institute of Medical Science, Mohali, Punjab, India
| | - Yamini Tawde
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Tushar K. Dutta
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Shivaprakash M. Rudramurthy
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harsimran Kaur
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Tushar Shaw
- Department of Life and Allied Health Sciences, Ramaiah university of Applied sciences, Bangalore, India
| | - Anup Ghosh
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
9
|
Schmidlin, Apodaca, Newell, Sastokas, Kinsler, Geiler-Samerotte. Distinguishing mutants that resist drugs via different mechanisms by examining fitness tradeoffs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.17.562616. [PMID: 37905147 PMCID: PMC10614906 DOI: 10.1101/2023.10.17.562616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
There is growing interest in designing multidrug therapies that leverage tradeoffs to combat resistance. Tradeoffs are common in evolution and occur when, for example, resistance to one drug results in sensitivity to another. Major questions remain about the extent to which tradeoffs are reliable, specifically, whether the mutants that provide resistance to a given drug all suffer similar tradeoffs. This question is difficult because the drug-resistant mutants observed in the clinic, and even those evolved in controlled laboratory settings, are often biased towards those that provide large fitness benefits. Thus, the mutations (and mechanisms) that provide drug resistance may be more diverse than current data suggests. Here, we perform evolution experiments utilizing lineage-tracking to capture a fuller spectrum of mutations that give yeast cells a fitness advantage in fluconazole, a common antifungal drug. We then quantify fitness tradeoffs for each of 774 evolved mutants across 12 environments, finding these mutants group into 6 classes with characteristically different tradeoffs. Their unique tradeoffs may imply that each group of mutants affects fitness through different underlying mechanisms. Some of the groupings we find are surprising. For example, we find some mutants that resist single drugs do not resist their combination, while others do. And some mutants to the same gene have different tradeoffs than others. These findings, on one hand, demonstrate the difficulty in relying on consistent or intuitive tradeoffs when designing multidrug treatments. On the other hand, by demonstrating that hundreds of adaptive mutations can be reduced to a few groups with characteristic tradeoffs, our findings may yet empower multidrug strategies that leverage tradeoffs to combat resistance. More generally speaking, by grouping mutants that likely affect fitness through similar underlying mechanisms, our work guides efforts to map the phenotypic effects of mutation.
Collapse
Affiliation(s)
- Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Apodaca
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Newell
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Sastokas
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Kinsler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| |
Collapse
|
10
|
Sultanaev V, Yakimova L, Nazarova A, Sedov I, Mostovaya O, Mukhametzyanov T, Davletshin D, Takuntseva D, Gilyazova E, Bulatov E, Stoikov I. Pillar[5]arene/albumin biosupramolecular systems for simultaneous native protein preservation and encapsulation of a water-soluble substrate. J Mater Chem B 2024; 12:3103-3114. [PMID: 38450640 DOI: 10.1039/d3tb02961a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The growing resistance of pathogens, bacteria, viruses, and fungi to a number of drugs has encouraged researchers to use natural and synthetic biomimetic systems to overcome this challenge. Multicomponent systems are an attractive approach for drug design and multitarget therapy. In this study, we report the assembly of a three-component (pillar[5]arene, bovine serum albumin, and methyl orange) biosupramolecular system as a potential drug delivery system. We estimated the cytotoxic activity and transfection ability of pillar[5]arene derivatives and investigated the effect of the nature of macrocycle functions (L-phenylalanine, glycine, L-alanine) on the native conformation of serum albumin in a three-component system. NMR, UV-vis, fluorescence, CD spectroscopy, DLS, and molecular docking studies were performed in order to confirm the structure and possible pillar[5]arene/bovine serum albumin/methyl orange interactions occurring during the association process. Results indicate that pillar[5]arene with L-phenylalanine fragments retains the native form of BSA to the maximum extent and forms more stable associates.
Collapse
Affiliation(s)
- Vildan Sultanaev
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Luidmila Yakimova
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Anastasia Nazarova
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Igor Sedov
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Olga Mostovaya
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Timur Mukhametzyanov
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Damir Davletshin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Daria Takuntseva
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| | - Elvina Gilyazova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Ivan Stoikov
- A.M. Butlerov Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia.
| |
Collapse
|
11
|
Spruijtenburg B, Ahmad S, Asadzadeh M, Alfouzan W, Al-Obaid I, Mokaddas E, Meijer EFJ, Meis JF, de Groot T. Whole genome sequencing analysis demonstrates therapy-induced echinocandin resistance in Candida auris isolates. Mycoses 2023; 66:1079-1086. [PMID: 37712885 DOI: 10.1111/myc.13655] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/16/2023]
Abstract
Candida auris is an emerging, multidrug-resistant yeast, causing outbreaks in healthcare facilities. Echinocandins are the antifungal drugs of choice to treat candidiasis, as they cause few side effects and resistance is rarely found. Previously, immunocompromised patients from Kuwait with C. auris colonisation or infection were treated with echinocandins, and within days to months, resistance was reported in urine isolates. To determine whether the development of echinocandin resistance was due to independent introductions of resistant strains or resulted from intra-patient resistance development, whole genome sequencing (WGS) single-nucleotide polymorphism (SNP) analysis was performed on susceptible (n = 26) and echinocandin-resistant (n = 6) isolates from seven patients. WGS SNP analysis identified three distinct clusters differing 17-127 SNPs from two patients, and the remaining isolates from five patients, respectively. Sequential isolates within patients had a maximum of 11 SNP differences over a time period of 1-10 months. The majority of isolates with reduced susceptibility displayed unique FKS1 substitutions including a novel FKS1M690V substitution, and nearly all were genetically related, ranging from only three to six SNP differences compared to susceptible isolates from the same patient. Resistant isolates from three patients shared the common FKS1S639F substitution; however, WGS analysis did not suggest a common source. These findings strongly indicate that echinocandin resistance is induced during antifungal treatment. Future studies should determine whether such echinocandin-resistant strains are capable of long-term colonisation, cause subsequent breakthrough candidiasis, have a propensity to cross-infect other patients, or remain viable for longer time periods in the hospital environment.
Collapse
Affiliation(s)
- Bram Spruijtenburg
- Department of Medical Microbiology and Infectious Diseases, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
- Center of Expertise for Mycology Radboud University Medical Center/Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Suhail Ahmad
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Mohammad Asadzadeh
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Wadha Alfouzan
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
- Microbiology Unit, Department of Laboratory Medicine, Farwania Hospital, Kuwait City, Kuwait
| | - Inaam Al-Obaid
- Department of Microbiology, Al-Sabah Hospital, Shuwaikh, Kuwait
| | - Eiman Mokaddas
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
- Department of Microbiology, Ibn-Sina Hospital, Shuwaikh, Kuwait
| | - Eelco F J Meijer
- Department of Medical Microbiology and Infectious Diseases, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
- Center of Expertise for Mycology Radboud University Medical Center/Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Jacques F Meis
- Center of Expertise for Mycology Radboud University Medical Center/Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Theun de Groot
- Department of Medical Microbiology and Infectious Diseases, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
- Center of Expertise for Mycology Radboud University Medical Center/Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Metzner K, O’Meara MJ, Halligan B, Wotring JW, Sexton JZ, O’Meara TR. Imaging-Based Screening Identifies Modulators of the eIF3 Translation Initiation Factor Complex in Candida albicans. Antimicrob Agents Chemother 2023; 67:e0050323. [PMID: 37382550 PMCID: PMC10353439 DOI: 10.1128/aac.00503-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/07/2023] [Indexed: 06/30/2023] Open
Abstract
Fungal pathogens like Candida albicans can cause devastating human disease. Treatment of candidemia is complicated by the high rate of resistance to common antifungal therapies. Additionally, there is host toxicity associated with many antifungal compounds due to the conservation between essential mammalian and fungal proteins. An attractive new approach for antimicrobial development is to target virulence factors: non-essential processes that are required for the organism to cause disease in human hosts. This approach expands the potential target space while reducing the selective pressure toward resistance, as these targets are not essential for viability. In C. albicans, a key virulence factor is the ability to transition to hyphal morphology. We developed a high-throughput image analysis pipeline to distinguish between yeast and filamentous growth in C. albicans at the single cell level. Based on this phenotypic assay, we screened the FDA drug repurposing library of 2,017 compounds for their ability to inhibit filamentation and identified 33 compounds that block the hyphal transition in C. albicans with IC50 values ranging from 0.2 to 150 μM. Multiple compounds showed a phenyl sulfone chemotype, prompting further analysis. Of these phenyl sulfones, NSC 697923 displayed the most efficacy, and by selecting for resistant mutants, we identified eIF3 as the target of NSC 697923 in C. albicans.
Collapse
Affiliation(s)
- Katura Metzner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin Halligan
- University of Michigan Center for Drug Repurposing, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jesse W. Wotring
- University of Michigan Center for Drug Repurposing, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, Michigan, USA
| | - Jonathan Z. Sexton
- University of Michigan Center for Drug Repurposing, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, Michigan, USA
| | - Teresa R. O’Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Anderson FM, Visser ND, Amses KR, Hodgins-Davis A, Weber AM, Metzner KM, McFadden MJ, Mills RE, O’Meara MJ, James TY, O’Meara TR. Candida albicans selection for human commensalism results in substantial within-host diversity without decreasing fitness for invasive disease. PLoS Biol 2023; 21:e3001822. [PMID: 37205709 PMCID: PMC10234564 DOI: 10.1371/journal.pbio.3001822] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 06/01/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Candida albicans is a frequent colonizer of human mucosal surfaces as well as an opportunistic pathogen. C. albicans is remarkably versatile in its ability to colonize diverse host sites with differences in oxygen and nutrient availability, pH, immune responses, and resident microbes, among other cues. It is unclear how the genetic background of a commensal colonizing population can influence the shift to pathogenicity. Therefore, we examined 910 commensal isolates from 35 healthy donors to identify host niche-specific adaptations. We demonstrate that healthy people are reservoirs for genotypically and phenotypically diverse C. albicans strains. Using limited diversity exploitation, we identified a single nucleotide change in the uncharacterized ZMS1 transcription factor that was sufficient to drive hyper invasion into agar. We found that SC5314 was significantly different from the majority of both commensal and bloodstream isolates in its ability to induce host cell death. However, our commensal strains retained the capacity to cause disease in the Galleria model of systemic infection, including outcompeting the SC5314 reference strain during systemic competition assays. This study provides a global view of commensal strain variation and within-host strain diversity of C. albicans and suggests that selection for commensalism in humans does not result in a fitness cost for invasive disease.
Collapse
Affiliation(s)
- Faith M. Anderson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Noelle D. Visser
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kevin R. Amses
- Department of Ecology and Evolution, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrea Hodgins-Davis
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Alexandra M. Weber
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Katura M. Metzner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Michael J. McFadden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Ryan E. Mills
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Timothy Y. James
- Department of Ecology and Evolution, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Teresa R. O’Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
14
|
Metzner K, O’Meara MJ, Halligan B, Wotring JW, Sexton JZ, O’Meara TR. Imaging-based screening identifies modulators of the eIF3 translation initiation factor complex in Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537517. [PMID: 37131825 PMCID: PMC10153179 DOI: 10.1101/2023.04.19.537517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Fungal pathogens like Candida albicans can cause devastating human disease. Treatment of candidemia is complicated by the high rate of resistance to common antifungal therapies. Additionally, there is host toxicity associated with many antifungal compounds due to the conservation between essential mammalian and fungal proteins. An attractive new approach for antimicrobial development is to target virulence factors: non-essential processes that are required for the organism to cause disease in human hosts. This approach expands the potential target space while reducing the selective pressure towards resistance, as these targets are not essential for viability. In C. albicans, a key virulence factor is the ability to transition to hyphal morphology. We developed a high-throughput image analysis pipeline to distinguish between yeast and filamentous growth in C. albicans at the single cell level. Based on this phenotypic assay, we screened the FDA drug repurposing library of 2,017 compounds for their ability to inhibit filamentation and identified 33 compounds that block the hyphal transition in C. albicans with IC 50 values ranging from 0.2 to 150 µM. Multiple compounds showed a phenyl vinyl sulfone chemotype, prompting further analysis. Of these phenyl vinyl sulfones, NSC 697923 displayed the most efficacy, and by selecting for resistant mutants, we identified eIF3 as the target of NSC 697923 in C. albicans .
Collapse
Affiliation(s)
- Katura Metzner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew J O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin Halligan
- University of Michigan Center for Drug Repurposing, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jesse W. Wotring
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, MI, USA
- University of Michigan Center for Drug Repurposing, USA
| | - Jonathan Z Sexton
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, MI, USA
- University of Michigan Center for Drug Repurposing, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Teresa R O’Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Antifungal Tolerance and Resistance Emerge at Distinct Drug Concentrations and Rely upon Different Aneuploid Chromosomes. mBio 2023; 14:e0022723. [PMID: 36877011 PMCID: PMC10127634 DOI: 10.1128/mbio.00227-23] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Antifungal drug tolerance is a response distinct from resistance, in which cells grow slowly above the MIC. Here, we found that the majority (69.2%) of 133 Candida albicans clinical isolates, including standard lab strain SC5314, exhibited temperature-enhanced tolerance at 37°C and 39°C, and were not tolerant at 30°C. Other isolates were either always tolerant (23.3%) or never tolerant (7.5%) at these three temperatures, suggesting that tolerance requires different physiological processes in different isolates. At supra-MIC fluconazole concentrations (8 to 128 μg/mL), tolerant colonies emerged rapidly at a frequency of ~10-3. In liquid passages over a broader range of fluconazole concentrations (0.25 to 128 μg/mL), tolerance emerged rapidly (within one passage) at supra-MICs. In contrast, resistance appeared at sub-MICs after 5 or more passages. Of 155 adaptors that evolved higher tolerance, all carried one of several recurrent aneuploid chromosomes, often including chromosome R, alone or in combination with other chromosomes. Furthermore, loss of these recurrent aneuploidies was associated with a loss of acquired tolerance, indicating that specific aneuploidies confer fluconazole tolerance. Thus, genetic background and physiology and the degree of drug stress (above or below the MIC) influence the evolutionary trajectories and dynamics with which antifungal drug resistance or tolerance emerges. IMPORTANCE Antifungal drug tolerance differs from drug resistance: tolerant cells grow slowly in drug, while resistant cells usually grow well, due to mutations in a few known genes. More than half of Candida albicans clinical isolates have higher tolerance at body temperature than they do at the lower temperatures used for most lab experiments. This implies that different isolates achieve drug tolerance via several cellular processes. When we evolved different strains at a range of high drug concentrations above inhibitory levels, tolerance emerged rapidly and at high frequency (one in 1,000 cells) while resistance appeared only later at very low drug concentrations. An extra copy of all or part of chromosome R was associated with tolerance, while point mutations or different aneuploidies were seen with resistance. Thus, genetic background and physiology, temperature, and drug concentration all influence how drug tolerance or resistance evolves.
Collapse
|
16
|
Sun LL, Li H, Yan TH, Cao YB, Jiang YY, Yang F. Aneuploidy enables cross-tolerance to unrelated antifungal drugs in Candida parapsilosis. Front Microbiol 2023; 14:1137083. [PMID: 37113223 PMCID: PMC10126355 DOI: 10.3389/fmicb.2023.1137083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Candida parapsilosis is an emerging major human fungal pathogen. Echinocandins are first-line antifungal drugs for the treatment of invasive Candida infections. In clinical isolates, tolerance to echinocandins in Candida species is mostly due to point mutations of FKS genes, which encode the target protein of echinocandins. However, here, we found chromosome 5 trisomy was the major mechanism of adaptation to the echinocandin drug caspofungin, and FKS mutations were rare events. Chromosome 5 trisomy conferred tolerance to echinocandin drugs caspofungin and micafungin and cross-tolerance to 5-flucytosine, another class of antifungal drugs. The inherent instability of aneuploidy caused unstable drug tolerance. Tolerance to echinocandins might be due to increased copy number and expression of CHS7, which encodes chitin synthase. Although copy number of chitinase genes CHT3 and CHT4 was also increased to the trisomic level, the expression was buffered to the disomic level. Tolerance to 5-flucytosine might be due to the decreased expression of FUR1. Therefore, the pleiotropic effect of aneuploidy on antifungal tolerance was due to the simultaneous regulation of genes on the aneuploid chromosome and genes on euploid chromosomes. In summary, aneuploidy provides a rapid and reversible mechanism of drug tolerance and cross-tolerance in C. parapsilosis.
Collapse
Affiliation(s)
- Liu-liu Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tian-hua Yan
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yong-bing Cao
- Department of Vascular Diseases, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-ying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Yuan-ying Jiang
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Feng Yang
| |
Collapse
|
17
|
Avramovska O, Smith AC, Rego E, Hickman MA. Tetraploidy accelerates adaptation under drug selection in a fungal pathogen. FRONTIERS IN FUNGAL BIOLOGY 2022; 3:984377. [PMID: 37746235 PMCID: PMC10512305 DOI: 10.3389/ffunb.2022.984377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/06/2022] [Indexed: 09/26/2023]
Abstract
Baseline ploidy significantly impacts evolutionary trajectories and, specifically, tetraploidy is associated with higher rates of adaptation relative to haploidy and diploidy. While the majority of experimental evolution studies investigating ploidy use the budding yeast Saccharomyces cerivisiae, the fungal pathogen Candida albicans is a powerful system to investigate ploidy dynamics, particularly in the context of acquiring antifungal drug resistance. C. albicans laboratory and clinical strains are predominantly diploid, but have been isolated as haploid and polyploid. Here, we evolved diploid and tetraploid C. albicans for ~60 days in the antifungal drug caspofungin. Tetraploid-evolved lines adapted faster than diploid-evolved lines and reached higher levels of caspofungin resistance. While diploid-evolved lines generally maintained their initial genome size, tetraploid-evolved lines rapidly underwent genome-size reductions and did so prior to caspofungin adaptation. While clinical resistance was largely due to mutations in FKS1, these mutations were caused by substitutions in diploid, and indels in tetraploid isolates. Furthermore, fitness costs in the absence of drug selection were significantly less in tetraploid-evolved lines compared to the diploid-evolved lines. Taken together, this work supports a model of adaptation in which the tetraploid state is transient but its ability to rapidly transition ploidy states improves adaptive outcomes and may drive drug resistance in fungal pathogens.
Collapse
Affiliation(s)
- Ognenka Avramovska
- Department of Biology, Emory University, Atlanta, GA, United States
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Amanda C. Smith
- Department of Biology, Emory University, Atlanta, GA, United States
- Division of Viral Disease, CDC Foundation, Atlanta, GA, United States
| | - Emily Rego
- Department of Biology, Emory University, Atlanta, GA, United States
| | | |
Collapse
|
18
|
Kaur H, Kanaujia R, Singh S, Kajal K, Jayashree M, Peter NJ, Verma S, Gupta M, Ray P, Ghosh A, Samujh R, Rudramurthy SM. Clinical utility of time to positivity of blood cultures in cases of fungaemia: A prospective study. Indian J Med Microbiol 2022; 43:85-89. [PMID: 36153287 DOI: 10.1016/j.ijmmb.2022.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/18/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022]
Abstract
PURPOSE Fungaemia due to yeast is a major cause of morbidity and mortality in critically ill patients. Although, automated blood cultures have improved the time to diagnosis, very few studies have systematically evaluated the utility of blood culture time to positivity (TTP) of fungaemia in the clinical scenario. In this study, we evaluated the TTP for different yeast species to determine its clinical utility. MATERIAL AND METHODS A prospective study including 244 consecutive patients admitted to the adult (n = 76) and paediatric (n = 168) intensive care units (ICUs) was conducted between December 2017 through March 2019. The clinical and demographic characteristics, BACTEC blood culture results and TTP for yeast positive blood cultures were recorded for analysis. RESULTS A total of 244 patients with 357 episodes of candidaemia were enrolled during the study period. The TTP (mean ± SD) for all yeast species was 26.8 ± 23.6 h while it was significantly longer in paediatric than adult patients (30.5 ± 24.7 vs. 25.2 ± 22.9 h; p = <0.0001). Wickerhamomyces anomalus and Cyberlindnera jadinii (previously C. utilis) were exclusively isolated from paediatric population where W. anomalus demonstrated significantly longer TTP than C. jadinii. Among adult cases, C. albicans exhibited significantly longer TTP than C. tropicalis. In paediatric cases, >80% of C. tropicalis and C. utilis flagged positive in blood culture before 24 h while majority (65.9%) of W. anomalus isolates flagged positive later than 24 h. Similarly in adult samples, 63% of C. tropicalis isolates beeped positive before 24 h. CONCLUSION TTP for yeast may provide insight regarding the responsible yeast species before final identification among critical patients with candidaemia. Larger studies are warranted for evaluating clinical utility of TTP considering other complex factors like yeast burden, generation time, virulence and host factors, which may affect TTP.
Collapse
Affiliation(s)
- Harsimran Kaur
- Department of Medical Microbiology, PGIMER, Chandigarh, India.
| | | | - Shreya Singh
- Dr B.R. Ambedkar State Institute of Medical Sciences (AIMS Mohali), India
| | - Kamal Kajal
- Department of Anaesthesia and Intensive Care, PGIMER, Chandigarh, India
| | - Muralidharan Jayashree
- Department of Paediatric Medicine, Advanced Paediatric Centre, PGIMER, Chandigarh, India
| | - Nitin James Peter
- Department of Paediatric Surgery, Advanced Paediatric Centre, PGIMER, Chandigarh, India
| | - Shristi Verma
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Mantavya Gupta
- Precise Healthcare Clinical Laboratory, Sector 30, Chandigarh, India
| | - Pallab Ray
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Anup Ghosh
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Ram Samujh
- Department of Paediatric Surgery, Advanced Paediatric Centre, PGIMER, Chandigarh, India
| | | |
Collapse
|
19
|
Jenull S, Shivarathri R, Tsymala I, Penninger P, Trinh PC, Nogueira F, Chauhan M, Singh A, Petryshyn A, Stoiber A, Chowdhary A, Chauhan N, Kuchler K. Transcriptomics and Phenotyping Define Genetic Signatures Associated with Echinocandin Resistance in Candida auris. mBio 2022; 13:e0079922. [PMID: 35968956 PMCID: PMC9426441 DOI: 10.1128/mbio.00799-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Candida auris emerged as a human fungal pathogen only during the past decade. Remarkably, C. auris displays high degrees of genomic diversity and phenotypic plasticity, with four major clades causing hospital outbreaks with high mortality and morbidity rates. C. auris can show clinical resistance to all classes of antifungal drugs, including echinocandins that are usually recommended as first-line therapies for invasive candidiasis. Here, we exploit transcriptomics coupled with phenotypic profiling to characterize a set of clinical C. auris isolates displaying pronounced echinocandin resistance (ECN-R). A hot spot mutation in the echinocandin FKS1 target gene is present in all resistant isolates. Moreover, ECN-R strains share a core signature set of 362 genes differentially expressed in ECN-R isolates. Among others, mitochondrial gene expression and genes affecting cell wall function appear to be the most prominent, with the latter correlating well with enhanced adhesive traits, increased cell wall mannan content, and altered sensitivity to cell wall stress of ECN-R isolates. Moreover, ECN-R phenotypic signatures were also linked to pathogen recognition and interaction with immune cells. Hence, transcriptomics paired with phenotyping is a suitable tool to predict resistance and fitness traits as well as treatment outcomes in pathogen populations with complex phenotypic diversity. IMPORTANCE The surge in antimicrobial drug resistance in some bacterial and fungal pathogens constitutes a significant challenge to health care facilities. The emerging human fungal pathogen Candida auris has been particularly concerning, as isolates can display pan-antifungal resistance traits against all drugs, including echinocandins. However, the mechanisms underlying this phenotypic diversity remain poorly understood. We identify transcriptomic signatures in C. auris isolates resistant to otherwise fungicidal echinocandins. We identify a set of differentially expressed genes shared by resistant strains compared to unrelated susceptible isolates. Moreover, phenotyping demonstrates that resistant strains show distinct behaviors, with implications for host-pathogen interactions. Hence, this work provides a solid basis to identify the mechanistic links between antifungal multidrug resistance and fitness costs that affect the interaction of C. auris with host immune defenses.
Collapse
Affiliation(s)
- Sabrina Jenull
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Raju Shivarathri
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Irina Tsymala
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Philipp Penninger
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Phan-Canh Trinh
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Filomena Nogueira
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
- CCRI-St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Manju Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Ashutosh Singh
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Andriy Petryshyn
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anton Stoiber
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anuradha Chowdhary
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Neeraj Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Karl Kuchler
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| |
Collapse
|
20
|
Genomic Diversity across Candida auris Clinical Isolates Shapes Rapid Development of Antifungal Resistance
In Vitro
and
In Vivo. mBio 2022; 13:e0084222. [PMID: 35862787 PMCID: PMC9426540 DOI: 10.1128/mbio.00842-22] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Antifungal drug resistance and tolerance pose a serious threat to global public health. In the human fungal pathogen, Candida auris, resistance to triazole, polyene, and echinocandin antifungals is rising, resulting in multidrug resistant isolates. Here, we use genome analysis and in vitro evolution of 17 new clinical isolates of C. auris from clades I and IV to determine how quickly resistance mutations arise, the stability of resistance in the absence of drug, and the impact of genetic background on evolutionary trajectories. We evolved each isolate in the absence of drug as well as in low and high concentrations of fluconazole. In just three passages, we observed genomic and phenotypic changes including karyotype alterations, aneuploidy, acquisition of point mutations, and increases in MIC values within the populations. Fluconazole resistance was stable in the absence of drug, indicating little to no fitness cost associated with resistance. Importantly, two isolates substantially increased resistance to ≥256 μg/mL fluconazole. Multiple evolutionary pathways and mutations associated with increased fluconazole resistance occurred simultaneously within the same population. Strikingly, the subtelomeric regions of C. auris were highly dynamic as deletion of multiple genes near the subtelomeres occurred during the three passages in several populations. Finally, we discovered a mutator phenotype in a clinical isolate of C. auris. This isolate had elevated mutation rates compared to other isolates and acquired substantial resistance during evolution in vitro and in vivo supporting that the genetic background of clinical isolates can have a significant effect on evolutionary potential.
Collapse
|
21
|
Lee Y, Liston SD, Lee D, Robbins N, Cowen LE. Functional analysis of the Candida albicans kinome reveals Hrr25 as a regulator of antifungal susceptibility. iScience 2022; 25:104432. [PMID: 35663022 PMCID: PMC9160768 DOI: 10.1016/j.isci.2022.104432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/05/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022] Open
Abstract
Candida albicans is a leading cause of death due to systemic fungal infections. Poor patient outcomes are attributable to the limited number of antifungal classes and the increasing prevalence of drug resistance. Protein kinases have emerged as rewarding targets in the development of drugs for diverse diseases, yet kinases remain untapped in the quest for new antifungals. Here, we performed a comprehensive analysis of the C. albicans kinome to identify genes for which loss-of-function confers hypersensitivity to the two most widely deployed antifungals, echinocandins and azoles. Through this analysis, we found a role for the casein kinase 1 (CK1) homologue Hrr25 in regulating tolerance to both antifungals as well as target-mediated echinocandin resistance. Follow-up investigations established that Hrr25 regulates these responses through its interaction with the SBF transcription factor. Thus, we provide insights into the circuitry governing cellular responses to antifungals and implicate Hrr25 as a key mediator of drug resistance. Screening Candida albicans kinase mutants reveals 47 regulators of antifungal tolerance Hrr25 is important for growth and cell wall/membrane stress tolerance Hrr25 enables target-mediated echinocandin resistance Hrr25 interacts with the SBF transcription factor complex
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sean D Liston
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Dongyeob Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
22
|
Stevenson EM, Gaze WH, Gow NAR, Hart A, Schmidt W, Usher J, Warris A, Wilkinson H, Murray AK. Antifungal Exposure and Resistance Development: Defining Minimal Selective Antifungal Concentrations and Testing Methodologies. FRONTIERS IN FUNGAL BIOLOGY 2022; 3:918717. [PMID: 37746188 PMCID: PMC10512330 DOI: 10.3389/ffunb.2022.918717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 09/26/2023]
Abstract
This scoping review aims to summarise the current understanding of selection for antifungal resistance (AFR) and to compare and contrast this with selection for antibacterial resistance, which has received more research attention. AFR is an emerging global threat to human health, associated with high mortality rates, absence of effective surveillance systems and with few alternative treatment options available. Clinical AFR is well documented, with additional settings increasingly being recognised to play a role in the evolution and spread of AFR. The environment, for example, harbours diverse fungal communities that are regularly exposed to antifungal micropollutants, potentially increasing AFR selection risk. The direct application of effect concentrations of azole fungicides to agricultural crops and the incomplete removal of pharmaceutical antifungals in wastewater treatment systems are of particular concern. Currently, environmental risk assessment (ERA) guidelines do not require assessment of antifungal agents in terms of their ability to drive AFR development, and there are no established experimental tools to determine antifungal selective concentrations. Without data to interpret the selective risk of antifungals, our ability to effectively inform safe environmental thresholds is severely limited. In this review, potential methods to generate antifungal selective concentration data are proposed, informed by approaches used to determine antibacterial minimal selective concentrations. Such data can be considered in the development of regulatory guidelines that aim to reduce selection for AFR.
Collapse
Affiliation(s)
- Emily M. Stevenson
- European Centre for Environment and Human Health, University of Exeter Medical School, Cornwall, United Kingdom
- Environment and Sustainability Institute, University of Exeter Medical School, Cornwall, United Kingdom
| | - William H. Gaze
- European Centre for Environment and Human Health, University of Exeter Medical School, Cornwall, United Kingdom
- Environment and Sustainability Institute, University of Exeter Medical School, Cornwall, United Kingdom
| | - Neil A. R. Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Alwyn Hart
- Chief Scientist’s Group, Environment Agency, Horizon House, Bristol, England, United Kingdom
| | - Wiebke Schmidt
- Chief Scientist’s Group, Environment Agency, Horizon House, Bristol, England, United Kingdom
| | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Helen Wilkinson
- Chief Scientist’s Group, Environment Agency, Horizon House, Bristol, England, United Kingdom
| | - Aimee K. Murray
- European Centre for Environment and Human Health, University of Exeter Medical School, Cornwall, United Kingdom
- Environment and Sustainability Institute, University of Exeter Medical School, Cornwall, United Kingdom
| |
Collapse
|
23
|
High-Throughput Chemical Screen Identifies a 2,5-Disubstituted Pyridine as an Inhibitor of Candida albicans Erg11. mSphere 2022; 7:e0007522. [PMID: 35531664 PMCID: PMC9241532 DOI: 10.1128/msphere.00075-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Fungal infections contribute to over 1.5 million deaths annually, with Candida albicans representing one of the most concerning human fungal pathogens. While normally commensal in nature, compromise of host immunity can result in C. albicans disseminating into the human bloodstream, causing infections with mortality rates of up to 40%. A contributing factor to this high mortality rate is the limited arsenal of antifungals approved to treat systemic infections. The most widely used antifungal class, the azoles, inhibits ergosterol biosynthesis by targeting Erg11. The rise of drug resistance among C. albicans clinical isolates, particularly against the azoles, has escalated the need to explore novel antifungal strategies. To address this challenge, we screened a 9,600-compound subset of the University of Tokyo Core Chemical Library to identify molecules with novel antifungal activity against C. albicans. The most potent hit molecule was CpdLC-6888, a 2,5-disubstituted pyridine compound, which inhibited growth of C. albicans and closely-related species. Chemical-genetic, biochemical, and modeling analyses suggest that CpdLC-6888 inhibits Erg11 in a manner similar to the azoles despite lacking the canonical five-membered nitrogen-containing azole ring. This work characterizes the antifungal activity of a 2,5-disubstituted pyridine against C. albicans, supporting the mining of existing chemical collections to identify compounds with novel antifungal activity. IMPORTANCE Pathogenic fungi represent a serious but underacknowledged threat to human health. The treatment and management of these infections relies heavily on the use of azole antifungals, a class of molecules that contain a five-membered nitrogen-containing ring and inhibit the biosynthesis of the key membrane sterol ergosterol. By employing a high-throughput chemical screen, we identified a 2,5-disubstituted pyridine, termed CpdLC-6888, as possessing antifungal activity against the prominent human fungal pathogen Candida albicans. Upon further investigation, we determined this molecule exhibits azole-like activity despite being structurally divergent. Specifically, transcriptional repression of the azole target gene ERG11 resulted in hypersensitivity to CpdLC-6888, and treatment of C. albicans with this molecule blocked the production of the key membrane sterol ergosterol. Therefore, this work describes a chemical scaffold with novel antifungal activity against a prevalent and threatening fungal pathogen affecting human health, expanding the repertoire of compounds that can inhibit this useful antifungal drug target.
Collapse
|
24
|
Iyer KR, Robbins N, Cowen LE. The role of Candida albicans stress response pathways in antifungal tolerance and resistance. iScience 2022; 25:103953. [PMID: 35281744 PMCID: PMC8905312 DOI: 10.1016/j.isci.2022.103953] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human fungal pathogens are the causative agents of devastating diseases across the globe, and the increasing prevalence of drug resistance threatens to undermine the already limited treatment options. One prominent pathogen is the opportunistic fungus Candida albicans, which can cause both superficial and serious systemic infections in immunocompromised individuals. C. albicans antifungal drug resistance and antifungal tolerance are supported by diverse and expansive cellular stress response pathways. Some of the major players are the Ca2+-calmodulin-activated phosphatase calcineurin, the protein kinase C cell wall integrity pathway, and the molecular chaperone heat shock protein 90. Beyond these core signal transducers, several other enzymes and transcription factors have been implicated in both tolerance and resistance. Here, we highlight some of the major stress response pathways, key advances in identifying chemical matter to inhibit these pathways, and implications for C. albicans persistence in the host. Candida albicans can cause superficial and serious systemic infections in humans Stress response pathways regulate C. albicans antifungal resistance and tolerance Stress response regulators include calcineurin, Pkc1, Hsp90, and many others Stress response inhibitors could reduce the likelihood of fungi persisting in humans
Collapse
Affiliation(s)
- Kali R. Iyer
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, West Tower, Room 1638, Toronto, ON M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, West Tower, Room 1638, Toronto, ON M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, West Tower, Room 1638, Toronto, ON M5G 1M1, Canada
- Corresponding author
| |
Collapse
|
25
|
Zeng G, Xu X, Gao J, da Silva Dantas A, Gow NA, Wang Y. Inactivating the mannose-ethanolamine phosphotransferase Gpi7 confers caspofungin resistance in the human fungal pathogen Candida albicans. Cell Surf 2021; 7:100057. [PMID: 34258484 PMCID: PMC8254124 DOI: 10.1016/j.tcsw.2021.100057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022] Open
Abstract
Understanding the molecular mechanisms governing antifungal resistance is crucial for identifying new cellular targets for developing new antifungal therapeutics. In this study, we performed a transposon-mediated genome-wide genetic screen in haploid Candida albicans to identify mutants resistant to caspofungin, the first member of the echinocandin class of antifungal drugs. A mutant exhibiting the highest resistance possessed a transposon insertion that inactivates GPI7, a gene encoding the mannose-ethanolamine phosphotransferase. Deleting GPI7 in diploid C. albicans caused similar caspofungin resistance. gpi7Δ/Δ cells showed significantly elevated cell wall chitin content and enhanced phosphorylation of Mkc1, a core component of the PKC-MAPK cell-wall integrity pathway. Deleting MKC1 suppressed the chitin elevation and caspofungin resistance of gpi7Δ/Δ cells, but overexpressing the dominant inactive form of RHO1, an upstream activator of PKC-MAPK signaling, did not. Transcriptome analysis uncovered 406 differentially expressed genes in gpi7Δ/Δ cells, many related to cell wall construction. Our results suggest that GPI7 deletion impairs cell wall integrity, which triggers the cell-wall salvage mechanism via the PKC-MAPK pathway independently of Rho1, resulting in the compensatory chitin synthesis to confer caspofungin resistance.
Collapse
Affiliation(s)
- Guisheng Zeng
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Xiaoli Xu
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Jiaxin Gao
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Alessandra da Silva Dantas
- MRC Centre for Medical Mycology, School of Biosciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Neil A.R. Gow
- MRC Centre for Medical Mycology, School of Biosciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Yue Wang
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
26
|
Li W, Shrivastava M, Lu H, Jiang Y. Calcium-calcineurin signaling pathway in Candida albicans: A potential drug target. Microbiol Res 2021; 249:126786. [PMID: 33989979 DOI: 10.1016/j.micres.2021.126786] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 12/26/2022]
Abstract
Increased morbidity and mortality of candidiasis are a notable threat to the immunocompromised patients. At present, the types of drugs available to treat C. albicans infection are relatively limited. Moreover, the emergence of antifungal drug resistance of C. albicans makes the treatment of C. albicans infection more difficult. The calcium-calcineurin signaling pathway plays a crucial role in the survival and pathogenicity of C. albicans and may act as a potential target against C. albicans. In this review, we summarized functions of the calcium-calcineurin signaling pathway in several biological processes, compared the differences of this signaling pathway between C. albicans and humans, and described anti-C. albicans activity of inhibitors of this signaling pathway. We believe that targeting the calcium-calcineurin signaling pathway is a promising strategy to cope with C. albicans infection.
Collapse
Affiliation(s)
- Wanqian Li
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Hui Lu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Dutta A, Hartmann FE, Francisco CS, McDonald BA, Croll D. Mapping the adaptive landscape of a major agricultural pathogen reveals evolutionary constraints across heterogeneous environments. THE ISME JOURNAL 2021; 15:1402-1419. [PMID: 33452474 PMCID: PMC8115182 DOI: 10.1038/s41396-020-00859-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
The adaptive potential of pathogens in novel or heterogeneous environments underpins the risk of disease epidemics. Antagonistic pleiotropy or differential resource allocation among life-history traits can constrain pathogen adaptation. However, we lack understanding of how the genetic architecture of individual traits can generate trade-offs. Here, we report a large-scale study based on 145 global strains of the fungal wheat pathogen Zymoseptoria tritici from four continents. We measured 50 life-history traits, including virulence and reproduction on 12 different wheat hosts and growth responses to several abiotic stressors. To elucidate the genetic basis of adaptation, we used genome-wide association mapping coupled with genetic correlation analyses. We show that most traits are governed by polygenic architectures and are highly heritable suggesting that adaptation proceeds mainly through allele frequency shifts at many loci. We identified negative genetic correlations among traits related to host colonization and survival in stressful environments. Such genetic constraints indicate that pleiotropic effects could limit the pathogen's ability to cause host damage. In contrast, adaptation to abiotic stress factors was likely facilitated by synergistic pleiotropy. Our study illustrates how comprehensive mapping of life-history trait architectures across diverse environments allows to predict evolutionary trajectories of pathogens confronted with environmental perturbations.
Collapse
Affiliation(s)
- Anik Dutta
- grid.5801.c0000 0001 2156 2780Plant Pathology, Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Fanny E. Hartmann
- grid.5801.c0000 0001 2156 2780Plant Pathology, Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland ,grid.417885.70000 0001 2185 8223Ecologie Systématique Evolution, CNRS, Université Paris-Saclay, AgroParisTech, 91400 Orsay, France
| | - Carolina Sardinha Francisco
- grid.5801.c0000 0001 2156 2780Plant Pathology, Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland ,Present Address: Environmental Genomics Group, Botanical Institute, CAU Kiel, Germany
| | - Bruce A. McDonald
- grid.5801.c0000 0001 2156 2780Plant Pathology, Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Daniel Croll
- grid.10711.360000 0001 2297 7718Laboratory of Evolutionary Genetics, Institute of Biology, University of Neuchâtel, 2000 Neuchâtel, Switzerland
| |
Collapse
|
28
|
Iyer KR, Camara K, Daniel-Ivad M, Trilles R, Pimentel-Elardo SM, Fossen JL, Marchillo K, Liu Z, Singh S, Muñoz JF, Kim SH, Porco JA, Cuomo CA, Williams NS, Ibrahim AS, Edwards JE, Andes DR, Nodwell JR, Brown LE, Whitesell L, Robbins N, Cowen LE. An oxindole efflux inhibitor potentiates azoles and impairs virulence in the fungal pathogen Candida auris. Nat Commun 2020; 11:6429. [PMID: 33353950 PMCID: PMC7755909 DOI: 10.1038/s41467-020-20183-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Candida auris is an emerging fungal pathogen that exhibits resistance to multiple drugs, including the most commonly prescribed antifungal, fluconazole. Here, we use a combinatorial screening approach to identify a bis-benzodioxolylindolinone (azoffluxin) that synergizes with fluconazole against C. auris. Azoffluxin enhances fluconazole activity through the inhibition of efflux pump Cdr1, thus increasing intracellular fluconazole levels. This activity is conserved across most C. auris clades, with the exception of clade III. Azoffluxin also inhibits efflux in highly azole-resistant strains of Candida albicans, another human fungal pathogen, increasing their susceptibility to fluconazole. Furthermore, azoffluxin enhances fluconazole activity in mice infected with C. auris, reducing fungal burden. Our findings suggest that pharmacologically targeting Cdr1 in combination with azoles may be an effective strategy to control infection caused by azole-resistant isolates of C. auris.
Collapse
Affiliation(s)
- Kali R Iyer
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Kaddy Camara
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
- Clark+Elbing LLP, Boston, MA, USA
| | | | - Richard Trilles
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | | | - Jen L Fossen
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Karen Marchillo
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Zhongle Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Shakti Singh
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
| | - José F Muñoz
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sang Hu Kim
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - John A Porco
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Ashraf S Ibrahim
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - John E Edwards
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - David R Andes
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Justin R Nodwell
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren E Brown
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
29
|
Experimental Evolution Identifies Adaptive Aneuploidy as a Mechanism of Fluconazole Resistance in Candida auris. Antimicrob Agents Chemother 2020; 65:AAC.01466-20. [PMID: 33077664 DOI: 10.1128/aac.01466-20] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Candida auris is a newly emerging fungal pathogen of humans and has attracted considerable attention from both the clinical and basic research communities. Clinical isolates of C. auris are often resistant to one or more antifungal agents. To explore how antifungal resistance develops, we performed experimental evolution assays using a fluconazole-susceptible isolate of C. auris (BJCA001). After a series of passages through medium containing increasing concentrations of fluconazole, fungal cells acquired resistance. By sequencing and comparing the genomes of the parental fluconazole-susceptible strain and 26 experimentally evolved strains of C. auris, we found that a portion of fluconazole-resistant strains carried one extra copy of chromosome V. In the absence of fluconazole, C. auris cells rapidly became susceptible and lost the extra copy of chromosome V. Genomic and transcriptome sequencing (RNA-Seq) analyses indicate that this chromosome carries a number of drug resistance-related genes, which were transcriptionally upregulated in the resistant, aneuploid strains. Moreover, missense mutations were identified in the genes TAC1B, RRP6, and SFT2 in all experimentally evolved strains. Our findings suggest that the gain of an extra copy of chromosome V is associated with the rapid acquisition of fluconazole resistance and may represent an important evolutionary mechanism of antifungal resistance in C. auris.
Collapse
|
30
|
Vallières C, Singh N, Alexander C, Avery SV. Repurposing Nonantifungal Approved Drugs for Synergistic Targeting of Fungal Pathogens. ACS Infect Dis 2020; 6:2950-2958. [PMID: 33141557 DOI: 10.1021/acsinfecdis.0c00405] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With the spread of drug resistance, new antimicrobials are urgently needed. Here, we set out to tackle this problem by high-throughput exploration for novel antifungal synergies among combinations of approved, nonantifungal drugs; a novel strategy exploiting the potential of alternative targets, low chemicals usage and low development risk. We screened the fungal pathogen Candida albicans by combining a small panel of nonantifungal drugs (all in current use for other clinical applications) with 1280 compounds from an approved drug library. Screens at sublethal concentrations of the antibiotic paromomycin (PM), the antimalarial primaquine (PQ), or the anti-inflammatory drug ibuprofen (IF) revealed a total of 17 potential strong, synergistic interactions with the library compounds. Susceptibility testing with the most promising combinations corroborated marked synergies [fractional inhibitory concentration (FIC) indices ≤0.5] between PM + β-escin, PQ + celecoxib, and IF + pentamidine, reducing the MICs of PM, PQ, and IF in C. albicans by >64-, 16-, and 8-fold, respectively. Paromomycin + β-escin and PQ + celecoxib were effective also against C. albicans biofilms, azole-resistant clinical isolates, and other fungal pathogens. Actions were specific, as no synergistic effect was observed in mammalian cells. Mode of action was investigated for one of the combinations, revealing that PM + β-escin synergistically increase the error-rate of mRNA translation and suggesting a different molecular target to current antifungals. The study unveils the potential of the described combinatorial strategy in enabling acceleration of drug-repurposing discovery for combatting fungal pathogens.
Collapse
Affiliation(s)
- Cindy Vallières
- School of Life Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
| | - Nishant Singh
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
| | - Cameron Alexander
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
| | - Simon V. Avery
- School of Life Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
| |
Collapse
|
31
|
Arastehfar A, Lass-Flörl C, Garcia-Rubio R, Daneshnia F, Ilkit M, Boekhout T, Gabaldon T, Perlin DS. The Quiet and Underappreciated Rise of Drug-Resistant Invasive Fungal Pathogens. J Fungi (Basel) 2020; 6:E138. [PMID: 32824785 PMCID: PMC7557958 DOI: 10.3390/jof6030138] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/22/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Human fungal pathogens are attributable to a significant economic burden and mortality worldwide. Antifungal treatments, although limited in number, play a pivotal role in decreasing mortality and morbidities posed by invasive fungal infections (IFIs). However, the recent emergence of multidrug-resistant Candida auris and Candida glabrata and acquiring invasive infections due to azole-resistant C. parapsilosis, C. tropicalis, and Aspergillus spp. in azole-naïve patients pose a serious health threat considering the limited number of systemic antifungals available to treat IFIs. Although advancing for major fungal pathogens, the understanding of fungal attributes contributing to antifungal resistance is just emerging for several clinically important MDR fungal pathogens. Further complicating the matter are the distinct differences in antifungal resistance mechanisms among various fungal species in which one or more mechanisms may contribute to the resistance phenotype. In this review, we attempt to summarize the burden of antifungal resistance for selected non-albicansCandida and clinically important Aspergillus species together with their phylogenetic placement on the tree of life. Moreover, we highlight the different molecular mechanisms between antifungal tolerance and resistance, and comprehensively discuss the molecular mechanisms of antifungal resistance in a species level.
Collapse
Affiliation(s)
- Amir Arastehfar
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Rocio Garcia-Rubio
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
| | - Farnaz Daneshnia
- Westerdijk Fungal Biodiversity Institute, 3584 CT Utrecht, The Netherlands; (F.D.); (T.B.)
| | - Macit Ilkit
- Division of Mycology, University of Çukurova, 01330 Adana, Turkey;
| | - Teun Boekhout
- Westerdijk Fungal Biodiversity Institute, 3584 CT Utrecht, The Netherlands; (F.D.); (T.B.)
- Institute of Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, 1012 WX Amsterdam, The Netherlands
| | - Toni Gabaldon
- Life Sciences Programme, Barcelona, Supercomputing Center (BSC-CNS), Jordi Girona, 08034 Barcelona, Spain;
- Mechanisms of Disease Programme, Institute for Research in Biomedicine (IRB), 08024 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
| |
Collapse
|
32
|
Chavhan Y, Malusare S, Dey S. Larger bacterial populations evolve heavier fitness trade-offs and undergo greater ecological specialization. Heredity (Edinb) 2020; 124:726-736. [PMID: 32203249 DOI: 10.1038/s41437-020-0308-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 11/09/2022] Open
Abstract
Evolutionary studies over the last several decades have invoked fitness trade-offs to explain why species prefer some environments to others. However, the effects of population size on trade-offs and ecological specialization remain largely unknown. To complicate matters, trade-offs themselves have been visualized in multiple ways in the literature. Thus, it is not clear how population size can affect the various aspects of trade-offs. To address these issues, we conducted experimental evolution with Escherichia coli populations of two different sizes in two nutritionally limited environments, and studied fitness trade-offs from three different perspectives. We found that larger populations evolved greater fitness trade-offs, regardless of how trade-offs are conceptualized. Moreover, although larger populations adapted more to their selection conditions, they also became more maladapted to other environments, ultimately paying heavier costs of adaptation. To enhance the generalizability of our results, we further investigated the evolution of ecological specialization across six different environmental pairs, and found that larger populations specialized more frequently and evolved consistently steeper reaction norms of fitness. This is the first study to demonstrate a relationship between population size and fitness trade-offs, and the results are important in understanding the population genetics of ecological specialization and vulnerability to environmental changes.
Collapse
Affiliation(s)
- Yashraj Chavhan
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Sarthak Malusare
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India.,Gaia Doctoral School, Institut des Sciences de l'Evolution (ISEM), 1093-1317 Route de Mende, 34090, Montpellier, France
| | - Sutirth Dey
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India.
| |
Collapse
|
33
|
Global Screening of Genomic and Transcriptomic Factors Associated with Phenotype Differences between Multidrug-Resistant and -Susceptible Candida haemulonii Strains. mSystems 2019; 4:4/6/e00459-19. [PMID: 31848301 PMCID: PMC6918027 DOI: 10.1128/msystems.00459-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A comprehensive, multi-omic survey was performed to disclose the genetic backgrounds and differences between multidrug-resistant and -susceptible C. haemulonii strains. Genes were identified with mutations or significant expression differences in multidrug-resistant compared to multidrug-susceptible strains, which were mainly involved in multidrug resistance, stress fitness, and morphology. The Cdr1-encoding gene, C. haemulonii2486 (CH_2486), was expressed at a significantly increased level in multidrug-resistant strains. Functional inhibition experiments further implicated potential roles of CH_2486 in drug resistance. A gene spontaneously mutated in resistant strains, CH_4347, was experimentally validated to influence the morphology of spores, possibly by controlling cell wall integrity. Candida haemulonii, a close relative of Candida auris, is an emerging pathogen which frequently shows multidrug resistance especially to triazoles, the most used antifungal drugs. The mechanisms of drug resistance in C. haemulonii, however, are largely unknown. Here, we sequenced and annotated the genomes of two reference strains from the C. haemulonii complex, compared the phenotypes, genomes, and transcriptomes of a triazole-susceptible and two triazole-resistant C. haemulonii strains, and identified triazole susceptibility, morphology, fitness, and the major genetic and gene expression differences between the strains. A multidrug efflux gene, CDR1, was recurrently found to be upregulated for expression in triazole-resistant strains. Blocking the activity of Cdr1 increased the susceptibility to triazoles strikingly. Comparative transcriptome analysis also demonstrated impaired cell wall integrity, filamentous growth, and iron homeostasis in triazole-resistant strains. Finally, we also identified a zinc-binding MHR family transcription regulator gene that mutated in triazole-resistant strains spontaneously, contributing to the changes of morphology and, possibly, cell wall integrity between the strains. The study provided important clues for future studies exploring the mechanisms of multidrug resistance and related phenotypic differences of C. haemulonii strains. IMPORTANCE A comprehensive, multi-omic survey was performed to disclose the genetic backgrounds and differences between multidrug-resistant and -susceptible C. haemulonii strains. Genes were identified with mutations or significant expression differences in multidrug-resistant compared to multidrug-susceptible strains, which were mainly involved in multidrug resistance, stress fitness, and morphology. The Cdr1-encoding gene, C. haemulonii2486 (CH_2486), was expressed at a significantly increased level in multidrug-resistant strains. Functional inhibition experiments further implicated potential roles of CH_2486 in drug resistance. A gene spontaneously mutated in resistant strains, CH_4347, was experimentally validated to influence the morphology of spores, possibly by controlling cell wall integrity.
Collapse
|
34
|
de Castro Spadari C, da Silva de Bastiani FWM, Pisani PBB, de Azevedo Melo AS, Ishida K. Efficacy of voriconazole in vitro and in invertebrate model of cryptococcosis. Arch Microbiol 2019; 202:773-784. [PMID: 31832690 DOI: 10.1007/s00203-019-01789-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/11/2019] [Accepted: 12/03/2019] [Indexed: 11/30/2022]
Abstract
Cryptococcosis is a common opportunistic infection in patients with advanced HIV infection and may also affect immunocompetent patients. The available antifungal agents are few and other options are needed for the cryptococcosis treatment. In this work, we first analyzed the virulence of twelve C. neoformans and C. gattii strains assessing capsule thickness, biofilms formation, and survival and morbidity in the invertebrate model of Galleria mellonella and then we evaluated the antifungal activity of voriconazole (VRC) in vitro and in vivo also using G. mellonella. Our results showed that all Cryptococcus spp. isolates were able to produce capsule and biofilms, and were virulent using G. mellonella model. The VRC has inhibitory activity on planktonic cells with MIC values ranging from 0.03 to 0.25 μg/mL on Cryptococcus spp.; and these isolates were more tolerant to fluconazole (ranging from 0.25 to 16 μg/mL), the triazol agent often recommended alone or in combination with amphotericin B in the cryptococcosis therapy. In contrast, mature biofilms were less susceptible to the VRC treatment. The VRC (10 or 20 mg/kg) treatment of infected G. mellonella larvae significantly increased the larval survival when compared to the untreated group for the both Cryptococcus species and significantly decreased the fungal burden and dissemination in the larval tissue. Our findings corroborate with the literature data, supporting the potential use of VRC as an alternative for cryptococcosis treatment. Here, we emphasize the use of G. mellonella larval model as an alternative animal model for studies of antifungal efficacy on mycosis, including cryptococcosis.
Collapse
Affiliation(s)
- Cristina de Castro Spadari
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Prof. Lineu Prestes Avenue, 1374, ICB II, Lab 150, São Paulo, SP, 05508-000, Brazil
| | - Fernanda Walt Mendes da Silva de Bastiani
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Prof. Lineu Prestes Avenue, 1374, ICB II, Lab 150, São Paulo, SP, 05508-000, Brazil
| | - Pietro Bruno Bautista Pisani
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Prof. Lineu Prestes Avenue, 1374, ICB II, Lab 150, São Paulo, SP, 05508-000, Brazil
| | | | - Kelly Ishida
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Prof. Lineu Prestes Avenue, 1374, ICB II, Lab 150, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
35
|
Abstract
Natural or synthetic genetic modules can lose their function over long-term evolution if the function is costly. How populations can evolve to restore such broken function is poorly understood. To test the reversibility of evolutionary breakdown, we use yeast cell populations with a chromosomally integrated synthetic gene circuit. In previous evolution experiments the gene circuit lost its costly function through various mutations. By exposing such mutant populations to conditions where regaining gene circuit function would be beneficial, we find adaptation scenarios with or without repairing lost gene circuit function. These results are important for drug resistance or future synthetic biology applications where evolutionary loss and regain of function play a significant role. Evolutionary reversibility—the ability to regain a lost function—is an important problem both in evolutionary and synthetic biology, where repairing natural or synthetic systems broken by evolutionary processes may be valuable. Here, we use a synthetic positive-feedback (PF) gene circuit integrated into haploid Saccharomyces cerevisiae cells to test if the population can restore lost PF function. In previous evolution experiments, mutations in a gene eliminated the fitness costs of PF activation. Since PF activation also provides drug resistance, exposing such compromised or broken mutants to both drug and inducer should create selection pressure to regain drug resistance and possibly PF function. Indeed, evolving 7 PF mutant strains in the presence of drug revealed 3 adaptation scenarios through genomic, PF-external mutations that elevate PF basal expression, possibly by affecting transcription, translation, degradation, and other fundamental cellular processes. Nonfunctional mutants gained drug resistance without ever developing high expression, while quasifunctional and dysfunctional PF mutants developed high expression nongenetically, which then diminished, although more slowly for dysfunctional mutants where revertant clones arose. These results highlight how intracellular context, such as the growth rate, can affect regulatory network dynamics and evolutionary dynamics, which has important consequences for understanding the evolution of drug resistance and developing future synthetic biology applications.
Collapse
|
36
|
James TY, Michelotti LA, Glasco AD, Clemons RA, Powers RA, James ES, Simmons DR, Bai F, Ge S. Adaptation by Loss of Heterozygosity in Saccharomyces cerevisiae Clones Under Divergent Selection. Genetics 2019; 213:665-683. [PMID: 31371407 PMCID: PMC6781901 DOI: 10.1534/genetics.119.302411] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/29/2019] [Indexed: 01/14/2023] Open
Abstract
Loss of heterozygosity (LOH) is observed during vegetative growth and reproduction of diploid genotypes through mitotic crossovers, aneuploidy caused by nondisjunction, and gene conversion. We aimed to test the role that LOH plays during adaptation of two highly heterozygous Saccharomyces cerevisiae genotypes to multiple environments over a short time span in the laboratory. We hypothesized that adaptation would be observed through parallel LOH events across replicate populations. Using genome resequencing of 70 clones, we found that LOH was widespread with 5.2 LOH events per clone after ∼500 generations. The most common mode of LOH was gene conversion (51%) followed by crossing over consistent with either break-induced replication or double Holliday junction resolution. There was no evidence that LOH involved nondisjunction of whole chromosomes. We observed parallel LOH in both an environment-specific and environment-independent manner. LOH largely involved recombining existing variation between the parental genotypes, but also was observed after de novo, presumably beneficial, mutations occurred in the presence of canavanine, a toxic analog of arginine. One highly parallel LOH event involved the ENA salt efflux pump locus on chromosome IV, which showed repeated LOH to the allele from the European parent, an allele originally derived by introgression from S. paradoxus Using CRISPR-engineered LOH we showed that the fitness advantage provided by this single LOH event was 27%. Overall, we found extensive evidence that LOH could be adaptive and is likely to be a greater source of initial variation than de novo mutation for rapid evolution of diploid genotypes.
Collapse
Affiliation(s)
- Timothy Y James
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Lucas A Michelotti
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Alexander D Glasco
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Rebecca A Clemons
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Robert A Powers
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Ellen S James
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - D Rabern Simmons
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Fengyan Bai
- Institute of Microbiology, Chinese Academy of Sciences, State Key Laboratory of Mycology, Chaoyang District, Beijing 100101, China
| | - Shuhua Ge
- Technology Development and Transfer Center, Institute of Microbiology, Chinese Academy of Sciences, Chaoyang District, Beijing 100029, China
| |
Collapse
|
37
|
Genetic Analysis of Candida auris Implicates Hsp90 in Morphogenesis and Azole Tolerance and Cdr1 in Azole Resistance. mBio 2019; 10:mBio.02529-18. [PMID: 30696744 PMCID: PMC6355988 DOI: 10.1128/mbio.02529-18] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida auris is an emerging fungal pathogen and a serious global health threat as the majority of clinical isolates display elevated resistance to currently available antifungal drugs. Despite the increased prevalence of C. auris infections, the mechanisms governing drug resistance remain largely elusive. In diverse fungi, the evolution of drug resistance is enabled by the essential molecular chaperone Hsp90, which stabilizes key regulators of cellular responses to drug-induced stress. Hsp90 also orchestrates temperature-dependent morphogenesis in Candida albicans, a key virulence trait. However, the role of Hsp90 in the pathobiology of C. auris remains unknown. In order to study regulatory functions of Hsp90 in C. auris, we placed HSP90 under the control of a doxycycline-repressible promoter to enable transcriptional repression. We found that Hsp90 is essential for growth in C. auris and that it enables tolerance of clinical isolates with respect to the azoles, which inhibit biosynthesis of the membrane sterol ergosterol. High-level azole resistance was independent of Hsp90 but dependent on the ABC transporter CDR1, deletion of which resulted in abrogated resistance. Strikingly, we discovered that C. auris undergoes a morphogenetic transition from yeast to filamentous growth in response to HSP90 depletion or cell cycle arrest but not in response to other cues that induce C. albicans filamentation. Finally, we observed that this developmental transition is associated with global transcriptional changes, including the induction of cell wall-related genes. Overall, this report provides a novel insight into mechanisms of drug tolerance and resistance in C. auris and describes a developmental transition in response to perturbation of a core regulator of protein homeostasis.IMPORTANCE Fungal pathogens pose a serious threat to public health. Candida auris is an emerging fungal pathogen that is often resistant to commonly used antifungal drugs. However, the mechanisms governing drug resistance and virulence in this organism remain largely unexplored. In this study, we adapted a conditional expression system to modulate the transcription of an essential gene, HSP90, which regulates antifungal resistance and virulence in diverse fungal pathogens. We showed that Hsp90 is essential for growth in C. auris and is important for tolerance of the clinically important azole antifungals, which block ergosterol biosynthesis. Further, we established that the Cdr1 efflux transporter regulates azole resistance. Finally, we discovered that C. auris transitions from yeast to filamentous growth in response to Hsp90 inhibition, accompanied by global transcriptional remodeling. Overall, this work provides a novel insight into mechanisms regulating azole resistance in C. auris and uncovers a distinct developmental program regulated by Hsp90.
Collapse
|
38
|
Chew KL, Octavia S, Lin RTP, Yan GZ, Teo JWP. Delay in effective therapy in anidulafungin-resistant Candida tropicalis fungaemia: Potential for rapid prediction of antifungal resistance with whole-genome-sequencing. J Glob Antimicrob Resist 2018; 16:105-107. [PMID: 30583013 DOI: 10.1016/j.jgar.2018.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES This study investigated the feasibility of using whole-genome sequencing (WGS) for the prediction of antifungal resistance in anidulafungin-resistant Candida tropicalis candidaemia isolates. METHODS Next-generation sequencing was performed for three anidulafungin-resistant C. tropicalis isolates on an Illumina MiSeq system with in-house bioinformatics analysis. RESULTS Mutations in Fks1p associated with anidulafungin resistance were identified. Other mutations associated with varying levels of phenotypic resistance to fluconazole were also identified. CONCLUSIONS These data demonstrate the potential to predict antifungal resistance using WGS. With improving technology, real-time WGS may be used for tailoring effective antifungal therapy in patients with candidaemia.
Collapse
Affiliation(s)
- Ka Lip Chew
- Department of Laboratory Medicine, National University Hospital, Singapore.
| | - Sophie Octavia
- National Public Health Laboratory, National Centre for Infectious Diseases, Singapore
| | - Raymond Tzer Pin Lin
- Department of Laboratory Medicine, National University Hospital, Singapore; National Public Health Laboratory, National Centre for Infectious Diseases, Singapore
| | | | | |
Collapse
|
39
|
Vallières C, Raulo R, Dickinson M, Avery SV. Novel Combinations of Agents Targeting Translation That Synergistically Inhibit Fungal Pathogens. Front Microbiol 2018; 9:2355. [PMID: 30349511 PMCID: PMC6186996 DOI: 10.3389/fmicb.2018.02355] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/14/2018] [Indexed: 12/29/2022] Open
Abstract
A range of fungicides or antifungals are currently deployed to control fungi in agriculture or medicine, but resistance to current agents is growing so new approaches and molecular targets are urgently needed. Recently, different aminoglycoside antibiotics combined with particular transport inhibitors were found to produce strong, synergistic growth-inhibition of fungi, by synergistically increasing the error rate of mRNA translation. Here, focusing on translation fidelity as a novel target for combinatorial antifungal treatment, we tested the hypothesis that alternative combinations of agents known to affect the availability of functional amino acids would synergistically inhibit growth of major fungal pathogens. We screened 172 novel combinations against three phytopathogens (Rhizoctonia solani, Zymoseptoria tritici, and Botrytis cinerea) and three human pathogens (Cryptococcus neoformans, Candida albicans, and Aspergillus fumigatus), showing that 48 combinations inhibited strongly the growth of the pathogens; the growth inhibition effect was significantly greater with the agents combined than by a simple product of their individual effects at the same doses. Of these, 23 combinations were effective against more than one pathogen, including combinations comprising food-and-drug approved compounds, e.g., quinine with bicarbonate, and quinine with hygromycin. These combinations [fractional inhibitory combination (FIC) index ≤0.5] gave up to 100% reduction of fungal growth yield at concentrations of agents which, individually, had negligible effect. No synergy was evident against bacterial, plant or mammalian cells, indicating specificity for fungi. Mode-of-action analyses for quinine + hygromycin indicated that synergistic mistranslation was the antifungal mechanism. That mechanism was not universal as bicarbonate exacerbated quinine action by increasing drug uptake. The study unveils chemical combinations and a target process with potential for control of diverse fungal pathogens, and suggests repurposing possibilities for several current therapeutics.
Collapse
Affiliation(s)
- Cindy Vallières
- School of Life Sciences, University of Nottingham, University Park Campus, Nottingham, United Kingdom
| | - Roxane Raulo
- School of Life Sciences, University of Nottingham, University Park Campus, Nottingham, United Kingdom
| | - Matthew Dickinson
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Simon V Avery
- School of Life Sciences, University of Nottingham, University Park Campus, Nottingham, United Kingdom
| |
Collapse
|
40
|
Abstract
The fungal pathogens Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus have transitioned from a rare curiosity to a leading cause of human mortality. The management of infections caused by these organisms is intimately dependent on the efficacy of antifungal agents; however, fungi that are resistant to these treatments are regularly isolated in the clinic, impeding our ability to control infections. Given the significant impact fungal pathogens have on human health, it is imperative to understand the molecular mechanisms that govern antifungal drug resistance. This review describes our current knowledge of the mechanisms by which antifungal drug resistance evolves in experimental populations and clinical settings. We explore current antifungal treatment options and discuss promising strategies to impede the evolution of drug resistance. By tackling antifungal drug resistance as an evolutionary problem, there is potential to improve the utility of current treatments and accelerate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; , ,
| | - Tavia Caplan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; , ,
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; , ,
| |
Collapse
|
41
|
Mount HO, Revie NM, Todd RT, Anstett K, Collins C, Costanzo M, Boone C, Robbins N, Selmecki A, Cowen LE. Global analysis of genetic circuitry and adaptive mechanisms enabling resistance to the azole antifungal drugs. PLoS Genet 2018; 14:e1007319. [PMID: 29702647 PMCID: PMC5922528 DOI: 10.1371/journal.pgen.1007319] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
Invasive fungal infections caused by the pathogen Candida albicans have transitioned from a rare curiosity to a major cause of human mortality. This is in part due to the emergence of resistance to the limited number of antifungals available to treat fungal infections. Azoles function by targeting the biosynthesis of ergosterol, a key component of the fungal cell membrane. Loss-of-function mutations in the ergosterol biosynthetic gene ERG3 mitigate azole toxicity and enable resistance that depends upon fungal stress responses. Here, we performed a genome-wide synthetic genetic array screen in Saccharomyces cerevisiae to map ERG3 genetic interactors and uncover novel circuitry important for azole resistance. We identified nine genes that enabled erg3-mediated azole resistance in the model yeast and found that only two of these genes had a conserved impact on resistance in C. albicans. Further, we screened a C. albicans homozygous deletion mutant library and identified 13 genes for which deletion enhances azole susceptibility. Two of the genes, RGD1 and PEP8, were also important for azole resistance acquired by diverse mechanisms. We discovered that loss of function of retrograde transport protein Pep8 overwhelms the functional capacity of the stress response regulator calcineurin, thereby abrogating azole resistance. To identify the mechanism through which the GTPase activator protein Rgd1 enables azole resistance, we selected for mutations that restore resistance in strains lacking Rgd1. Whole genome sequencing uncovered parallel adaptive mechanisms involving amplification of both chromosome 7 and a large segment of chromosome 3. Overexpression of a transporter gene on the right portion of chromosome 3, NPR2, was sufficient to enable azole resistance in the absence of Rgd1. Thus, we establish a novel mechanism of adaptation to drug-induced stress, define genetic circuitry underpinning azole resistance, and illustrate divergence in resistance circuitry over evolutionary time. Fungal infections caused by the pathogen Candida albicans pose a serious threat to human health. Treating these infections relies heavily on the azole antifungals, however, resistance to these drugs develops readily demanding novel therapeutic strategies. We performed large-scale systematic screens in both C. albicans and the model yeast Saccharomyces cerevisiae to identify genes that enable azole resistance. Our genome-wide screen in S. cerevisiae identified nine determinants of azole resistance, only two of which were important for resistance in C. albicans. Our screen of C. albicans mutants identified 13 genes for which deletion enhances susceptibility to azoles, including RGD1 and PEP8. We found that loss of Pep8 overwhelms the functional capacity of a key stress response regulator, calcineurin. In contrast, amplification of chromosome 7 and the right portion of chromosome 3 can restore resistance in strains lacking Rgd1, suggesting that Rgd1 may enable azole resistance by inducing genes in these amplified regions. Specifically, overexpression of a gene involved in transport on chromosome 3, NPR2, was sufficient to restore azole resistance in the absence of Rgd1. Thus, we establish novel circuitry important for antifungal drug resistance, and uncover adaptive mechanisms involving genomic plasticity that occur in response to drug induced stress.
Collapse
Affiliation(s)
| | - Nicole M. Revie
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Robert T. Todd
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Kaitlin Anstett
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Cathy Collins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Costanzo
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Anna Selmecki
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
42
|
Yadav A, Sinha H. Gene-gene and gene-environment interactions in complex traits in yeast. Yeast 2018; 35:403-416. [PMID: 29322552 DOI: 10.1002/yea.3304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 12/23/2017] [Indexed: 01/05/2023] Open
Abstract
One of the fundamental questions in biology is how the genotype regulates the phenotype. An increasing number of studies indicate that, in most cases, the effect of a genetic locus on the phenotype is context-dependent, i.e. it is influenced by the genetic background and the environment in which the phenotype is measured. Still, the majority of the studies, in both model organisms and humans, that map the genetic regulation of phenotypic variation in complex traits primarily identify additive loci with independent effects. This does not reflect an absence of the contribution of genetic interactions to phenotypic variation, but instead is a consequence of the technical limitations in mapping gene-gene interactions (GGI) and gene-environment interactions (GEI). Yeast, with its detailed molecular understanding, diverse population genomics and ease of genetic manipulation, is a unique and powerful resource to study the contributions of GGI and GEI in the regulation of phenotypic variation. Here we review studies in yeast that have identified GGI and GEI that regulate phenotypic variation, and discuss the contribution of these findings in explaining missing heritability of complex traits, and how observations from these GGI and GEI studies enhance our understanding of the mechanisms underlying genetic robustness and adaptability that shape the architecture of the genotype-phenotype map.
Collapse
Affiliation(s)
- Anupama Yadav
- Center for Cancer Systems Biology, and Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA.,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Himanshu Sinha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.,Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, 600036, India.,Robert Bosch Centre for Data Sciences and Artificial Intelligence, Indian Institute of Technology Madras, Chennai, 600036, India
| |
Collapse
|
43
|
Bleuven C, Landry CR. Molecular and cellular bases of adaptation to a changing environment in microorganisms. Proc Biol Sci 2017; 283:rspb.2016.1458. [PMID: 27798299 DOI: 10.1098/rspb.2016.1458] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/04/2016] [Indexed: 12/27/2022] Open
Abstract
Environmental heterogeneity constitutes an evolutionary challenge for organisms. While evolutionary dynamics under variable conditions has been explored for decades, we still know relatively little about the cellular and molecular mechanisms involved. It is of paramount importance to examine these molecular bases because they may play an important role in shaping the course of evolution. In this review, we examine the diversity of adaptive mechanisms in the face of environmental changes. We exploit the recent literature on microbial systems because those have benefited the most from the recent emergence of genetic engineering and experimental evolution followed by genome sequencing. We identify four emerging trends: (i) an adaptive molecular change in a pathway often results in fitness trade-off in alternative environments but the effects are dependent on a mutation's genetic background; (ii) adaptive changes often modify transcriptional and signalling pathways; (iii) several adaptive changes may occur within the same molecular pathway but be associated with pleiotropy of different signs across environments; (iv) because of their large associated costs, macromolecular changes such as gene amplification and aneuploidy may be a rapid mechanism of adaptation in the short-term only. The course of adaptation in a variable environment, therefore, depends on the complexity of the environment but also on the molecular relationships among the genes involved and between the genes and the phenotypes under selection.
Collapse
Affiliation(s)
- Clara Bleuven
- Département de Biologie, Université Laval, Québec, Québec, Canada .,Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada.,PROTEO, The Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Québec, Canada
| | - Christian R Landry
- Département de Biologie, Université Laval, Québec, Québec, Canada.,Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada.,Big Data Research Center, Université Laval, Québec, Québec, Canada.,PROTEO, The Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Québec, Canada
| |
Collapse
|
44
|
Impact of Morphological Sectors on Antifungal Susceptibility Testing and Virulence Studies. Antimicrob Agents Chemother 2017; 61:AAC.00755-17. [PMID: 28993330 DOI: 10.1128/aac.00755-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/30/2017] [Indexed: 12/11/2022] Open
Abstract
Morphological heterogeneity of Aspergillus terreus cultures was observed during continued cultivation of amphotericin B (AMB)-resistant isolates on drug-free medium. Outgrowth leads to the emergence of multiple sectors that might result from increased growth rates at drug-free conditions. We evaluated the differences in AMB susceptibility and virulence between sector subcultures (ATSec), AMB-resistant (ATR) strains, and AMB-susceptible (ATS) strains. By comparing A. terreus AMB-resistant (ATR) strains and A. terreus sector (ATSec) cultures we observed a highly significant reduction of AMB MICs in ATSec (ATR MIC, 2 to 32 μg/ml; ATSec MIC, 0.12 to 5 μg/ml). Furthermore, Galleria mellonella survival studies revealed an enhanced virulence of ATSec, which was comparable with that of AMB-sensitive Aspergillus terreus strains (median survival rates for ATS isolates, 72 h; for ATSec isolate ATSecG1, 84 h; for ATR isolates, 144 h). Our findings clearly demonstrate that spontaneous culture degeneration occurs in A. terreus and, most importantly, crucially impacts drug efficacy and virulence.
Collapse
|
45
|
Juvvadi PR, Lee SC, Heitman J, Steinbach WJ. Calcineurin in fungal virulence and drug resistance: Prospects for harnessing targeted inhibition of calcineurin for an antifungal therapeutic approach. Virulence 2017; 8:186-197. [PMID: 27325145 PMCID: PMC5354160 DOI: 10.1080/21505594.2016.1201250] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/26/2023] Open
Abstract
Increases in the incidence and mortality due to the major invasive fungal infections such as aspergillosis, candidiasis and cryptococcosis caused by the species of Aspergillus, Candida and Cryptococcus, are a growing threat to the immunosuppressed patient population. In addition to the limited armamentarium of the current classes of antifungal agents available (pyrimidine analogs, polyenes, azoles, and echinocandins), their toxicity, efficacy and the emergence of resistance are major bottlenecks limiting successful patient outcomes. Although these drugs target distinct fungal pathways, there is an urgent need to develop new antifungals that are more efficacious, fungal-specific, with reduced or no toxicity and simultaneously do not induce resistance. Here we review several lines of evidence which indicate that the calcineurin signaling pathway, a target of the immunosuppressive drugs FK506 and cyclosporine A, orchestrates growth, virulence and drug resistance in a variety of fungal pathogens and can be exploited for novel antifungal drug development.
Collapse
Affiliation(s)
- Praveen R. Juvvadi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Soo Chan Lee
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph Heitman
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - William J. Steinbach
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
46
|
Li X, Robbins N, O'Meara TR, Cowen LE. Extensive functional redundancy in the regulation of Candida albicans drug resistance and morphogenesis by lysine deacetylases Hos2, Hda1, Rpd3 and Rpd31. Mol Microbiol 2016; 103:635-656. [PMID: 27868254 DOI: 10.1111/mmi.13578] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2016] [Indexed: 12/22/2022]
Abstract
Current treatment efforts for fungal infections are hampered by the limited availability of antifungal drugs and by the emergence of drug resistance. A powerful strategy to enhance the efficacy of antifungal drugs is to inhibit the molecular chaperone Hsp90. Hsp90 governs drug resistance, morphogenesis and virulence in a leading fungal pathogen of humans, Candida albicans. Our previous work with Saccharomyces cerevisiae established acetylation as a novel mechanism of posttranslational control of Hsp90 function in fungi. We implicated lysine deacetylases (KDACs) as key regulators of resistance to the most widely deployed class of antifungals, the azoles, in both S. cerevisiae and C. albicans. Here, we demonstrate high levels of functional redundancy among the KDACs that are important for regulating Hsp90 function. We identify Hos2, Hda1, Rpd3 and Rpd31 as the KDACs mediating azole resistance and morphogenesis in C. albicans. Furthermore, we identify lysine 30 and 271 as critical acetylation sites on C. albicans Hsp90, and substitutions at these residues compromise Hsp90 function. Finally, we show that pharmacological inhibition of KDACs phenocopies pharmacological inhibition of Hsp90 and abrogates Hsp90-dependent azole resistance in numerous Candida species. This work illuminates new facets to the impact of KDACs on fungal drug resistance and morphogenesis, provides important insights into the divergence of the C. albicans Hsp90 regulatory network and reveals new targets for development of antifungal drugs.
Collapse
Affiliation(s)
- Xinliu Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Teresa R O'Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
47
|
Abstract
ABSTRACT
Invasive fungal infections are becoming an increasingly important cause of human mortality and morbidity, particularly for immunocompromised populations. The fungal pathogens
Candida albicans
,
Cryptococcus neoformans
, and
Aspergillus fumigatus
collectively contribute to over 1 million human deaths annually. Hence, the importance of safe and effective antifungal therapeutics for the practice of modern medicine has never been greater. Given that fungi are eukaryotes like their human host, the number of unique molecular targets that can be exploited for drug development remains limited. Only three classes of molecules are currently approved for the treatment of invasive mycoses. The efficacy of these agents is compromised by host toxicity, fungistatic activity, or the emergence of drug resistance in pathogen populations. Here we describe our current arsenal of antifungals and highlight current strategies that are being employed to improve the therapeutic safety and efficacy of these drugs. We discuss state-of-the-art approaches to discover novel chemical matter with antifungal activity and highlight some of the most promising new targets for antifungal drug development. We feature the benefits of combination therapy as a strategy to expand our current repertoire of antifungals and discuss the antifungal combinations that have shown the greatest potential for clinical development. Despite the paucity of new classes of antifungals that have come to market in recent years, it is clear that by leveraging innovative approaches to drug discovery and cultivating collaborations between academia and industry, there is great potential to bolster the antifungal armamentarium.
Collapse
|
48
|
O’Meara TR, Veri AO, Polvi EJ, Li X, Valaei SF, Diezmann S, Cowen LE. Mapping the Hsp90 Genetic Network Reveals Ergosterol Biosynthesis and Phosphatidylinositol-4-Kinase Signaling as Core Circuitry Governing Cellular Stress. PLoS Genet 2016; 12:e1006142. [PMID: 27341673 PMCID: PMC4920384 DOI: 10.1371/journal.pgen.1006142] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/27/2022] Open
Abstract
Candida albicans is a leading human fungal pathogen that causes life-threatening systemic infections. A key regulator of C. albicans stress response, drug resistance, morphogenesis, and virulence is the molecular chaperone Hsp90. Targeting Hsp90 provides a powerful strategy to treat fungal infections, however, the therapeutic utility of current inhibitors is compromised by toxicity due to inhibition of host Hsp90. To identify components of the Hsp90-dependent circuitry governing virulence and drug resistance that are sufficiently divergent for selective targeting in the pathogen, we pioneered chemical genomic profiling of the Hsp90 genetic network in C. albicans. Here, we screen mutant collections covering ~10% of the genome for hypersensitivity to Hsp90 inhibition in multiple environmental conditions. We identify 158 HSP90 chemical genetic interactors, most of which are important for growth only in specific environments. We discovered that the sterol C-22 desaturase gene ERG5 and the phosphatidylinositol-4-kinase (PI4K) gene STT4 are HSP90 genetic interactors under multiple conditions, suggesting a function upstream of Hsp90. By systematic analysis of the ergosterol biosynthetic cascade, we demonstrate that defects in ergosterol biosynthesis induce cellular stress that overwhelms Hsp90's functional capacity. By analysis of the phosphatidylinositol pathway, we demonstrate that there is a genetic interaction between the PI4K Stt4 and Hsp90. We also establish that Stt4 is required for normal actin polarization through regulation of Wal1, and suggest a model in which defects in actin remodeling induces stress that creates a cellular demand for Hsp90 that exceeds its functional capacity. Consistent with this model, actin inhibitors are synergistic with Hsp90 inhibitors. We highlight new connections between Hsp90 and virulence traits, demonstrating that Erg5 and Stt4 enable activation of macrophage pyroptosis. This work uncovers novel circuitry regulating Hsp90 functional capacity and new effectors governing drug resistance, morphogenesis and virulence, revealing new targets for antifungal drug development.
Collapse
Affiliation(s)
- Teresa R. O’Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda O. Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth J. Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Xinliu Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Stephanie Diezmann
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Claverton Down, Bath, United Kingdom
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
49
|
Zhao B, Sedlak JC, Srinivas R, Creixell P, Pritchard JR, Tidor B, Lauffenburger DA, Hemann MT. Exploiting Temporal Collateral Sensitivity in Tumor Clonal Evolution. Cell 2016; 165:234-246. [PMID: 26924578 DOI: 10.1016/j.cell.2016.01.045] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/19/2015] [Accepted: 01/26/2016] [Indexed: 12/14/2022]
Abstract
The prevailing approach to addressing secondary drug resistance in cancer focuses on treating the resistance mechanisms at relapse. However, the dynamic nature of clonal evolution, along with potential fitness costs and cost compensations, may present exploitable vulnerabilities-a notion that we term "temporal collateral sensitivity." Using a combined pharmacological screen and drug resistance selection approach in a murine model of Ph(+) acute lymphoblastic leukemia, we indeed find that temporal and/or persistent collateral sensitivity to non-classical BCR-ABL1 drugs arises in emergent tumor subpopulations during the evolution of resistance toward initial treatment with BCR-ABL1-targeted inhibitors. We determined the sensitization mechanism via genotypic, phenotypic, signaling, and binding measurements in combination with computational models and demonstrated significant overall survival extension in mice. Additional stochastic mathematical models and small-molecule screens extended our insights, indicating the value of focusing on evolutionary trajectories and pharmacological profiles to identify new strategies to treat dynamic tumor vulnerabilities.
Collapse
Affiliation(s)
- Boyang Zhao
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joseph C Sedlak
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA
| | - Raja Srinivas
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Pau Creixell
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Justin R Pritchard
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bruce Tidor
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Douglas A Lauffenburger
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Michael T Hemann
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
50
|
Kim SH, Clark ST, Surendra A, Copeland JK, Wang PW, Ammar R, Collins C, Tullis DE, Nislow C, Hwang DM, Guttman DS, Cowen LE. Global Analysis of the Fungal Microbiome in Cystic Fibrosis Patients Reveals Loss of Function of the Transcriptional Repressor Nrg1 as a Mechanism of Pathogen Adaptation. PLoS Pathog 2015; 11:e1005308. [PMID: 26588216 PMCID: PMC4654494 DOI: 10.1371/journal.ppat.1005308] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/03/2015] [Indexed: 01/10/2023] Open
Abstract
The microbiome shapes diverse facets of human biology and disease, with the importance of fungi only beginning to be appreciated. Microbial communities infiltrate diverse anatomical sites as with the respiratory tract of healthy humans and those with diseases such as cystic fibrosis, where chronic colonization and infection lead to clinical decline. Although fungi are frequently recovered from cystic fibrosis patient sputum samples and have been associated with deterioration of lung function, understanding of species and population dynamics remains in its infancy. Here, we coupled high-throughput sequencing of the ribosomal RNA internal transcribed spacer 1 (ITS1) with phenotypic and genotypic analyses of fungi from 89 sputum samples from 28 cystic fibrosis patients. Fungal communities defined by sequencing were concordant with those defined by culture-based analyses of 1,603 isolates from the same samples. Different patients harbored distinct fungal communities. There were detectable trends, however, including colonization with Candida and Aspergillus species, which was not perturbed by clinical exacerbation or treatment. We identified considerable inter- and intra-species phenotypic variation in traits important for host adaptation, including antifungal drug resistance and morphogenesis. While variation in drug resistance was largely between species, striking variation in morphogenesis emerged within Candida species. Filamentation was uncoupled from inducing cues in 28 Candida isolates recovered from six patients. The filamentous isolates were resistant to the filamentation-repressive effects of Pseudomonas aeruginosa, implicating inter-kingdom interactions as the selective force. Genome sequencing revealed that all but one of the filamentous isolates harbored mutations in the transcriptional repressor NRG1; such mutations were necessary and sufficient for the filamentous phenotype. Six independent nrg1 mutations arose in Candida isolates from different patients, providing a poignant example of parallel evolution. Together, this combined clinical-genomic approach provides a high-resolution portrait of the fungal microbiome of cystic fibrosis patient lungs and identifies a genetic basis of pathogen adaptation. Microbial cells vastly outnumber human cells in our bodies, yet we are only beginning to understand how these microbes influence human health and disease. One disease for which microbial communities are especially important is cystic fibrosis, where persistent lung infections can be lethal. Fungi are associated with poor respiratory function, but how fungal communities change with disease progression or treatment remains enigmatic. Here, we assess the dynamics of fungal communities by combining high-throughput sequencing of sputum samples from 28 patients with detailed analysis of phenotypes and genotypes of 1,603 fungal isolates. We found stable communities dominated by Candida and Aspergillus, and diversity in traits important for host adaptation. Antifungal drug resistance varied largely between species, while morphogenesis varied within species. For Candida species, the capacity to transition between yeast and filaments is a key virulence trait that is normally regulated by inducing cues, however, 28 isolates grew as filaments without such cues. Filamentation was due to loss-of-function mutations in the transcriptional regulator NRG1 in most isolates, which conferred resistance to the filament-repressive effects of a common bacterial pathogen. This work provides a portrait of the fungal microbiome associated with a lethal disease, and illuminates a genetic basis of pathogen adaptation.
Collapse
Affiliation(s)
- Sang Hu Kim
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Shawn T. Clark
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Anuradha Surendra
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Julia K. Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Pauline W. Wang
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ron Ammar
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Cathy Collins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Corey Nislow
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - David M. Hwang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - David S. Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|