1
|
Zajac C, Scott NE, Kline S, Erayil SE, Selmecki A. Hotspot gene conversion between FKS1 and FKS2 in echinocandin resistant Candida glabrata serial isolates. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:31. [PMID: 40247099 PMCID: PMC12006411 DOI: 10.1038/s44259-025-00102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025]
Abstract
Candida glabrata (Nakaseomyces glabratus) is the most common cause of drug-resistant candidemia and is associated with a high mortality rate. Only a few mechanisms of drug resistance are known in C. glabrata, predominantly involving recurrent single nucleotide polymorphisms. The importance of structural variation in acquired drug resistance is not understood. We performed comparative phenotypic and genomic analyses of six serial bloodstream isolates of C. glabrata and identified novel mutations associated with resistance to echinocandins. Critically, we identified a novel gene conversion event between the hotspot 2 regions of FKS1 and FKS2 that was associated with increased resistance to micafungin. We further analyzed 621 publicly available C. glabrata genomes and found three additional examples of structural variation involving FKS1 and FKS2. Ultimately, drug resistance in C. glabrata involves structural variants that are missed with current diagnostic methods and need to be considered when designing and implementing more effective antifungal management strategies.
Collapse
Affiliation(s)
- Christopher Zajac
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Nancy E Scott
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- University of Minnesota, Bioinformatics and Computational Biology Program, Minneapolis, MN, USA
| | - Susan Kline
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Serin E Erayil
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA.
- University of Minnesota, Bioinformatics and Computational Biology Program, Minneapolis, MN, USA.
| |
Collapse
|
2
|
Sharma A, Homayoon A, Weyler M, Frazer C, Ramírez-Zavala B, Morschhäuser J, Bennett RJ. Transcriptional control of C. albicans white-opaque switching and modulation by environmental cues and strain background. mBio 2025:e0058125. [PMID: 40202334 DOI: 10.1128/mbio.00581-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
The opportunistic fungal pathogen Candida albicans can undergo cellular transitions in response to environmental cues that impact its lifestyle and its interactions with the human host. This is exemplified by the white-opaque switch, which is a heritable transition between two phenotypic states that is regulated by a highly interconnected network of transcription factors (TFs). To obtain greater understanding of the transcriptional regulation of the switch, we generated a genome-wide, tetracycline-inducible TF library in the WO-1 strain background and identified those TFs whose forced expression induces white cells to switch to the opaque state. This set of opaque-inducing TFs was also evaluated for their ability to induce switching in a second strain background, that of the standard reference strain SC5314, as well as during growth on different laboratory media. These experiments identify 14 TFs that can drive white-to-opaque switching when overexpressed but that do so in a highly strain- and media-specific manner. In particular, changes in pH, amino acids, and zinc concentrations had marked effects on the ability of TFs to drive phenotypic switching. These results provide insights into the complex transcriptional regulation of switching in C. albicans and reveal that an interplay between genetic and environmental factors determines TF function and cell fate.IMPORTANCEThe white-opaque switch in Candida albicans represents a model system for understanding an epigenetic switch in a eukaryotic pathogen. Here, we generated an inducible library of the set of transcription factors (TFs) present in C. albicans and identify 14 TFs that can drive the white-to-opaque transition when ectopically expressed. We demonstrate that several of these TFs induce the switch in a highly strain- and media-specific manner. This highlights that both strain background and changes in experimental conditions (including different water sources) can profoundly impact the phenotypic consequences of TF overexpression. Moreover, the inducible TF library provides an invaluable tool for the further analysis of TF function in this important human pathogen.
Collapse
Affiliation(s)
- Anupam Sharma
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Ameen Homayoon
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Michael Weyler
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Corey Frazer
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Bernardo Ramírez-Zavala
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Joachim Morschhäuser
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Richard J Bennett
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
3
|
Mackey AI, Fillinger RJ, Hendricks PS, Thomson GJ, Cuomo CA, Bennett RJ, Anderson MZ. Aneuploidy confers a unique transcriptional and phenotypic profile to Candida albicans. Nat Commun 2025; 16:3287. [PMID: 40189588 PMCID: PMC11973194 DOI: 10.1038/s41467-025-58457-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Inaccurate chromosome segregation can lead to the formation of aneuploid cells that harbor an imbalanced complement of chromosomes. Several fungal species are not only able to tolerate the detrimental effects of aneuploidy but can use it to adapt to environmental pressures. The fungal pathobiont Candida albicans frequently acquires supernumerary chromosomes that enable growth in the presence of antifungal drugs or in specific host niches, yet the transcriptional changes associated with aneuploidy are not globally defined. Here, a karyotypically diverse set of C. albicans strains revealed that expression generally correlated with gene copy number regardless of the strain karyotype. Unexpectedly, aneuploid strains shared a characteristic transcriptional profile that was distinct from a generalized environmental stress response previously defined in aneuploid yeast cells. This aneuploid transcriptional response led to altered growth and oxidative balances relative to euploid control strains. The increased expression of reactive oxygen species (ROS) mitigating enzymes in aneuploid cells reduced the levels of ROS but caused an acute sensitivity to both internal and external sources of oxidative stress. Taken together, our work demonstrates common transcriptional and phenotypic features of aneuploid C. albicans cells with consequences for infection of different host niches and susceptibility to environmental stimuli.
Collapse
Affiliation(s)
- Anna I Mackey
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Robert J Fillinger
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - P Shane Hendricks
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Gregory J Thomson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Christina A Cuomo
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02412, USA
| | - Richard J Bennett
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Matthew Z Anderson
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA.
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA.
- Center for Genomic Science Innovation, University of Wisconsin - Madison, Madison, WI, 53706, USA.
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, WI, 53706, USA.
| |
Collapse
|
4
|
Phan-Canh T, Bitencourt T, Kuchler K. Gene dosage of PDR16 modulates azole susceptibility in Candida auris. Microbiol Spectr 2025:e0265924. [PMID: 40130854 DOI: 10.1128/spectrum.02659-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Affiliation(s)
- Trinh Phan-Canh
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, Vienna, Austria
| | - Tamires Bitencourt
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, Vienna, Austria
- Labdia - Labordiagnostik GmbH, CCRI - St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, Vienna, Austria
| |
Collapse
|
5
|
Xiong EH, Zhang X, Robbins N, Myers CL, Cowen LE. Protocol to identify genes important for Candida albicans fitness in diverse environmental conditions using pooled bar-seq screening approach. STAR Protoc 2025; 6:103645. [PMID: 39985775 PMCID: PMC11889967 DOI: 10.1016/j.xpro.2025.103645] [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: 11/22/2024] [Revised: 01/10/2025] [Accepted: 01/31/2025] [Indexed: 02/24/2025] Open
Abstract
Identifying genes important for fitness in Candida albicans advances our understanding of this important pathogen of humans. Here, we present a functional genomics approach for assessing fitness through the quantification of strain-specific barcodes. We describe steps for library preparation, propagation of strains, genomic DNA extraction, amplification of barcodes, and sequencing. We then detail the computational analysis of data to determine effect size and statistical significance. For complete details on the use and execution of this protocol, please refer to Xiong et al.1.
Collapse
Affiliation(s)
- Emily H Xiong
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiang Zhang
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
6
|
Lew-Smith J, Binkley J, Sherlock G. The Candida Genome Database: annotation and visualization updates. Genetics 2025; 229:iyaf001. [PMID: 39776186 DOI: 10.1093/genetics/iyaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
The Candida Genome Database (CGD; www.candidagenome.org) is unique in being both a model organism database and a fungal pathogen database. As a fungal pathogen database, CGD hosts locus pages for 5 species of the best-studied pathogenic fungi in the Candida group. As a model organism database, the species Candida albicans serves as a model both for other Candida spp. and for non-Candida fungi that form biofilms and undergo routine morphogenic switching from the planktonic form to the filamentous form, which is not done by other model yeasts. As pathogenic Candida species have become increasingly drug resistant, the high lethality of invasive candidiasis in immunocompromised people is increasingly alarming. There is a pressing need for additional research into basic Candida biology, epidemiology and phylogeny, and potential new antifungals. CGD serves the needs of this diverse research community by curating the entire gene-based Candida experimental literature as it is published, extracting, organizing, and standardizing gene annotations. Gene pages were added for the species Candida auris, recent outbreaks of which have been labeled an "urgent" threat. Most recently, we have begun linking clinical data on disease to relevant Literature Topics to improve searchability for clinical researchers. Because CGD curates for multiple species and most research focuses on aspects related to pathogenicity, we focus our curation efforts on assigning Literature Topic tags, collecting detailed mutant phenotype data, and assigning controlled Gene Ontology terms with accompanying evidence codes. Our Summary pages for each feature include the primary name and all aliases for that locus, a description of the gene and/or gene product, detailed ortholog information with links, a JBrowse window with a visual view of the gene on its chromosome, summarized phenotype, Gene Ontology, and sequence information, references cited on the summary page itself, and any locus notes. The database serves as a community hub, where we link to various types of reference material of relevance to Candida researchers, including colleague information, news, and notice of upcoming meetings. We routinely survey the community to learn how the field is evolving and how needs may have changed. For example, we asked our users which species we should next add to CGD, and the clear answer was Candida tropicalis. A key future challenge is management of the flood of high-throughput expression data to make it as useful as possible to as many researchers as possible. The central challenge for any community database is to turn data into knowledge, which the community can access, use, and build upon.
Collapse
Affiliation(s)
- Jodi Lew-Smith
- Department of Genetics, Stanford University, Stanford CA 94305-5120, USA
| | - Jonathan Binkley
- Department of Genetics, Stanford University, Stanford CA 94305-5120, USA
| | - Gavin Sherlock
- Department of Genetics, Stanford University, Stanford CA 94305-5120, USA
| |
Collapse
|
7
|
Nemeth T, Zarnocki A, Ladanyi A, Papp C, Ayaydin F, Szebeni GJ, Gacser A. PCR-based CRISPR/Cas9 system for fluorescent tagging: A tool for studying Candida parapsilosis virulence. PLoS One 2025; 20:e0312948. [PMID: 39992908 DOI: 10.1371/journal.pone.0312948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/16/2024] [Indexed: 02/26/2025] Open
Abstract
Candida parapsilosis is persistent in a hospital environment hence it is often associated with nosocomial infections especially amongst low-birth weight neonates. Genetic modification is therefore important to characterise the physiological and virulence related properties of this fungus. A PCR-based CRISPR/Cas9 system has been adopted to facilitate the generation of fluorescent tagged prototroph isolates. We examined a total of eight fluorescent protein coding genes, out of which three were found to be applicable for simultaneous utilisation. We investigated three clinical isolates of C. parapsilosis in terms of their adherence to silicone and their uptake by J774.2 murine macrophages in competition assays. Interestingly, we found significant differences between them in both experiments where GA1 isolate was significantly less resistant to macrophage uptake and CDC317 was significantly more adherent to silicone material. In silico analysis of the agglutinin-like sequences (Als) exposed remarkable diversity in this protein family and additionally, the thorough analysis of the ALS genes revealed evidence of formation of a new gene by intrachromosomal recombination in the GA1 isolate. Finally, we provide a step by step protocol for the application of the PCR-based CRISPR/Cas9 system for fluorescently labelling C. parapsilosis isolates.
Collapse
Affiliation(s)
- Tibor Nemeth
- Department of Biotechnology and Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Andrea Zarnocki
- Department of Biotechnology and Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anett Ladanyi
- Department of Biotechnology and Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Csaba Papp
- Department of Biotechnology and Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferhan Ayaydin
- Functional Cell Biology and Immunology Advanced Core Facility (FCBI-ACF), Hungarian Centre of Excellence for Molecular Medicine (HCEMM), University of Szeged, Szeged, Hungary
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Plant Biology, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gabor Janos Szebeni
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Attila Gacser
- Department of Biotechnology and Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Pathomechanisms of Fungal Infections Research Group, University of Szeged, Szeged, Hungary
- IKIKK, Competence Centre for Molecular Biology, Bionics and Biotechnology, University of Szeged, Szeged, Hungary
| |
Collapse
|
8
|
Spettel K, Bumberger D, Kriz R, Frank S, Loy M, Galazka S, Suchomel M, Lagler H, Makristathis A, Willinger B. In vitro long-term exposure to chlorhexidine or triclosan induces cross-resistance against azoles in Nakaseomyces glabratus. Antimicrob Resist Infect Control 2025; 14:2. [PMID: 39849551 PMCID: PMC11755926 DOI: 10.1186/s13756-024-01511-4] [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: 03/12/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Topical antiseptics are crucial for preventing infections and reducing transmission of pathogens. However, commonly used antiseptic agents have been reported to cause cross-resistance to other antimicrobials in bacteria, which has not yet been described in yeasts. This study aims to assess the in vitro efficacy of antiseptics against clinical and reference isolates of Candida albicans and Nakaseomyces glabratus, and whether prolonged exposure to antiseptics promotes the development of antifungal (cross)resistance. METHODS A high-throughput approach for in vitro resistance development was established to simultaneously expose 96 C. albicans and N. glabratus isolates to increasing concentrations of a given antiseptic - chlorhexidine, triclosan or octenidine. Susceptibility testing and whole genome sequencing of yeast isolates pre- and post-exposure were performed. RESULTS Long-term exposure to antiseptics does not result in the development of stable resistance to the antiseptics themselves. However, 50 N. glabratus isolates acquired resistance to azole antifungals after long-term exposure to triclosan or chlorhexidine, revealing newly acquired mutations in the PDR1 and PMA1 genes. CONCLUSIONS Chlorhexidine as well as triclosan, but not octenidine, were able to introduce selective pressure promoting resistance to azole antifungals. Although we assessed this phenomenon only in vitro, these findings warrant critical monitoring in clinical settings.
Collapse
Affiliation(s)
- Kathrin Spettel
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, 1100, Austria
| | - Dominik Bumberger
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Richard Kriz
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, 1100, Austria
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
| | - Sarah Frank
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Madita Loy
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Sonia Galazka
- Division of Data, Statistics and Risk Assessment, Austrian Agency for Health and Food Safety AGES, Vienna, 1220, Austria
| | - Miranda Suchomel
- Institute for Hygiene and Applied Immunology, Medical University of Vienna, Vienna, 1090, Austria
| | - Heimo Lagler
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
| | - Athanasios Makristathis
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Birgit Willinger
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria.
| |
Collapse
|
9
|
James SA, Parker A, Purse C, Baker DJ, Funnell SGP, Carding SR. Draft genome sequence of a non-human primate-derived isolate of Candida parapsilosis. Microbiol Resour Announc 2025; 14:e0081124. [PMID: 39655927 PMCID: PMC11737174 DOI: 10.1128/mra.00811-24] [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: 07/29/2024] [Accepted: 11/15/2024] [Indexed: 01/18/2025] Open
Abstract
Candida parapsilosis is a common human commensal and opportunistic fungal pathogen that is also found in non-human primates (NHPs). Here, we report the first draft sequence of C. parapsilosis NCYC 4418, a fecal isolate from an adult cynomolgus macaque.
Collapse
Affiliation(s)
- Steve A. James
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Aimee Parker
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Catherine Purse
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - David J. Baker
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Simon G. P. Funnell
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
- UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Simon R. Carding
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
10
|
Syrjanen JL, Nore A, Roselletti E, Chakraborty T, Perera RL, Wilson D. Structural insights into mechanisms of zinc scavenging by the Candida albicans zincophore Pra1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632233. [PMID: 39829916 PMCID: PMC11741385 DOI: 10.1101/2025.01.09.632233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Candida albicans causes more than 400,000 life-threatening, and half a billion mucosal infections annually. In response to infection, the host limits availability of essential micronutrients, including zinc, to restrict growth of the invading pathogen. As assimilation of zinc is essential for C. albicans pathogenicity, its limitation induces the secretion of the zincophore protein Pra1 to scavenge zinc from the host. Pra1 also plays a number of important roles in host-pathogen interactions and is conserved in most fungi. However, the structure of fungal zincophores is not known. Here, we present the first cryogenic electron microscopy structures of C. albicans Pra1 in its apo- and zinc-bound states, at 2.8 and 2.5 Å resolution respectively. Our work reveals a hexameric ring-like assembly with multiple zinc binding sites. Through genetic studies, we show that one of these zinc binding sites is essential for C. albicans growth under zinc restriction but does not affect the inflammatory properties of Pra1. These data provide a foundation for future work to explore the structural basis of Pra1-mediated host-pathogen interactions, C. albicans zinc uptake, as well therapeutics development.
Collapse
Affiliation(s)
- Johanna L Syrjanen
- W.M Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA
- Current Address: Institute of Biotechnology, HiLIFE, University of Helsinki, FI 00014, Helsinki, Finland
| | - Alexandre Nore
- Faculty of Health and Life Sciences, Medical Research Council Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter, EX4 4QD, UK
| | - Elena Roselletti
- Faculty of Health and Life Sciences, Medical Research Council Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter, EX4 4QD, UK
| | - Tanmoy Chakraborty
- Faculty of Health and Life Sciences, Medical Research Council Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter, EX4 4QD, UK
| | | | - Duncan Wilson
- Faculty of Health and Life Sciences, Medical Research Council Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter, EX4 4QD, UK
| |
Collapse
|
11
|
Kim H, Heredia MY, Chen X, Ahmed M, Qasim M, Callender TL, Hernday AD, Rauceo JM. Mitochondrial targeting of Candida albicans SPFH proteins and requirement of stomatins for SDS-induced stress tolerance. Microbiol Spectr 2025; 13:e0173324. [PMID: 39641539 PMCID: PMC11705831 DOI: 10.1128/spectrum.01733-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
The SPFH (stomatin, prohibitin, flotillin, and HflK/HflC) protein superfamily is conserved across all domains of life. Fungal SPFH proteins are required for respiration, stress adaptation, and membrane scaffolding. In the yeast Candida albicans, stomatin-like protein 3 (Slp3) forms punctate foci at the plasma membrane, and SLP3 overexpression causes cell death following exposure to the surfactant, SDS, and the oxidative stressor, H2O2. Here, we sought to determine the cellular localization and functionally characterize stomatin-like protein 2 (Slp2), prohibitin-1 (Phb1), prohibitin-2 (Phb2), and prohibitin-12 (Phb12) in C. albicans. Cytological and western blotting results showed that Slp2-Gfp/Rfp and prohibitin-Gfp fusion proteins localize to the mitochondrion in yeast cells. Growth assay results did not identify any respiration defects in a panel of stomatin and prohibitin mutant strains, suggesting that SPFH respiratory function has diverged in C. albicans from other model eukaryotes. However, a slp2Δ/Δ/slp3Δ/Δ double mutant strain grew poorly in the presence of 0.08% SDS, accumulated intracellular reactive oxidative species, and displayed aberrant ergosterol distribution in the plasma membrane. These phenotypes were not observed in slp2Δ/Δ or slp3Δ/Δ single mutants, indicating a possible indirect genetic interaction between SLP2 and SLP3. In addition, slp2Δ/Δ and slp2Δ/Δ/slp3Δ/Δ mutant strains were slightly resistant to the antifungal drug, fluconazole. Collectively, these findings reveal the cellular localization of Slp2, Phb1, Phb2, and Phb12, highlight the significance of stomatins in C. albicans SDS stress tolerance, and, for the first time, associate stomatins with antifungal resistance. IMPORTANCE Stomatins and prohibitins coordinate respiration and stress adaptation in fungi. Invasive mycoses caused by Candida albicans are a significant cause of morbidity, and candidemia patients show high mortality rates worldwide. Mitochondria are essential for C. albicans commensalism and virulence, and mitochondrial proteins are targets for antifungal interventions. C. albicans encodes five SPFH proteins: two stomatin-like proteins and three prohibitins. We have previously shown that Slp3 is important for C. albicans adaptation to various types of environmental stress. Moreover, synthetic compounds that bind to mammalian prohibitins inhibit C. albicans filamentation and are fungicidal. However, there is limited information available regarding the remaining SPFH proteins. Our findings show that mitochondrial localization of SPFH proteins is conserved in C. albicans. In addition, we demonstrate the importance of stomatins in plasma membrane and mitochondrial stress tolerance.
Collapse
Affiliation(s)
- Hyunjeong Kim
- Department of Sciences, John Jay College of the City University of New York, New York, New York, USA
| | - Marienela Y. Heredia
- Department of Sciences, John Jay College of the City University of New York, New York, New York, USA
| | - Xiao Chen
- Department of Sciences, John Jay College of the City University of New York, New York, New York, USA
| | - Maisha Ahmed
- Department of Sciences, John Jay College of the City University of New York, New York, New York, USA
| | - Mohammad Qasim
- Department of Molecular and Cellular Biology, School of Natural Sciences, University of California, Merced, California, USA
| | - Tracy L. Callender
- Department of Biology, Farmingdale State College of the State University of New York, Farmingdale, New York, USA
| | - Aaron D. Hernday
- Department of Molecular and Cellular Biology, School of Natural Sciences, University of California, Merced, California, USA
| | - Jason M. Rauceo
- Department of Sciences, John Jay College of the City University of New York, New York, New York, USA
| |
Collapse
|
12
|
Jaeger M, Dietschmann A, Austermeier S, Dinçer S, Porschitz P, Vornholz L, Maas RJ, Sprenkeler EG, Ruland J, Wirtz S, Azam T, Joosten LA, Hube B, Netea MG, Dinarello CA, Gresnigt MS. Alpha1-antitrypsin impacts innate host-pathogen interactions with Candida albicans by stimulating fungal filamentation. Virulence 2024; 15:2333367. [PMID: 38515333 PMCID: PMC11008552 DOI: 10.1080/21505594.2024.2333367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
Our immune system possesses sophisticated mechanisms to cope with invading microorganisms, while pathogens evolve strategies to deal with threats imposed by host immunity. Human plasma protein α1-antitrypsin (AAT) exhibits pleiotropic immune-modulating properties by both preventing immunopathology and improving antimicrobial host defence. Genetic associations suggested a role for AAT in candidemia, the most frequent fungal blood stream infection in intensive care units, yet little is known about how AAT influences interactions between Candida albicans and the immune system. Here, we show that AAT differentially impacts fungal killing by innate phagocytes. We observed that AAT induces fungal transcriptional reprogramming, associated with cell wall remodelling and downregulation of filamentation repressors. At low concentrations, the cell-wall remodelling induced by AAT increased immunogenic β-glucan exposure and consequently improved fungal clearance by monocytes. Contrastingly, higher AAT concentrations led to excessive C. albicans filamentation and thus promoted fungal immune escape from monocytes and macrophages. This underscores that fungal adaptations to the host protein AAT can differentially define the outcome of encounters with innate immune cells, either contributing to improved immune recognition or fungal immune escape.
Collapse
Affiliation(s)
- Martin Jaeger
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Sude Dinçer
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Pauline Porschitz
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Larsen Vornholz
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Ralph J.A. Maas
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evelien G.G. Sprenkeler
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, USA
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Mark S. Gresnigt
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| |
Collapse
|
13
|
Carolus H, Sofras D, Boccarella G, Sephton-Clark P, Biriukov V, Cauldron NC, Lobo Romero C, Vergauwen R, Yazdani S, Pierson S, Jacobs S, Vandecruys P, Wijnants S, Meis JF, Gabaldón T, van den Berg P, Rybak JM, Cuomo CA, Van Dijck P. Acquired amphotericin B resistance leads to fitness trade-offs that can be mitigated by compensatory evolution in Candida auris. Nat Microbiol 2024; 9:3304-3320. [PMID: 39567662 DOI: 10.1038/s41564-024-01854-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/10/2024] [Indexed: 11/22/2024]
Abstract
Candida auris is a growing concern due to its resistance to antifungal drugs, particularly amphotericin B (AMB), detected in 30 to 60% of clinical isolates. However, the mechanisms of AMB resistance remain poorly understood. Here we investigated 441 in vitro- and in vivo-evolved C. auris lineages from 4 AMB-susceptible clinical strains of different clades. Genetic and sterol analyses revealed four major types of sterol alterations as a result of clinically rare variations in sterol biosynthesis genes ERG6, NCP1, ERG11, ERG3, HMG1, ERG10 and ERG12. In addition, aneuploidies in chromosomes 4 and 6 emerged during resistance evolution. Fitness trade-off phenotyping and mathematical modelling identified diverse strain- and mechanism-dependent fitness trade-offs. Variation in CDC25 rescued fitness trade-offs, thereby increasing the infection capacity. This possibly contributed to therapy-induced acquired AMB resistance in the clinic. Our findings highlight sterol-modulating mechanisms and fitness trade-off compensation as risks for AMB treatment failure in clinical settings.
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 Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | | | - Vladislav Biriukov
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Nicholas C Cauldron
- Molecular Microbiology and Immunology Department, Brown University, Providence, RI, USA
| | - Celia Lobo Romero
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Rudy Vergauwen
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Saleh Yazdani
- 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
| | - Stef Jacobs
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Paul Vandecruys
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Stefanie Wijnants
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Jacques F Meis
- Centre of Expertise in Mycology, Radboudumc/CWZ, Nijmegen, The Netherlands
- Institute of Translational Research, CECAD, University of Cologne, Cologne, Germany
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB), 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
| | - Pieter van den Berg
- Evolutionary Modelling Group, Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
| | | | - Christina A Cuomo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Molecular Microbiology and Immunology Department, Brown University, Providence, RI, USA
| | - 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
|
14
|
Cotter CJ, Trinh CT. CRISPR-GRIT: Guide RNAs with Integrated Repair Templates Enable Precise Multiplexed Genome Editing in the Diploid Fungal Pathogen Candida albicans. CRISPR J 2024; 7:385-394. [PMID: 39436817 DOI: 10.1089/crispr.2024.0052] [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: 10/25/2024] Open
Abstract
Candida albicans, an opportunistic fungal pathogen, causes severe infections in immunocompromised individuals. Limited classes and overuse of current antifungals have led to the rapid emergence of antifungal resistance. Thus, there is an urgent need to understand fungal pathogen genetics to develop new antifungal strategies. Genetic manipulation of C. albicans is encumbered by its diploid chromosomes requiring editing both alleles to elucidate gene function. Although the recent development of CRISPR-Cas systems has facilitated genome editing in C. albicans, large-scale and multiplexed functional genomic studies are still hindered by the necessity of cotransforming repair templates for homozygous knockouts. Here, we present CRISPR-GRIT (Guide RNAs with Integrated Repair Templates), a repair template-integrated guide RNA design for expedited gene knockouts and multiplexed gene editing in C. albicans. We envision that this method can be used for high-throughput library screens and identification of synthetic lethal pairs in both C. albicans and other diploid organisms with strong homologous recombination machinery.
Collapse
Affiliation(s)
- Christopher J Cotter
- Department of Chemical and Biomolecular Engineering, The University of Tennessee, Knoxville, Tennessee, USA
| | - Cong T Trinh
- Department of Chemical and Biomolecular Engineering, The University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
15
|
Gupta D, Shukla R, Mishra K. SUMO-targeted Ubiquitin Ligases as crucial mediators of protein homeostasis in Candida glabrata. PLoS Pathog 2024; 20:e1012742. [PMID: 39642165 DOI: 10.1371/journal.ppat.1012742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/18/2024] [Accepted: 11/11/2024] [Indexed: 12/08/2024] Open
Abstract
Candida glabrata is an opportunistic human pathogen, capable of causing severe systemic infections that are often resistant to standard antifungal treatments. To understand the importance of protein SUMOylation in the physiology and pathogenesis of C. glabrata, we earlier identified the components of SUMOylation pathway and demonstrated that the deSUMOylase CgUlp2 is essential for pathogenesis. In this work we show that the CgUlp2 is essential to maintain protein homeostasis via the SUMO-targeted ubiquitin ligase pathway. The dual loss of deSUMOylase and specific ubiquitin ligase, CgSlx8, results in heightened protein degradation, rendering the cells vulnerable to various stressors. This degradation affects crucial processes such as purine biosynthesis and compromises mitochondrial function in the mutants. Importantly, the absence of these ubiquitin ligases impedes the proliferation of C. glabrata in macrophages. These findings underscore the significance of SUMOylation and SUMO-mediated protein homeostasis as pivotal regulators of C. glabrata physiology and capacity to survive in host cells. Understanding these mechanisms could pave the way for the development of effective antifungal treatments.
Collapse
Affiliation(s)
- Dipika Gupta
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Renu Shukla
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Krishnaveni Mishra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
16
|
Kim MJ, White AM, Mitchell AP. Strain variation in Candida albicans glycolytic gene regulation. mSphere 2024; 9:e0057924. [PMID: 39431903 PMCID: PMC11580466 DOI: 10.1128/msphere.00579-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Central carbon metabolism is vital for the proliferation of Candida albicans, a fungus that is prominent as a commensal and pathogen. Glycolytic genes are activated by overlapping activities of the transcription factors Tye7 and Gal4, as shown by studies in the SC5314 genetic background. However, regulatory relationships can vary among C. albicans isolates. Here, we analyzed Tye7- and Gal4-related phenotypes in five diverse clinical isolates of C. albicans. We tested growth properties and gene expression impact through Nanostring profiling and, for the two strains SC5314 and P87, RNA sequencing. Our results lead to three main conclusions. First, the functional redundancy of Tye7 and Gal4 for glycolytic gene activation is preserved among all strains tested. Second, at the gene expression level, strain P87 is an outlier with regard to tye7Δ/Δ impact, and strain SC5314 is an outlier with regard to gal4Δ/Δ impact. Third, while Gal4 is well known to be dispensable for induction of the GAL1, GAL7, and GAL10 galactose-specific metabolic genes, we find that gal4Δ/Δ mutants of several strains have a mild galactose fermentation defect, as assayed by growth on galactose with the respiration inhibitor antimycin A. Our findings indicate that even a central metabolic regulatory network is subject to strain variation and illustrates an unexpected genotype-phenotype relationship.The fungal commensal and pathogen Candida albicans rely upon metabolic flexibility to colonize and infect host niches. Central carbon metabolism is governed by two regulators, Tye7 and Gal4, as defined in the reference strain SC5314. Here, we have explored the impact of Tye7 and Gal4 on carbon utilization and gene expression across five diverse C. albicans clinical isolates. Novel aspects of this study are the finding that even a central metabolic regulatory network is subject to strain variation and the observation of an unexpected mutant phenotype.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Amelia M. White
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
17
|
Kuplińska A, Rząd K, Stefaniak-Skorupa J, Kozłowska-Tylingo K, Wojciechowski M, Milewski S, Gabriel I. Targeting Candida albicans O-acetyl-L-homoserine sulfhydrylase (Met15p) in antifungal treatment. Sci Rep 2024; 14:28188. [PMID: 39548263 PMCID: PMC11568248 DOI: 10.1038/s41598-024-79886-y] [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: 07/24/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024] Open
Abstract
Fungal infections are a serious threat to public health as they are becoming increasingly frequent. A major problem stems also from a rising fungal resistance to currently available antifungal therapies, therefore novel molecular targets are highly desirable. Exploration of enzymes participating in the biosynthesis pathways of essential amino acids such as L-methionine (L-Met) may provide new insights into pharmaceutical development. The MET15 gene from Candida albicans, encoding O-acetyl-L-homoserine sulfhydrylase (Met15p), an enzyme catalyzing the second step in that pathway, was cloned and expressed in two versions: as N and C-terminal oligo-His-tagged fusion proteins. The recombinant enzymes revealed appropriate activity, and catalyzed conversion of O-acetyl-L-homoserine and a sulfide ion to produce L-homocysteine. A new RP-HPLC-DAD method, using the enzymatic reaction product pre-column derivatization with 5,5'-dithio-bis-(2-nitrobenzoic acid) was developed and used by us to determine Met15p activity. Newly synthesized compounds as well as two commercially available exhibited a Met15p inhibitory effect which was related to antifungal activity. Fungal cells' sensitivity to inhibitors depending on the presence or absence of L-Met in the medium clearly indicated Met15p targeting. Moreover, the synergistic effect of the first methionine biosynthetic enzyme affecting inhibitor and Met15p inhibitors indicate that methionine biosynthesis pathway enzymes are promising molecular targets.
Collapse
Affiliation(s)
- Aleksandra Kuplińska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Kamila Rząd
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | | | - Katarzyna Kozłowska-Tylingo
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Marek Wojciechowski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Sławomir Milewski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Iwona Gabriel
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland.
| |
Collapse
|
18
|
Shivarathri R, Chauhan M, Datta A, Das D, Karuli A, Aptekmann A, Jenull S, Kuchler K, Thangamani S, Chowdhary A, Desai JV, Chauhan N. The Candida auris Hog1 MAP kinase is essential for the colonization of murine skin and intradermal persistence. mBio 2024; 15:e0274824. [PMID: 39422509 PMCID: PMC11558994 DOI: 10.1128/mbio.02748-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Candida auris, a multidrug-resistant human fungal pathogen, was first identified in 2009 in Japan. Since then, systemic C. auris infections have now been reported in more than 50 countries, with mortality rates of 30%-60%. A major contributing factor to its high inter- and intrahospital clonal transmission is that C. auris, unlike most Candida species, displays unique skin tropism and can stay on human skin for a prolonged period. However, the molecular mechanisms responsible for C. auris skin colonization, intradermal persistence, and systemic virulence are poorly understood. Here, we report that C. auris Hog1 mitogen-activated protein kinase is essential for efficient skin colonization, intradermal persistence as well as systemic virulence. RNA-seq analysis of wild-type parental and hog1Δ mutant strains revealed marked downregulation of genes involved in processes such as cell adhesion, cell wall rearrangement, and pathogenesis in hog1Δ mutant compared to the wild-type parent. Consistent with these data, we found a prominent role for Hog1 in maintaining cell wall architecture, as the hog1Δ mutant demonstrated a significant increase in cell-surface β-glucan exposure and a concomitant reduction in chitin content. Additionally, we observed that Hog1 was required for biofilm formation in vitro and fungal survival when challenged with primary murine macrophages and neutrophils ex vivo. Collectively, these findings have important implications for understanding the C. auris skin adherence mechanisms and penetration of skin epithelial layers preceding bloodstream infections. IMPORTANCE Candida auris is a World Health Organization fungal priority pathogen and an urgent public health threat recognized by the Centers for Disease Control and Prevention. C. auris has a unique ability to colonize human skin. It also persists on abiotic surfaces in healthcare environments for an extended period of time. These attributes facilitate the inter- and intrahospital clonal transmission of C. auris. Therefore, understanding C. auris skin colonization mechanisms is critical for infection control, especially in hospitals and nursing homes. However, despite its profound clinical relevance, the molecular and genetic basis of C. auris skin colonization mechanisms are poorly understood. Herein, we present data on the identification of the Hog1 MAP kinase as a key regulator of C. auris skin colonization. These findings lay the foundation for further characterization of unique mechanisms that promote fungal persistence on human skin.
Collapse
Affiliation(s)
- Raju Shivarathri
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Manju Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Abhishek Datta
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Diprasom Das
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Adela Karuli
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Ariel Aptekmann
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Sabrina Jenull
- Department of Medical Biochemistry, Medical University Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karl Kuchler
- Department of Medical Biochemistry, Medical University Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Shankar Thangamani
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Anuradha Chowdhary
- Department of Medical Mycology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Jigar V. Desai
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Neeraj Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
19
|
Gonçalves B, Pires DP, Fernandes L, Pacheco M, Ferreira T, Osório H, Soares AR, Henriques M, Silva S. Biofilm matrix regulation by Candida glabrata Zap1 under acidic conditions: transcriptomic and proteomic analyses. Microbiol Spectr 2024; 12:e0120124. [PMID: 39494883 PMCID: PMC11619577 DOI: 10.1128/spectrum.01201-24] [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: 05/14/2024] [Accepted: 08/06/2024] [Indexed: 11/05/2024] Open
Abstract
The vaginal acidic environment potentiates the formation of Candida glabrata biofilms, leading to complicated and recurrent infections. Importantly, the production of matrix is known to contribute to the recalcitrant features of Candida biofilms. In this study, we reveal that Zap1 regulates the matrix of C. glabrata acidic biofilms and analyzed the modulation of their transcriptome (by microarrays) and matrix proteome (by LC-MS/MS) by Zap1. For that, the deletion mutant zap1Δ and its complemented strain zap1Δ::ZAP1 were constructed, and their biofilms were developed at pH 4 (adjusted with lactic acid). The results revealed that Zap1 is a negative regulator of the total amount of protein and carbohydrate in the biofilm matrix. Accordingly, various genes and matrix proteins with predicted functions in the regulation of carbohydrate metabolism, sugar binding, sugar transport, and adhesion (including Epa family) were repressed by Zap1. Nevertheless, the results also suggested that Zap1 is essential to the delivery and organization of some matrix components. Indeed, Zap1 was required for the secretion of 122 proteins to the matrix and induced the expression of 557 genes, including various targets involved in glucan metabolism. Additionally, Zap1 induced targets with roles in virulence, resistance to antifungals, and host immunity evasion, including yapsins, ERG family, and moonlighting proteins. Zap1 was also required for the secretion of acidic-specific matrix proteins, indicating a contribution to the response to the acidic environment. Overall, this study demonstrates that Zap1 is a relevant regulator of the biofilm matrix, contributing to a better understanding of C. glabrata acidic biofilms.IMPORTANCEThe rising prevalence of vulvovaginal candidiasis (VVC) and the increasing presence of Candida spp. with aggressive virulence features and low susceptibility to common antifungals, particularly Candida glabrata, have resulted in more severe, prolonged, and recurrent cases of VVC, with significant implications for patients. This research offers valuable insights into the molecular changes that contribute to the formation of C. glabrata biofilms in the acidic vaginal environment, representing a significant advancement in the understanding of C. glabrata's virulence. Notably, this study identified Zap1 as a critical regulator of C. glabrata biofilm matrix, with additional potential roles in adhesion, antifungal resistance, evasion of host immunity, and response to acidic conditions, making it a promising target for new therapeutic approaches. Importantly, Zap1 is the first regulator of the biofilm matrix to be identified in C. glabrata, and the elucidation of its targets (including genes and matrix proteins) lays a strong foundation for future research.
Collapse
Affiliation(s)
- Bruna Gonçalves
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| | - Diana Priscila Pires
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| | - Liliana Fernandes
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Miguel Pacheco
- Informatics Department, University of Minho, Braga, Portugal
| | - Tiago Ferreira
- Informatics Department, University of Minho, Braga, Portugal
| | - Hugo Osório
- i3S—Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ana Raquel Soares
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Mariana Henriques
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| | - Sónia Silva
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
- National Institute for Agrarian and Veterinary Research, Vairão, Vila do Conde, Portugal
| |
Collapse
|
20
|
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
|
21
|
Zhou X, Hilk A, Solis NV, Scott N, Beach A, Soisangwan N, Billings CL, Burrack LS, Filler SG, Selmecki A. Single-cell detection of copy number changes reveals dynamic mechanisms of adaptation to antifungals in Candida albicans. Nat Microbiol 2024; 9:2923-2938. [PMID: 39227665 PMCID: PMC11524788 DOI: 10.1038/s41564-024-01795-7] [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: 12/16/2023] [Accepted: 07/24/2024] [Indexed: 09/05/2024]
Abstract
Genomic copy number changes are associated with antifungal drug resistance and virulence across diverse fungal pathogens, but the rate and dynamics of these genomic changes in the presence of antifungal drugs are unknown. Here we optimized a dual-fluorescent reporter system in the diploid pathogen Candida albicans to quantify haplotype-specific copy number variation (CNV) and loss of heterozygosity (LOH) at the single-cell level with flow cytometry. We followed the frequency and dynamics of CNV and LOH at two distinct genomic locations in the presence and absence of antifungal drugs in vitro and in a murine model of candidiasis. Copy number changes were rapid and dynamic during adaptation to fluconazole and frequently involved competing subpopulations with distinct genotypes. This study provides quantitative evidence for the rapid speed at which diverse genotypes arise and undergo dynamic population-level fluctuations during adaptation to antifungal drugs in vitro and in vivo.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Audrey Hilk
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Norma V Solis
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, USA
| | - Nancy Scott
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Annette Beach
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Natthapon Soisangwan
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Clara L Billings
- Gustavus Adolphus College, Department of Biology, Saint Peter, MN, USA
| | - Laura S Burrack
- Gustavus Adolphus College, Department of Biology, Saint Peter, MN, USA
| | - Scott G Filler
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
22
|
Wang TW, Sofras D, Montelongo-Jauregui D, Paiva TO, Carolus H, Dufrêne YF, Alfaifi AA, McCracken C, Bruno VM, Van Dijck P, Jabra-Rizk MA. Functional redundancy in Candida auris cell surface adhesins crucial for cell-cell interaction and aggregation. Nat Commun 2024; 15:9212. [PMID: 39455573 PMCID: PMC11511831 DOI: 10.1038/s41467-024-53588-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Candida auris is an emerging nosocomial fungal pathogen associated with life-threatening invasive disease due to its persistent colonization, high level of transmissibility and multi-drug resistance. Aggregative and non-aggregative growth phenotypes for C. auris strains with different biofilm forming abilities, drug susceptibilities and virulence characteristics have been described. Using comprehensive transcriptional analysis we identified key cell surface adhesins that were highly upregulated in the aggregative phenotype during in vitro and in vivo grown biofilms using a mouse model of catheter infection. Phenotypic and functional evaluations of generated null mutants demonstrated crucial roles for the adhesins Als4112 and Scf1 in mediating cell-cell adherence, coaggregation and biofilm formation. While individual mutants were largely non-aggregative, in combination cells were able to co-adhere and aggregate, as directly demonstrated by measuring cell adhesion forces using single-cell atomic force spectroscopy. This co-adherence indicates their role as complementary adhesins, which despite their limited similarity, may function redundantly to promote cell-cell interaction and biofilm formation. Functional diversity of cell wall proteins may be a form of regulation that provides the aggregative phenotype of C. auris with flexibility and rapid adaptation to the environment, potentially impacting persistence and virulence.
Collapse
Affiliation(s)
- Tristan W Wang
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Dimitrios Sofras
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Telmo O Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Areej A Alfaifi
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- Department of Restorative and Prosthetic Dental Sciences, College of Dentistry King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vincent M Bruno
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD, USA
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium.
- KU Leuven One-Health Institute, KU Leuven, Leuven, Belgium.
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA.
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
23
|
Hwang IJ, Kwon YJ, Lim HJ, Hong KH, Lee H, Yong D, Won EJ, Byun SA, Lee GY, Kim SH, Song ES, Shin JH. Nosocomial transmission of fluconazole-resistant Candida glabrata bloodstream isolates revealed by whole-genome sequencing. Microbiol Spectr 2024; 12:e0088324. [PMID: 39162519 PMCID: PMC11448407 DOI: 10.1128/spectrum.00883-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
The clonal transmission of fluconazole-resistant Candida glabrata isolates within hospitals has seldom been analyzed by whole-genome sequencing (WGS). We performed WGS on 79 C. glabrata isolates, comprising 31 isolates from three premature infants with persistent C. glabrata bloodstream infection despite antifungal treatment in the same neonatal intensive care unit (NICU) in 2022 and 48 (27 fluconazole-resistant and 21 fluconazole-susceptible dose-dependent) bloodstream isolates from 48 patients in 15 South Korean hospitals from 2010 to 2022. Phylogenetic analysis based on WGS single-nucleotide polymorphisms (SNPs) distinguished the 79 isolates according to multilocus sequence typing (MLST) (17 sequence type [ST]3, 13 ST7, two ST22, 41 ST26, four ST55, and two ST59 isolates) and unveiled two possible clusters of nosocomial transmission among ST26 isolates. One cluster from two premature infants with overlapping NICU hospitalizations in 2022 encompassed 15 fluconazole-resistant isolates harboring pleiotropic drug-resistance transcription factor (Pdr1p) P258L (13 isolates) or N1086I (two isolates), together with 10 fluconazole-susceptible dose-dependent isolates lacking Pdr1p SNPs. The other cluster indicated unforeseen clonal transmission of fluconazole-resistant bloodstream isolates among five patients (four post-lung transplantation and one with diffuse interstitial lung disease) in the same hospital over 8 months. Among these five isolates, four obtained after exposure to azole antifungals harbored distinct Pdr1p SNPs (N1091D, E388Q, K365E, and R376Q). The findings reveal the transmission patterns of clonal bloodstream isolates of C. glabrata among patients undergoing antifungal treatment, exhibiting different levels of fluconazole susceptibility or distinct Pdr1p SNP profiles. IMPORTANCE The prevalence of fluconazole-resistant bloodstream infections caused by Candida glabrata is increasing globally, but the transmission of these resistant strains within hospitals has rarely been documented. Through whole-genome sequencing and epidemiological analyses, this study identified two potential clusters of C. glabrata bloodstream infections within the same hospital, revealing the transmission of clonal C. glabrata strains with different levels of fluconazole susceptibility or distinct transcription factor pleiotropic drug resistance protein 1 (Pdr1p) single-nucleotide polymorphism profiles among patients receiving antifungal therapy.
Collapse
Affiliation(s)
- In Ji Hwang
- Department of Pediatrics, Chonnam National University Medical School, Gwangju, South Korea
| | - Yong Jun Kwon
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Ha Jin Lim
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Ki Ho Hong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyukmin Lee
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, South Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Jeong Won
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung A Byun
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Ga Yeong Lee
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Soo Hyun Kim
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Eun Song Song
- Department of Pediatrics, Chonnam National University Medical School, Gwangju, South Korea
| | - Jong Hee Shin
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
24
|
Rai MN, Lan Q, Parsania C, Rai R, Shirgaonkar N, Chen R, Shen L, Tan K, Wong KH. Temporal transcriptional response of Candida glabrata during macrophage infection reveals a multifaceted transcriptional regulator CgXbp1 important for macrophage response and fluconazole resistance. eLife 2024; 13:e73832. [PMID: 39356739 PMCID: PMC11554308 DOI: 10.7554/elife.73832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/01/2024] [Indexed: 10/04/2024] Open
Abstract
Candida glabrata can thrive inside macrophages and tolerate high levels of azole antifungals. These innate abilities render infections by this human pathogen a clinical challenge. How C. glabrata reacts inside macrophages and what is the molecular basis of its drug tolerance are not well understood. Here, we mapped genome-wide RNA polymerase II (RNAPII) occupancy in C. glabrata to delineate its transcriptional responses during macrophage infection in high temporal resolution. RNAPII profiles revealed dynamic C. glabrata responses to macrophages with genes of specialized pathways activated chronologically at different times of infection. We identified an uncharacterized transcription factor (CgXbp1) important for the chronological macrophage response, survival in macrophages, and virulence. Genome-wide mapping of CgXbp1 direct targets further revealed its multi-faceted functions, regulating not only virulence-related genes but also genes associated with drug resistance. Finally, we showed that CgXbp1 indeed also affects fluconazole resistance. Overall, this work presents a powerful approach for examining host-pathogen interaction and uncovers a novel transcription factor important for C. glabrata's survival in macrophages and drug tolerance.
Collapse
Affiliation(s)
| | - Qing Lan
- Faculty of Health Sciences, University of MacauTaipaChina
| | | | - Rikky Rai
- Faculty of Health Sciences, University of MacauTaipaChina
| | | | - Ruiwen Chen
- Faculty of Health Sciences, University of MacauTaipaChina
| | - Li Shen
- Faculty of Health Sciences, University of MacauTaipaChina
- Gene Expression, Genomics and Bioinformatics Core, Faculty of Health Sciences, University of MacauTaipaChina
| | - Kaeling Tan
- Faculty of Health Sciences, University of MacauTaipaChina
- Gene Expression, Genomics and Bioinformatics Core, Faculty of Health Sciences, University of MacauTaipaChina
| | - Koon Ho Wong
- Faculty of Health Sciences, University of MacauTaipaChina
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau,Avenida da UniversidadeTaipaChina
- MoE Frontiers Science Center for Precision Oncology, University of MacauTaipaChina
| |
Collapse
|
25
|
Pongpom M, Khamto N, Sukantamala P, Kalawil T, Wangsanut T. Identification of Homeobox Transcription Factors in a Dimorphic Fungus Talaromyces marneffei and Protein-Protein Interaction Prediction of RfeB. J Fungi (Basel) 2024; 10:687. [PMID: 39452639 PMCID: PMC11508405 DOI: 10.3390/jof10100687] [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: 07/15/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Talaromyces marneffei is a thermally dimorphic fungus that can cause life-threatening systemic mycoses, particularly in immunocompromised individuals. Fungal homeobox transcription factors control various developmental processes, including the regulation of sexual reproduction, morphology, metabolism, and virulence. However, the function of homeobox proteins in T. marneffei has not been fully explored. Here, we searched the T. marneffei genome for the total homeobox transcription factors and predicted their biological relevance by performing gene expression analysis in different cell types, including conidia, mycelia, yeasts, and during phase transition. RfeB is selected for further computational analysis since (i) its transcripts were differentially expressed in different phases of T. marneffei, and (ii) this protein contains the highly conserved protein-protein interaction region (IR), which could be important for pathobiology and have therapeutic application. To assess the structure-function of the IR region, in silico alanine substitutions were performed at three-conserved IR residues (Asp276, Glu279, and Gln282) of RfeB, generating a triple RfeB mutated protein. Using 3D modeling and molecular dynamics simulations, we compared the protein complex formation of wild-type and mutated RfeB proteins with the putative partner candidate TmSwi5. Our results demonstrated that the mutated RfeB protein exhibited increased free binding energy, elevated protein compactness, and a reduced number of atomic contacts, suggesting disrupted protein stability and interaction. Notably, our model revealed that the IR residues primarily stabilized the RfeB binding sites located in the central region (CR). This computational approach for protein mutagenesis could provide a foundation for future experimental studies on the functional characterization of RfeB and other homeodomain-containing proteins in T. marneffei.
Collapse
Affiliation(s)
- Monsicha Pongpom
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (M.P.)
| | - Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Thitisuda Kalawil
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (M.P.)
| | - Tanaporn Wangsanut
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (M.P.)
| |
Collapse
|
26
|
Kim MJ, Cravener M, Solis N, Filler SG, Mitchell AP. A Brg1-Rme1 circuit in Candida albicans hyphal gene regulation. mBio 2024; 15:e0187224. [PMID: 39078139 PMCID: PMC11389389 DOI: 10.1128/mbio.01872-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
Major Candida albicans virulence traits include its ability to make hyphae, to produce a biofilm, and to damage host cells. These traits depend upon expression of hypha-associated genes. A gene expression comparison among clinical isolates suggested that transcription factor Rme1, established by previous studies to be a positive regulator of chlamydospore formation, may also be a negative regulator of hypha-associated genes. Engineered RME1 overexpression supported this hypothesis, but no relevant rme1Δ/Δ mutant phenotype was detected. We reasoned that Rme1 may function within a specific regulatory pathway. This idea was supported by our finding that an rme1Δ/Δ mutation relieves the need for biofilm regulator Brg1 in biofilm formation. The impact of the rme1Δ/Δ mutation is most prominent under static or "biofilm-like" growth conditions. RNA sequencing (RNA-seq) of cells grown under biofilm-like conditions indicates that Brg1 activates hypha-associated genes indirectly via repression of RME1: hypha-associated gene expression levels are substantially reduced in a brg1Δ/Δ mutant and partially restored in a brg1Δ/Δ rme1Δ/Δ double mutant. An rme1Δ/Δ mutation does not simply bypass Brg1, because iron homeostasis genes depend upon Brg1 regardless of Rme1. Rme1 thus connects Brg1 to the targets relevant to hypha and biofilm formation under biofilm growth conditions.IMPORTANCECandida albicans is a major fungal pathogen of humans, and its ability to grow as a surface-associated biofilm on implanted devices is a common cause of infection. Here, we describe a new regulator of biofilm formation, RME1, whose activity is most prominent under biofilm-like growth conditions.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Max Cravener
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Norma Solis
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Scott G Filler
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Aaron P Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
27
|
Luo G, Zhang J, Wang T, Cui H, Bai Y, Luo J, Zhang J, Zhang M, Di L, Yuan Y, Xiong K, Yu X, Zhang Y, Shen C, Zhu C, Wang Y, Su C, Lu Y. A human commensal-pathogenic fungus suppresses host immunity via targeting TBK1. Cell Host Microbe 2024; 32:1536-1551.e6. [PMID: 39084229 DOI: 10.1016/j.chom.2024.07.003] [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: 03/14/2024] [Revised: 06/11/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024]
Abstract
Candida albicans stably colonizes humans but is the leading cause of hospital-acquired fungemia. Traditionally, masking immunogenic moieties has been viewed as a tactic for immune evasion. Here, we demonstrate that C. albicans blocks type I interferon (IFN-I) signaling via translocating an effector protein Cmi1 into host cells. Mechanistically, Cmi1 binds and inhibits TANK-binding kinase 1 (TBK1) to abrogate IFN-regulatory factor 3 (IRF3) phosphorylation, thereby suppressing the IFN-I cascade. Murine infection with a cmi1 mutant displays an exaggerated IFN-I response in both kidneys and bone-marrow-derived macrophages, leading to rapid fungal clearance and host survival. Remarkably, the lack of CMI1 compromises gut commensalism and increases IFN-I response in mouse colonic cells. These phenotypes of cmi1 are rescued by the depletion of IFN-I receptor. This work establishes the importance of TBK1 inhibition in fungal pathogenesis and reveals that a human commensal-pathogenic fungus significantly impacts host immunity during gut colonization and infection via delivering effector proteins into host cells.
Collapse
Affiliation(s)
- Gang Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Jingkai Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Tianxu Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Hao Cui
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Yukun Bai
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Jianchen Luo
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Jinqiu Zhang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Mao Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Linyan Di
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Yuncong Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kang Xiong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiangtai Yu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yaling Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Chao Shen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Chang Su
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yang Lu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
28
|
Nayak A, Khedri A, Chavarria A, Sanders KN, Ghalei H, Khoshnevis S. Sinefungin, a natural nucleoside analog of S-adenosyl methionine, impairs the pathogenicity of Candida albicans. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:23. [PMID: 39268078 PMCID: PMC11391927 DOI: 10.1038/s44259-024-00040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/19/2024] [Indexed: 09/15/2024]
Abstract
Candida albicans, an opportunistic fungal pathogen, causes life-threatening infections in immunocompromised patients. Current antifungals are limited by toxicity, drug-drug interactions, and emerging resistance, underscoring the importance of identifying novel treatment approaches. Here, we elucidate the impact of sinefungin, an analog of S-adenosyl methionine, on the virulence of C. albicans strain SC5314 and clinical isolates. Our data indicate that sinefungin impairs pathogenic traits of C. albicans including hyphal morphogenesis, biofilm formation, adhesion to epithelial cells, and virulence towards Galleria mellonella, highlighting sinefungin as an avenue for therapeutic intervention. We determine that sinefungin particularly disturbs N6-methyladenosine (m6A) formation. Transcriptome analysis of C. albicans hyphae upon sinefungin treatment reveals an increase in transcripts related to the yeast form and decrease in those associated with hyphae formation and virulence. Collectively, our data propose sinefungin as a potent molecule against C. albicans and emphasize further exploration of post-transcriptional control mechanisms of pathogenicity for antifungal design.
Collapse
Affiliation(s)
- Anushka Nayak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Azam Khedri
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Alejandro Chavarria
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Kyla N. Sanders
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Sohail Khoshnevis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
29
|
Correia I, Oliveira C, Reis A, Guimarães AR, Aveiro S, Domingues P, Bezerra AR, Vitorino R, Moura G, Santos MAS. A Proteogenomic Pipeline for the Analysis of Protein Biosynthesis Errors in the Human Pathogen Candida albicans. Mol Cell Proteomics 2024; 23:100818. [PMID: 39047911 PMCID: PMC11420639 DOI: 10.1016/j.mcpro.2024.100818] [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: 11/24/2023] [Revised: 03/20/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Candida albicans is a diploid pathogen known for its ability to live as a commensal fungus in healthy individuals but causing both superficial infections and disseminated candidiasis in immunocompromised patients where it is associated with high morbidity and mortality. Its success in colonizing the human host is attributed to a wide range of virulence traits that modulate interactions between the host and the pathogen, such as optimal growth rate at 37 °C, the ability to switch between yeast and hyphal forms, and a remarkable genomic and phenotypic plasticity. A fascinating aspect of its biology is a prominent heterogeneous proteome that arises from frequent genomic rearrangements, high allelic variation, and high levels of amino acid misincorporations in proteins. This leads to increased morphological and physiological phenotypic diversity of high adaptive potential, but the scope of such protein mistranslation is poorly understood due to technical difficulties in detecting and quantifying amino acid misincorporation events in complex protein samples. We have developed and optimized mass spectrometry and bioinformatics pipelines capable of identifying rare amino acid misincorporation events at the proteome level. We have also analyzed the proteomic profile of an engineered C. albicans strain that exhibits high level of leucine misincorporation at protein CUG sites and employed an in vivo quantitative gain-of-function fluorescence reporter system to validate our LC-MS/MS data. C. albicans misincorporates amino acids above the background level at protein sites of diverse codons, particularly at CUG, confirming our previous data on the quantification of leucine incorporation at single CUG sites of recombinant reporter proteins, but increasing misincorporation of Leucine at these sites does not alter the translational fidelity of the other codons. These findings indicate that the C. albicans statistical proteome exceeds prior estimates, suggesting that its highly plastic phenome may also be modulated by environmental factors due to translational ambiguity.
Collapse
Affiliation(s)
- Inês Correia
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal.
| | - Carla Oliveira
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal
| | - Andreia Reis
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal
| | - Ana Rita Guimarães
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal
| | - Susana Aveiro
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Pedro Domingues
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Ana Rita Bezerra
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal
| | - Gabriela Moura
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal
| | - Manuel A S Santos
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences (DCM), University of Aveiro, Aveiro, Portugal; Multidisciplinary Institute of Ageing (MIA-Portugal), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
30
|
Després PC, Shapiro RS, Cuomo CA. New approaches to tackle a rising problem: Large-scale methods to study antifungal resistance. PLoS Pathog 2024; 20:e1012478. [PMID: 39236046 PMCID: PMC11376582 DOI: 10.1371/journal.ppat.1012478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Affiliation(s)
- Philippe C Després
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
31
|
Xiong EH, Zhang X, Yan H, Ward HN, Lin ZY, Wong CJ, Fu C, Gingras AC, Noble SM, Robbins N, Myers CL, Cowen LE. Functional genomic analysis of genes important for Candida albicans fitness in diverse environmental conditions. Cell Rep 2024; 43:114601. [PMID: 39126650 PMCID: PMC11416860 DOI: 10.1016/j.celrep.2024.114601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Fungal pathogens such as Candida albicans pose a significant threat to human health with limited treatment options available. One strategy to expand the therapeutic target space is to identify genes important for pathogen growth in host-relevant environments. Here, we leverage a pooled functional genomic screening strategy to identify genes important for fitness of C. albicans in diverse conditions. We identify an essential gene with no known Saccharomyces cerevisiae homolog, C1_09670C, and demonstrate that it encodes subunit 3 of replication factor A (Rfa3). Furthermore, we apply computational analyses to identify functionally coherent gene clusters and predict gene function. Through this approach, we predict the cell-cycle-associated function of C3_06880W, a previously uncharacterized gene required for fitness specifically at elevated temperatures, and follow-up assays confirm that C3_06880W encodes Iml3, a component of the C. albicans kinetochore with roles in virulence in vivo. Overall, this work reveals insights into the vulnerabilities of C. albicans.
Collapse
Affiliation(s)
- Emily H Xiong
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiang Zhang
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Huijuan Yan
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Henry N Ward
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Bioinformatics and Computational Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhen-Yuan Lin
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Cassandra J Wong
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Ci Fu
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Suzanne M Noble
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Bioinformatics and Computational Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
32
|
Lash E, Maufrais C, Janbon G, Robbins N, Herzel L, Cowen LE. The spliceosome impacts morphogenesis in the human fungal pathogen Candida albicans. mBio 2024; 15:e0153524. [PMID: 38980041 PMCID: PMC11323467 DOI: 10.1128/mbio.01535-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/10/2024] Open
Abstract
At human body temperature, the fungal pathogen Candida albicans can transition from yeast to filamentous morphologies in response to host-relevant cues. Additionally, elevated temperatures encountered during febrile episodes can independently induce C. albicans filamentation. However, the underlying genetic pathways governing this developmental transition in response to elevated temperatures remain largely unexplored. Here, we conducted a functional genomic screen to unravel the genetic mechanisms orchestrating C. albicans filamentation specifically in response to elevated temperature, implicating 45% of genes associated with the spliceosome or pre-mRNA splicing in this process. Employing RNA-Seq to elucidate the relationship between mRNA splicing and filamentation, we identified greater levels of intron retention in filaments compared to yeast, which correlated with reduced expression of the affected genes. Intriguingly, homozygous deletion of a gene encoding a spliceosome component important for filamentation (PRP19) caused even greater levels of intron retention compared with wild type and displayed globally dysregulated gene expression. This suggests that intron retention is a mechanism for fine-tuning gene expression during filamentation, with perturbations of the spliceosome exacerbating this process and blocking filamentation. Overall, this study unveils a novel biological process governing C. albicans filamentation, providing new insights into the complex regulation of this key virulence trait.IMPORTANCEFungal pathogens such as Candida albicans can cause serious infections with high mortality rates in immunocompromised individuals. When C. albicans is grown at temperatures encountered during human febrile episodes, yeast cells undergo a transition to filamentous cells, and this process is key to its virulence. Here, we expanded our understanding of how C. albicans undergoes filamentation in response to elevated temperature and identified many genes involved in mRNA splicing that positively regulate filamentation. Through transcriptome analyses, we found that intron retention is a mechanism for fine-tuning gene expression in filaments, and perturbation of the spliceosome exacerbates intron retention and alters gene expression substantially, causing a block in filamentation. This work adds to the growing body of knowledge on the role of introns in fungi and provides new insights into the cellular processes that regulate a key virulence trait in C. albicans.
Collapse
Affiliation(s)
- Emma Lash
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Corinne Maufrais
- Unité Biologie des ARN des Pathogènes Fongiques, Institut Pasteur, Université Paris Cité, Paris, France
- HUB Bioinformatique et Biostatistique, Institut Pasteur, Université Paris Cité, Paris, France
| | - Guilhem Janbon
- Unité Biologie des ARN des Pathogènes Fongiques, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Lydia Herzel
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Wickramasinghe DN, Lyon CM, Lee S, Hepworth OW, Priest EL, Maufrais C, Ryan AP, Permal E, Sullivan D, McManus BA, Hube B, Butler G, d'Enfert C, Naglik JR, Richardson JP. Variations in candidalysin amino acid sequence influence toxicity and host responses. mBio 2024; 15:e0335123. [PMID: 38953356 PMCID: PMC11323794 DOI: 10.1128/mbio.03351-23] [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] [Received: 12/09/2023] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
Candida albicans causes millions of mucosal infections in humans annually. Hyphal overgrowth on mucosal surfaces is frequently associated with tissue damage caused by candidalysin, a secreted peptide toxin that destabilizes the plasma membrane of host cells thereby promoting disease and immunopathology. Candidalysin was first identified in C. albicans strain SC5314, but recent investigations have revealed candidalysin "variants" of differing amino acid sequence in isolates of C. albicans, and the related species C. dubliniensis, and C tropicalis, suggesting that sequence variation among candidalysins may be widespread in natural populations of these Candida species. Here, we analyzed ECE1 gene sequences from 182 C. albicans isolates, 10 C. dubliniensis isolates, and 78 C. tropicalis isolates and identified 10, 3, and 2 candidalysin variants in these species, respectively. Application of candidalysin variants to epithelial cells revealed differences in the ability to cause cellular damage, changes in metabolic activity, calcium influx, MAPK signalling, and cytokine secretion, while biophysical analyses indicated that variants exhibited differences in their ability to interact with and permeabilize a membrane. This study identifies candidalysin variants with differences in biological activity that are present in medically relevant Candida species. IMPORTANCE Fungal infections are a significant burden to health. Candidalysin is a toxin produced by Candida albicans that damages host tissues, facilitating infection. Previously, we demonstrated that candidalysins exist in the related species C. dubliniensis and C. tropicalis, thereby identifying these molecules as a toxin family. Recent genomic analyses have highlighted the presence of a small number of candidalysin "variant" toxins, which have different amino acid sequences to those originally identified. Here, we screened genome sequences of isolates of C. albicans, C. dubliniensis, and C. tropicalis and identified candidalysin variants in all three species. When applied to epithelial cells, candidalysin variants differed in their ability to cause damage, activate intracellular signaling pathways, and induce innate immune responses, while biophysical analysis revealed differences in the ability of candidalysin variants to interact with lipid bilayers. These findings suggest that intraspecies variation in candidalysin amino acid sequence may influence fungal pathogenicity.
Collapse
Affiliation(s)
- Don N. Wickramasinghe
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Claire M. Lyon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Sejeong Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Olivia W. Hepworth
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Emily L. Priest
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Corinne Maufrais
- Institut Pasteur, Université Paris Cité, INRAe USC 2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Adam P. Ryan
- School of Biomedical and Biomolecular Science and UCD Conway Institute of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Emmanuelle Permal
- Institut Pasteur, Université Paris Cité, INRAe USC 2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Derek Sullivan
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Brenda A. McManus
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Geraldine Butler
- School of Biomedical and Biomolecular Science and UCD Conway Institute of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Christophe d'Enfert
- Institut Pasteur, Université Paris Cité, INRAe USC 2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
34
|
O’Connor-Moneley J, Fletcher J, Bean C, Parker J, Kelly SL, Moran GP, Sullivan DJ. Deletion of the Candida albicans TLO gene family results in alterations in membrane sterol composition and fluconazole tolerance. PLoS One 2024; 19:e0308665. [PMID: 39121069 PMCID: PMC11315338 DOI: 10.1371/journal.pone.0308665] [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: 07/03/2024] [Accepted: 07/27/2024] [Indexed: 08/11/2024] Open
Abstract
Development of resistance and tolerance to antifungal drugs in Candida albicans can compromise treatment of infections caused by this pathogenic yeast species. The uniquely expanded C. albicans TLO gene family is comprised of 14 paralogous genes which encode Med2, a subunit of the multiprotein Mediator complex which is involved in the global control of transcription. This study investigates the acquisition of fluconazole tolerance in a mutant in which the entire TLO gene family has been deleted. This phenotype was reversed to varying degrees upon reintroduction of representative members of the alpha- and beta-TLO clades (i.e. TLO1 and TLO2), but not by TLO11, a gamma-clade representative. Comparative RNA sequencing analysis revealed changes in the expression of genes involved in a range of cellular functions, including ergosterol biosynthesis, mitochondrial function, and redox homeostasis. This was supported by the results of mass spectrometry analysis, which revealed alterations in sterol composition of the mutant cell membrane. Our data suggest that members of the C. albicans TLO gene family are involved in the control of ergosterol biosynthesis and mitochondrial function and may play a role in the responses of C. albicans to azole antifungal agents.
Collapse
Affiliation(s)
- James O’Connor-Moneley
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Jessica Fletcher
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Cody Bean
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Josie Parker
- Institute of Life Science, Singleton Campus, Swansea University, Swansea, Wales, United Kingdom
| | - Steven L. Kelly
- Institute of Life Science, Singleton Campus, Swansea University, Swansea, Wales, United Kingdom
| | - Gary P. Moran
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Derek J. Sullivan
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
35
|
Farheen A, Case NT, MacAlpine J, Fu C, Robbins N, Cowen LE. The putative prenyltransferase Nus1 is required for filamentation in the human fungal pathogen Candida albicans. G3 (BETHESDA, MD.) 2024; 14:jkae124. [PMID: 38874344 PMCID: PMC11304969 DOI: 10.1093/g3journal/jkae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/03/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
Candida albicans is a major fungal pathogen of humans that can cause serious systemic infections in vulnerable immunocompromised populations. One of its virulence attributes is its capacity to transition between yeast and filamentous morphologies, but our understanding of this process remains incomplete. Here, we analyzed data from a functional genomic screen performed with the C. albicans Gene Replacement And Conditional Expression collection to identify genes crucial for morphogenesis in host-relevant conditions. Through manual scoring of microscopy images coupled with analysis of each image using a deep learning-based method termed Candescence, we identified 307 genes important for filamentation in tissue culture medium at 37°C with 5% CO2. One such factor was orf19.5963, which is predicted to encode the prenyltransferase Nus1 based on sequence homology to Saccharomyces cerevisiae. We further showed that Nus1 and its predicted interacting partner Rer2 are important for filamentation in multiple liquid filament-inducing conditions as well as for wrinkly colony formation on solid agar. Finally, we highlight that Nus1 and Rer2 likely govern C. albicans morphogenesis due to their importance in intracellular trafficking, as well as maintaining lipid homeostasis. Overall, this work identifies Nus1 and Rer2 as important regulators of C. albicans filamentation and highlights the power of functional genomic screens in advancing our understanding of gene function in human fungal pathogens.
Collapse
Affiliation(s)
- Aiman Farheen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Nicola T Case
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Ci Fu
- 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
|
36
|
Lindemann-Perez E, Perez JC. Candida albicans natural diversity: a resource to dissect fungal commensalism and pathogenesis. Curr Opin Microbiol 2024; 80:102493. [PMID: 38833793 DOI: 10.1016/j.mib.2024.102493] [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: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Candida albicans is a ubiquitous fungus of humans. It is not only a component of the oral and intestinal microbiota of most healthy adults but also a major cause of mucosal disorders and life-threatening disseminated infections. Until recently, research on the biology and pathogenesis of the fungus was largely based on a single clinical isolate. We review investigations that have started to dissect a diverse set of C. albicans strains. Using different approaches to leverage the species' phenotypic and/or genetic diversity, these studies illuminate the wide range of interactions between fungus and host. While connecting genetic variants to phenotypes of interest remains challenging, research on C. albicans' natural diversity is central to understand fungal commensalism and pathogenesis.
Collapse
Affiliation(s)
- Elena Lindemann-Perez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | - J Christian Perez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA.
| |
Collapse
|
37
|
Acosta-Zaldívar M, Qi W, Mishra A, Roy U, King WR, Li Y, Patton-Vogt J, Anderson MZ, Köhler JR. Candida albicans' inorganic phosphate transport and evolutionary adaptation to phosphate scarcity. PLoS Genet 2024; 20:e1011156. [PMID: 39137212 PMCID: PMC11343460 DOI: 10.1371/journal.pgen.1011156] [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: 01/29/2024] [Revised: 08/23/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Phosphorus is essential in all cells' structural, metabolic and regulatory functions. For fungal cells that import inorganic phosphate (Pi) up a steep concentration gradient, surface Pi transporters are critical capacitators of growth. Fungi must deploy Pi transporters that enable optimal Pi uptake in pH and Pi concentration ranges prevalent in their environments. Single, triple and quadruple mutants were used to characterize the four Pi transporters we identified for the human fungal pathogen Candida albicans, which must adapt to alkaline conditions during invasion of the host bloodstream and deep organs. A high-affinity Pi transporter, Pho84, was most efficient across the widest pH range while another, Pho89, showed high-affinity characteristics only within one pH unit of neutral. Two low-affinity Pi transporters, Pho87 and Fgr2, were active only in acidic conditions. Only Pho84 among the Pi transporters was clearly required in previously identified Pi-related functions including Target of Rapamycin Complex 1 signaling, oxidative stress resistance and hyphal growth. We used in vitro evolution and whole genome sequencing as an unbiased forward genetic approach to probe adaptation to prolonged Pi scarcity of two quadruple mutant lineages lacking all 4 Pi transporters. Lineage-specific genomic changes corresponded to divergent success of the two lineages in fitness recovery during Pi limitation. Initial, large-scale genomic alterations like aneuploidies and loss of heterozygosity eventually resolved, as populations gained small-scale mutations. Severity of some phenotypes linked to Pi starvation, like cell wall stress hypersensitivity, decreased in parallel to evolving populations' fitness recovery in Pi scarcity, while severity of others like membrane stress responses diverged from Pi scarcity fitness. Among preliminary candidate genes for contributors to fitness recovery, those with links to TORC1 were overrepresented. Since Pi homeostasis differs substantially between fungi and humans, adaptive processes to Pi deprivation may harbor small-molecule targets that impact fungal growth, stress resistance and virulence.
Collapse
Affiliation(s)
- Maikel Acosta-Zaldívar
- Division of Infectious Diseases, Boston Children’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wanjun Qi
- Division of Infectious Diseases, Boston Children’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Abhishek Mishra
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Udita Roy
- Division of Infectious Diseases, Boston Children’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - William R. King
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States of America
| | - Yuping Li
- Department of Microbiology and Immunology, University of California, San Francisco, California, United States of America
| | - Jana Patton-Vogt
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States of America
| | - Matthew Z. Anderson
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medical Genetics, Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Julia R. Köhler
- Division of Infectious Diseases, Boston Children’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
38
|
Vande Zande P, Gautier C, Kawar N, Maufrais C, Metzner K, Wash E, Beach AK, Bracken R, Maciel EI, Pereira de Sá N, Fernandes CM, Solis NV, Del Poeta M, Filler SG, Berman J, Ene IV, Selmecki A. Step-wise evolution of azole resistance through copy number variation followed by KSR1 loss of heterozygosity in Candida albicans. PLoS Pathog 2024; 20:e1012497. [PMID: 39213436 PMCID: PMC11392398 DOI: 10.1371/journal.ppat.1012497] [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: 03/12/2024] [Revised: 09/12/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Antimicrobial drug resistance poses a global health threat, requiring a deeper understanding of the evolutionary processes that lead to its emergence in pathogens. Complex evolutionary dynamics involve multiple mutations that can result in cooperative or competitive (clonal interference) effects. Candida albicans, a major fungal pathogen, displays high rates of copy number variation (CNV) and loss of heterozygosity (LOH). CNV and LOH events involve large numbers of genes and could synergize during evolutionary adaptation. Understanding the contributions of CNV and LOH to antifungal drug adaptation is challenging, especially in the context of whole-population genome sequencing. Here, we document the sequential evolution of fluconazole tolerance and then resistance in a C. albicans isolate involving an initial CNV on chromosome 4, followed by an LOH on chromosome R that involves KSR1. Similar LOH events involving KSR1, which encodes a reductase in the sphingolipid biosynthesis pathway, were also detected in independently evolved fluconazole resistant isolates. We dissect the specific KSR1 codons that affect fluconazole resistance and tolerance. The combination of the chromosome 4 CNV and KSR1 LOH results in a >500-fold decrease in azole susceptibility relative to the progenitor, illustrating a compelling example of rapid, yet step-wise, interplay between CNV and LOH in drug resistance evolution.
Collapse
Affiliation(s)
- Pétra Vande Zande
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cécile Gautier
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
| | - Nora Kawar
- Shmunis School of Biotechnology and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Corinne Maufrais
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
- Institut Pasteur Bioinformatic Hub, Université Paris Cité, Paris, France
| | - Katura Metzner
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Elizabeth Wash
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Annette K. Beach
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ryan Bracken
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Eli Isael Maciel
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
| | - Nívea Pereira de Sá
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
| | - Caroline Mota Fernandes
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
| | - Norma V. Solis
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, California, United States of America
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- Veterans Administration Medical Center, Northport, New York, United States of America
| | - Scott G. Filler
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Judith Berman
- Shmunis School of Biotechnology and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Iuliana V. Ene
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
39
|
Ramírez-Zavala B, Krüger I, Schwanfelder S, Barker KS, Rogers PD, Morschhäuser J. The zinc cluster transcription factor Znc1 regulates Rta3-dependent miltefosine resistance in Candida albicans. mSphere 2024; 9:e0027024. [PMID: 38860767 PMCID: PMC11288014 DOI: 10.1128/msphere.00270-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/03/2024] [Indexed: 06/12/2024] Open
Abstract
Zinc cluster transcription factors (ZCFs) are a family of transcription regulators that are almost exclusively found in the fungal kingdom. Activating mutations in the ZCFs Mrr1, Tac1, and Upc2 frequently cause acquired resistance to the widely used antifungal drug fluconazole in the pathogenic yeast Candida albicans. Similar to a hyperactive Tac1, a constitutively active form of the ZCF Znc1 causes increased fluconazole resistance by upregulating the multidrug efflux pump-encoding gene CDR1. Hyperactive forms of both Tac1 and Znc1 also cause overexpression of RTA3, which encodes a seven-transmembrane receptor protein involved in the regulation of asymmetric lipid distribution in the plasma membrane. RTA3 expression is also upregulated by miltefosine, an antiparasitic drug that is active against fungal pathogens and considered for treatment of invasive candidiasis, and rta3Δ mutants are hypersensitive to miltefosine. We found that activated forms of both Tac1 and Znc1 confer increased miltefosine resistance, which was dependent on RTA3 whereas CDR1 was dispensable. Intriguingly, the induction of RTA3 expression by miltefosine depended on Znc1, but not Tac1, in contrast to the known Tac1-dependent RTA3 upregulation by fluphenazine. In line with this observation, znc1Δ mutants were hypersensitive to miltefosine, whereas tac1Δ mutants showed wild-type tolerance. Forced expression of RTA3 reverted the hypersensitivity of znc1Δ mutants, demonstrating that the hypersensitivity was caused by the inability of the mutants to upregulate RTA3 in response to the drug. These findings establish Znc1 as a key regulator of miltefosine-induced RTA3 expression that is important for wild-type miltefosine tolerance. IMPORTANCE Transcription factors are central regulators of gene expression, and knowledge about which transcription factor regulates specific genes in response to a certain signal is important to understand the behavior of organisms. In the pathogenic yeast Candida albicans, the RTA3 gene is required for wild-type tolerance of miltefosine, an antiparasitic drug that is considered for treatment of invasive candidiasis. Activated forms of the transcription factors Tac1 and Znc1 cause constitutive overexpression of RTA3 and thereby increased miltefosine resistance, but only Tac1 mediates upregulation of RTA3 in response to the known inducer fluphenazine. RTA3 expression is also induced by miltefosine, and we found that this response depends on Znc1, whereas Tac1 is dispensable. Consequently, znc1Δ mutants were hypersensitive to miltefosine, whereas tac1Δ mutants showed wild-type tolerance. These findings demonstrate that Znc1 is the key regulator of RTA3 expression in response to miltefosine that is important for wild-type miltefosine tolerance.
Collapse
Affiliation(s)
| | - Ines Krüger
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Sonja Schwanfelder
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Katherine S. Barker
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - P. David Rogers
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Joachim Morschhäuser
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
40
|
Xiong L, Goerlich K, Do E, Mitchell AP. Strain variation in the Candida albicans iron limitation response. mSphere 2024; 9:e0037224. [PMID: 38980069 PMCID: PMC11288005 DOI: 10.1128/msphere.00372-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/16/2024] [Indexed: 07/10/2024] Open
Abstract
Iron acquisition is critical for pathogens to proliferate during invasive infection, and the human fungal pathogen Candida albicans is no exception. The iron regulatory network, established in reference strain SC5314 and derivatives, includes the central player Sef1, a transcription factor that activates iron acquisition genes in response to iron limitation. Here, we explored potential variation in this network among five diverse C. albicans strains through mutant analysis, Nanostring gene expression profiling, and, for two strains, RNA-Seq. Our findings highlight four features that may inform future studies of natural variation and iron acquisition in this species. (i) Conformity: In all strains, major iron acquisition genes are upregulated during iron limitation, and a sef1Δ/Δ mutation impairs that response and growth during iron limitation. (ii) Response variation: Some aspects of the iron limitation response vary among strains, notably the activation of hypha-associated genes. As this gene set is tied to tissue damage and virulence, variation may impact the progression of infection. (iii) Genotype-phenotype variation: The impact of a sef1Δ/Δ mutation on cell wall integrity varies, and for the two strains examined the phenotype correlated with sef1Δ/Δ impact on several cell wall integrity genes. (iv) Phenotype discovery: DNA repair genes were induced modestly by iron limitation in sef1Δ/Δ mutants, with fold changes we would usually ignore. However, the response occurred in both strains tested and was reminiscent of a much stronger response described in Cryptococcus neoformans, a suggestion that it may have biological meaning. In fact, we observed that the iron limitation of a sef1Δ/Δ mutant caused recessive phenotypes to emerge at two heterozygous loci. Overall, our results show that a network that is critical for pathogen proliferation presents variation outside of its core functions.IMPORTANCEA key virulence factor of Candida albicans is the ability to maintain iron homeostasis in the host where iron is scarce. We focused on a central iron regulator, SEF1. We found that iron regulator Sef1 is required for growth, cell wall integrity, and genome integrity during iron limitation. The novel aspect of this work is the characterization of strain variation in a circuit that is required for survival in the host and the connection of iron acquisition to genome integrity in C. albicans.
Collapse
Affiliation(s)
- Liping Xiong
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | | | - Eunsoo Do
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
41
|
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
|
42
|
Ksiezopolska E, Schikora-Tamarit MÀ, Carlos Nunez-Rodriguez J, Gabaldón T. Long-term stability of acquired drug resistance and resistance associated mutations in the fungal pathogen Nakaseomyces glabratus ( Candida glabrata). Front Cell Infect Microbiol 2024; 14:1416509. [PMID: 39077431 PMCID: PMC11284152 DOI: 10.3389/fcimb.2024.1416509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
The limited number of available antifungal drugs and the increasing number of fungal isolates that show drug or multidrug resistance pose a serious medical threat. Several yeast pathogens, such as Nakaseomyces glabratus (Candida glabrata), show a remarkable ability to develop drug resistance during treatment through the acquisition of genetic mutations. However, how stable this resistance and the underlying mutations are in non-selective conditions remains poorly characterized. The stability of acquired drug resistance has fundamental implications for our understanding of the appearance and spread of drug-resistant outbreaks and for defining efficient strategies to combat them. Here, we used an in vitro evolution approach to assess the stability under optimal growth conditions of resistance phenotypes and resistance-associated mutations that were previously acquired under exposure to antifungals. Our results reveal a remarkable stability of the resistant phenotype and the underlying mutations in a significant number of evolved populations, which conserved their phenotype for at least two months in the absence of drug-selective pressure. We observed a higher stability of anidulafungin resistance over fluconazole resistance, and of resistance-conferring point mutations as compared with aneuploidies. In addition, we detected accumulation of novel mutations in previously altered resistance-associated genes in non-selective conditions, which suggest a possible compensatory role. We conclude that acquired resistance, particularly to anidulafungin, is a long-lasting phenotype, which has important implications for the persistence and propagation of drug-resistant clinical outbreaks.
Collapse
Affiliation(s)
- Ewa Ksiezopolska
- Department of Life Sciences, Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Department of Mechanisms of Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Miquel Àngel Schikora-Tamarit
- Department of Life Sciences, Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Department of Mechanisms of Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Juan Carlos Nunez-Rodriguez
- Department of Life Sciences, Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Department of Mechanisms of Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Toni Gabaldón
- Department of Life Sciences, Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Department of Mechanisms of Disease, 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
- Department of CIBERinfect, Centro Investigación Biomédica En Red de Enfermedades Infecciosas, Barcelona, Spain
| |
Collapse
|
43
|
Determann B, Fu J, Wickes BL. Development of a Shuttle Vector That Transforms at High Frequency for the Emerging Human Fungal Pathogen: Candida auris. J Fungi (Basel) 2024; 10:477. [PMID: 39057362 PMCID: PMC11278357 DOI: 10.3390/jof10070477] [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: 06/17/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Routine molecular manipulation of any organism is inefficient and difficult without the existence of a plasmid. Although transformation is possible in C. auris, no plasmids are available that can serve as cloning or shuttle vectors. C. auris centromeres have been well characterized but have not been explored further as molecular tools. We tested C. auris centromeric sequences to identify which, if any, could be used to create a plasmid that was stably maintained after transformation. We cloned all seven C. auris centromeric sequences and tested them for transformation frequency and stability. Transformation frequency varied significantly; however, one was found to transform at a very high frequency. A 1.7 Kb subclone of this sequence was used to construct a shuttle vector. The vector was stable with selection and maintained at ~1 copy per cell but could be easily lost when selection was removed, which suggested that the properties of the centromeric sequence were more Autonomously Replicating Sequence (ARS)-like than centromere-like when part of a plasmid. Rescue of this plasmid from transformed C. auris cells into E. coli revealed that it remained intact after the initial C. auris transformation, even when carrying large inserts. The plasmid was found to be able to transform all four clades of C. auris, with varying frequencies. This plasmid is an important new reagent in the C. auris molecular toolbox, which will enhance the investigation of this human fungal pathogen.
Collapse
Affiliation(s)
| | | | - Brian L. Wickes
- The Department of Microbiology, Immunology, and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA (J.F.)
| |
Collapse
|
44
|
Zhou X, Hilk A, Solis NV, Pereira De Sa N, Hogan BM, Bierbaum TA, Del Poeta M, Filler SG, Burrack LS, Selmecki A. Erg251 has complex and pleiotropic effects on sterol composition, azole susceptibility, filamentation, and stress response phenotypes. PLoS Pathog 2024; 20:e1012389. [PMID: 39078851 PMCID: PMC11315318 DOI: 10.1371/journal.ppat.1012389] [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: 01/31/2024] [Revised: 08/09/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
Ergosterol is essential for fungal cell membrane integrity and growth, and numerous antifungal drugs target ergosterol. Inactivation or modification of ergosterol biosynthetic genes can lead to changes in antifungal drug susceptibility, filamentation and stress response. Here, we found that the ergosterol biosynthesis gene ERG251 is a hotspot for point mutations during adaptation to antifungal drug stress within two distinct genetic backgrounds of Candida albicans. Heterozygous point mutations led to single allele dysfunction of ERG251 and resulted in azole tolerance in both genetic backgrounds. This is the first known example of point mutations causing azole tolerance in C. albicans. Importantly, single allele dysfunction of ERG251 in combination with recurrent chromosome aneuploidies resulted in bona fide azole resistance. Homozygous deletions of ERG251 caused increased fitness in low concentrations of fluconazole and decreased fitness in rich medium, especially at low initial cell density. Homozygous deletions of ERG251 resulted in accumulation of ergosterol intermediates consistent with the fitness defect in rich medium. Dysfunction of ERG251, together with FLC exposure, resulted in decreased accumulation of the toxic sterol (14-ɑ-methylergosta-8,24(28)-dien-3β,6α-diol) and increased accumulation of non-toxic alternative sterols. The altered sterol composition of the ERG251 mutants had pleiotropic effects on transcription, filamentation, and stress responses including cell membrane, osmotic and oxidative stress. Interestingly, while dysfunction of ERG251 resulted in azole tolerance, it also led to transcriptional upregulation of ZRT2, a membrane-bound Zinc transporter, in the presence of FLC, and overexpression of ZRT2 is sufficient to increase azole tolerance in wild-type C. albicans. Finally, in a murine model of systemic infection, homozygous deletion of ERG251 resulted in decreased virulence while the heterozygous deletion mutants maintain their pathogenicity. Overall, this study demonstrates that single allele dysfunction of ERG251 is a recurrent and effective mechanism of acquired azole tolerance. We propose that altered sterol composition resulting from ERG251 dysfunction mediates azole tolerance as well as pleiotropic effects on stress response, filamentation and virulence.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Audrey Hilk
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Norma V. Solis
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, California, United States of America
| | - Nivea Pereira De Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
| | - Bode M. Hogan
- Gustavus Adolphus College, Department of Biology, Saint Peter, Minnesota, USA
| | - Tessa A. Bierbaum
- Gustavus Adolphus College, Department of Biology, Saint Peter, Minnesota, USA
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- Veterans Administration Medical Center, Northport, New York, United States of America
| | - Scott G. Filler
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Laura S. Burrack
- Gustavus Adolphus College, Department of Biology, Saint Peter, Minnesota, USA
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
45
|
Pham D, Sivalingam V, Tang HM, Montgomery JM, Chen SCA, Halliday CL. Molecular Diagnostics for Invasive Fungal Diseases: Current and Future Approaches. J Fungi (Basel) 2024; 10:447. [PMID: 39057332 PMCID: PMC11278267 DOI: 10.3390/jof10070447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Invasive fungal diseases (IFDs) comprise a growing healthcare burden, especially given the expanding population of immunocompromised hosts. Early diagnosis of IFDs is required to optimise therapy with antifungals, especially in the setting of rising rates of antifungal resistance. Molecular techniques including nucleic acid amplification tests and whole genome sequencing have potential to offer utility in overcoming limitations with traditional phenotypic testing. However, standardisation of methodology and interpretations of these assays is an ongoing undertaking. The utility of targeted Aspergillus detection has been well-defined, with progress in investigations into the role of targeted assays for Candida, Pneumocystis, Cryptococcus, the Mucorales and endemic mycoses. Likewise, whilst broad-range polymerase chain reaction assays have been in use for some time, pathology stewardship and optimising diagnostic yield is a continuing exercise. As costs decrease, there is also now increased access and experience with whole genome sequencing, including metagenomic sequencing, which offers unparalleled resolution especially in the investigations of potential outbreaks. However, their role in routine diagnostic use remains uncommon and standardisation of techniques and workflow are required for wider implementation.
Collapse
Affiliation(s)
- David Pham
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (D.P.)
| | - Varsha Sivalingam
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (D.P.)
| | - Helen M. Tang
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (D.P.)
| | - James M. Montgomery
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (D.P.)
| | - Sharon C.-A. Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (D.P.)
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Catriona L. Halliday
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (D.P.)
| |
Collapse
|
46
|
Saha D, Gregor JB, Hoda S, Eastman KE, Navarrete M, Wisecaver JH, Briggs SD. Candida glabrata maintains two Hap1 homologs, Zcf27 and Zcf4, for distinct roles in ergosterol gene regulation to mediate sterol homeostasis under azole and hypoxic conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599910. [PMID: 38979343 PMCID: PMC11230168 DOI: 10.1101/2024.06.20.599910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Candida glabrata exhibits innate resistance to azole antifungal drugs but also has the propensity to rapidly develop clinical drug resistance. Azole drugs, which target Erg11, is one of the three major classes of antifungals used to treat Candida infections. Despite their widespread use, the mechanism controlling azole-induced ERG gene expression and drug resistance in C. glabrata has primarily revolved around Upc2 and/or Pdr1. In this study, we determined the function of two zinc cluster transcription factors, Zcf27 and Zcf4, as direct but distinct regulators of ERG genes. Our phylogenetic analysis revealed C. glabrata Zcf27 and Zcf4 as the closest homologs to Saccharomyces cerevisiae Hap1. Hap1 is a known zinc cluster transcription factor in S. cerevisiae in controlling ERG gene expression under aerobic and hypoxic conditions. Interestingly, when we deleted HAP1 or ZCF27 in either S. cerevisiae or C. glabrata, respectively, both deletion strains showed altered susceptibility to azole drugs, whereas the strain deleted for ZCF4 did not exhibit azole susceptibility. We also determined that the increased azole susceptibility in a zcf27Δ strain is attributed to decreased azole-induced expression of ERG genes, resulting in decreased levels of total ergosterol. Surprisingly, Zcf4 protein expression is barely detected under aerobic conditions but is specifically induced under hypoxic conditions. However, under hypoxic conditions, Zcf4 but not Zcf27 was directly required for the repression of ERG genes. This study provides the first demonstration that Zcf27 and Zcf4 have evolved to serve distinct roles allowing C. glabrata to adapt to specific host and environmental conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Scott D Briggs
- Department of Biochemistry
- Purdue University Institute for Cancer Research
| |
Collapse
|
47
|
Rai LS, Chauvel M, Sanchez H, van Wijlick L, Maufrais C, Cokelaer T, Sertour N, Legrand M, Sanyal K, Andes DR, Bachellier-Bassi S, d’Enfert C. Metabolic reprogramming during Candida albicans planktonic-biofilm transition is modulated by the transcription factors Zcf15 and Zcf26. PLoS Biol 2024; 22:e3002693. [PMID: 38905306 PMCID: PMC11221756 DOI: 10.1371/journal.pbio.3002693] [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: 08/04/2023] [Revised: 07/03/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024] Open
Abstract
Candida albicans is a commensal of the human microbiota that can form biofilms on implanted medical devices. These biofilms are tolerant to antifungals and to the host immune system. To identify novel genes modulating C. albicans biofilm formation, we performed a large-scale screen with 2,454 C. albicans doxycycline-dependent overexpression strains and identified 16 genes whose overexpression significantly hampered biofilm formation. Among those, overexpression of the ZCF15 and ZCF26 paralogs that encode transcription factors and have orthologs only in biofilm-forming species of the Candida clade, caused impaired biofilm formation both in vitro and in vivo. Interestingly, overexpression of ZCF15 impeded biofilm formation without any defect in hyphal growth. Transcript profiling, transcription factor binding, and phenotypic microarray analyses conducted upon overexpression of ZCF15 and ZCF26 demonstrated their role in reprogramming cellular metabolism by regulating central metabolism including glyoxylate and tricarboxylic acid cycle genes. Taken together, this study has identified a new set of biofilm regulators, including ZCF15 and ZCF26, that appear to control biofilm development through their specific role in metabolic remodeling.
Collapse
Affiliation(s)
- Laxmi Shanker Rai
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
- Department of Life Sciences, GITAM University, Bengaluru, Karnataka 561203, India
| | - Murielle Chauvel
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Hiram Sanchez
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Lasse van Wijlick
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Corinne Maufrais
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Thomas Cokelaer
- Institut Pasteur, Université Paris Cité, Hub de Bioinformatique et Biostatistique, Paris, France
| | - Natacha Sertour
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Mélanie Legrand
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Kaustuv Sanyal
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN-80, Sector-V, Salt Lake City, Kolkata, India
| | - David R. Andes
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sophie Bachellier-Bassi
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Christophe d’Enfert
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| |
Collapse
|
48
|
Rahate K, Arshi A, Barai RS, Chakraborty S, Idicula-Thomas S. EpiCandIn: An open online resource for epidemiology of Candida infections in India. Indian J Med Res 2024; 159:576-580. [PMID: 39382461 PMCID: PMC11463880 DOI: 10.25259/ijmr_886_23] [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: 05/10/2023] [Indexed: 10/10/2024] Open
Abstract
Background & objectives Candida spp. cause candidiasis in humans under conditions disrupting the host defence. While Candida albicans is the most reported cause of candidiasis, there is a surge in the incidence of infections by non-albicans Candida species (NACs), such as C. tropicalis, C. glabrata and C. auris. These species can infect all organs of the human body. To effectively manage these outbreaks, it is important to track the epidemiology of candidiasis. A consolidated resource describing the landscape of candidiasis in India is absent. Methods To address this gap, we have developed an online resource named Epidemiology of Candida Infections in India (EpiCandIn) by manually curating published literature on Candida infections in the Indian population obtained from PubMed and ScienceDirect databases. Results EpiCandIn contains data available since 1972 from 51 sites across 16 States and four Union Territories of India. It provides information on geographical location, Candida species, niche affected, disease characteristics and drug therapy details extracted from the publications. This resource is integrated with visualization tools. Interpretation & conclusions EpiCandIn will be useful for public health researchers and policymakers as it will help them gain insights into the emerging trends and management of Candida infections in India. It can be accessed at epicandin.bicnirrh.res.in.
Collapse
Affiliation(s)
- Kshitija Rahate
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, India
| | - Anam Arshi
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, India
| | - Ram Shankar Barai
- Biological Sciences Division, ICMR-National Institute of Occupational Health, Meghani Nagar, Ahmedabad, India
| | - Shuvechha Chakraborty
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, India
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, India
| |
Collapse
|
49
|
Jiang J, Keniya MV, Puri A, Zhan X, Cheng J, Wang H, Lin G, Lee YK, Jaber N, Hassoun Y, Shor E, Shi Z, Lee SH, Xu M, Perlin DS, Dai W. Structural and Biophysical Dynamics of Fungal Plasma Membrane Proteins and Implications for Echinocandin Action in Candida glabrata. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596243. [PMID: 38854035 PMCID: PMC11160696 DOI: 10.1101/2024.05.29.596243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Fungal plasma membrane proteins represent key therapeutic targets for antifungal agents, yet their structure and spatial distribution in the native context remain poorly characterized. Herein, we employ an integrative multimodal approach to elucidate the structural and functional organization of plasma membrane protein complexes in Candida glabrata , focusing on prominent and essential membrane proteins, the polysaccharide synthase β-(1,3)-glucan synthase (GS) and the proton pump Pma1. Cryo-electron tomography (cryo-ET) and live cell imaging reveal that GS and Pma1 are heterogeneously distributed into distinct plasma membrane microdomains. Treatment with caspofungin, an echinocandin antifungal that targets GS, alters the plasma membrane and disrupts the native distribution of GS and Pma1. Based on these findings, we propose a model for echinocandin action that considers how drug interactions with the plasma membrane environment lead to inhibition of GS. Our work underscores the importance of interrogating the structural and dynamic characteristics of fungal plasma membrane proteins in situ to understand function and facilitate precisely targeted development of novel antifungal therapies.
Collapse
|
50
|
Mathur K, Singh B, Puria R, Nain V. In silico genome wide identification of long non-coding RNAs differentially expressed during Candida auris host pathogenesis. Arch Microbiol 2024; 206:253. [PMID: 38727738 DOI: 10.1007/s00203-024-03969-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/18/2024] [Indexed: 05/15/2024]
Abstract
Candida auris is an invasive fungal pathogen of high concern due to acquired drug tolerance against antifungals used in clinics. The prolonged persistence on biotic and abiotic surfaces can result in onset of hospital outbreaks causing serious health threat. An in depth understanding of pathology of C. auris is highly desirable for development of efficient therapeutics. Non-coding RNAs play crucial role in fungal pathology. However, the information about ncRNAs is scanty to be utilized. Herein our aim is to identify long noncoding RNAs with potent role in pathobiology of C. auris. Thereby, we analyzed the transcriptomics data of C. auris infection in blood for identification of potential lncRNAs with regulatory role in determining invasion, survival or drug tolerance under infection conditions. Interestingly, we found 275 lncRNAs, out of which 253 matched with lncRNAs reported in Candidamine, corroborating for our accurate data analysis pipeline. Nevertheless, we obtained 23 novel lncRNAs not reported earlier. Three lncRNAs were found to be under expressed throughout the course of infection, in the transcriptomics data. 16 of potent lncRNAs were found to be coexpressed with coding genes, emphasizing for their functional role. Noteworthy, these ncRNAs are expressed from intergenic regions of the genes associated with transporters, metabolism, cell wall biogenesis. This study recommends for possible association between lncRNA expression and C. auris pathogenesis.
Collapse
Affiliation(s)
- Kartavya Mathur
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Bharti Singh
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Rekha Puria
- School of Biotechnology, Gautam Buddha University, Greater Noida, India.
| | - Vikrant Nain
- School of Biotechnology, Gautam Buddha University, Greater Noida, India.
| |
Collapse
|