1
|
Kowalewski ME, Zagler S, Redinbo MR. Structural Insights into Selectively Targeting Candida albicans Hsp90. Biochemistry 2025. [PMID: 40397669 DOI: 10.1021/acs.biochem.5c00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
The threat of drug-resistant pathogens continues to rise and underscores the need for new antimicrobial and antifungal strategies. Diverse chemical scaffolds have been shown with high affinity to bind the human heat-shock protein Hsp90. Orthologous proteins are present in microbial pathogens and have been shown to be particularly abundant in these organisms, suggesting they may serve as therapeutic targets. Here, we examine the potency and selectivity of human Hsp90 ligands for their capacity to bind to the nucleotide binding domain of Hsp90 from the pathogenic fungi, Candida albicans. Using a series of biochemical, structural, and fragment and in silico screening investigations, we define key chemical features that lead to effective C. albicans Hsp90 (CaHsp90) binding. We support these studies with crystal structures of five diverse human Hsp90 ligands in complex with CaHsp90, as well as the structure of this protein with a nonhydrolyzable ATP analog. We demonstrate the structural basis for the selectivity of the human Hsp90 inhibitor TAS116 for CaHsp90, features that may be exploited in the future development of improved CaHsp90 inhibitors.
Collapse
Affiliation(s)
- Mark E Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Sebastian Zagler
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Matthew R Redinbo
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Microbiology and Immunology, and Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
2
|
Ghogare SS, Pathan EK. Intratumor fungi specific mechanisms to influence cell death pathways and trigger tumor cell apoptosis. Cell Death Discov 2025; 11:188. [PMID: 40258837 PMCID: PMC12012188 DOI: 10.1038/s41420-025-02483-z] [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: 12/26/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer, uncontrolled cell growth due to the loss of cell cycle regulation, is often found to be associated with viral infections and, as recent studies show, with bacterial infections as well. Emerging reports also suggest a strong link between fungi and cancer. The crucial virulence trait of fungi, the switch from yeast (Y) to hyphal (H) form, is found to be associated with carcinogenesis. The physicochemical properties and signal transduction pathways involved in the switch to the hyphal form overlap with those of tumor cell formation. Inhibiting differentiation causes apoptosis in fungi, whereas preventing apoptosis leads to cancer in multicellular organisms. Literature on the fungi-cancer linkage, though limited, is increasing rapidly. This review examines cancer-specific fungal communities, the impact of fungal microbiome on cancer cell progression, similarities between fungal differentiation and cells turning cancerous at biochemical and molecular levels, including the overlaps in signal transduction pathways between fungi and cancer. Based on the available evidence, we suggest that molecules inhibiting the yeast-hyphal transition in fungi can be combined with those targeting tumor cell apoptosis for effective cancer treatment. The review points out fertile research areas where mycologists and cancer researchers can collaborate to unravel common molecular mechanisms. Moreover, antibodies targeting fungal-specific chitin and glucan can be used for the selective neutralization of tumor cells. These new combinations of potential therapies are expected to facilitate the development of target-specific, less harmful and commercially feasible anticancer therapies. We bring together available evidence to argue that fungal infections could either trigger cancer or have a significant role in the development and progression of cancer. Hence, cancer-associated fungal populations could be utilized as a target for a combination therapy involving the integration of anticancer and antifungal drugs as well as inhibitors of fungal morphogenesis to develop more effective anticancer therapies.
Collapse
Affiliation(s)
- Simran S Ghogare
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India
| | - Ejaj K Pathan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
3
|
Fang T, Lu H, Jiang Y. Extracellular fungal Hsp90 represents a promising therapeutic target for combating fungal infections. Eur J Pharm Sci 2025; 207:107041. [PMID: 39947525 DOI: 10.1016/j.ejps.2025.107041] [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: 09/14/2024] [Revised: 12/17/2024] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
Heat shock protein 90 (Hsp90) is a pivotal virulence factor in pathogenic fungi, playing a significant role in conferring drug resistance. However, due to the high amino acid sequence similarity between fungal and mammalian Hsp90, targeting fungal intracellular Hsp90 therapeutically is associated with marked toxic side effects, thereby limiting clinical application. Studies have demonstrated that intracellular fungal Hsp90 can be secreted as extracellular Hsp90 (eHsp90), which plays a crucial role in fungal infections. Strategies targeting fungal eHsp90 have exhibited promising therapeutic outcomes. Unlike intracellular targeting, such antifungal approaches can operate without cell penetration, thereby circumventing the toxic side effects due to Hsp90's high conservation. This review summarizes the potential extracellular secretion pathways of fungal eHsp90, its roles in fungal pathogenesis, as well as the development of vaccines and antibodies targeting fungal eHsp90. The review underlines the significance of eHsp90 in fungal infections and suggests that eHsp90 represents a promising therapeutic target for fungal infection treatment.
Collapse
Affiliation(s)
- Ting Fang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
4
|
Bende G, Zsindely N, Laczi K, Kristóffy Z, Papp C, Farkas A, Tóth L, Sáringer S, Bodai L, Rákhely G, Marx F, Galgóczy L. The Neosartorya (Aspergillus) fischeri antifungal protein NFAP2 has low potential to trigger resistance development in Candida albicans in vitro. Microbiol Spectr 2025; 13:e0127324. [PMID: 39560388 PMCID: PMC11705825 DOI: 10.1128/spectrum.01273-24] [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: 05/23/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Due to the increase in the number of drug-resistant Candida albicans strains, new antifungal compounds with limited potential for the development of resistance are urgently needed. NFAP2, an antifungal protein (AFP) secreted by Neosartorya (Aspergillus) fischeri, is a promising candidate. We investigated the ability of C. albicans to develop resistance to NFAP2 in a microevolution experiment compared with generic fluconazole (FLC). C. albicans adapted to only 1× minimum inhibitory concentration (MIC) of NFAP2, which can be considered tolerance rather than resistance, compared with 32× MIC of FLC. Genome analysis revealed non-silent mutations in only two genes in NFAP2-tolerant strains and in several genes in FLC-resistant strains. Tolerance development to NFAP2 did not influence cell morphology. The susceptibility of NFAP2-tolerant strains did not change to FLC, amphotericin B, micafungin, and terbinafine. These strains did not show altered susceptibility to AFPs from Penicillium chrysogenum, except one which had less susceptibility to Penicillium chrysogenum antifungal protein B. FLC-resistant strains had decreased susceptibility to terbinafine and NFAP2, but not to other drugs and AFPs from P. chrysogenum. NFAP2-tolerant and FLC-resistant strains showed decreased and increased NFAP2 binding and uptake, respectively. The development of tolerance to NFAP2 decreased tolerance to cell wall, heat, and UV stresses. The development of FLC resistance increased tolerance to cell wall stress and decreased tolerance to heat and UV stresses. Tolerance to NFAP2 did not have significant metabolic fitness cost and could not increase virulence, compared with resistance to FLC.IMPORTANCEDue to the increasing number of (multi)drug-resistant strains, only a few effective antifungal drugs are available to treat infections caused by opportunistic Candida species. Therefore, the incidence of hard-to-treat candidiasis has increased dramatically in the past decade, and the demand to identify antifungal compounds with minimal potential to trigger resistance is substantial. The features of NFAP2 make it a promising candidate for the topical treatment of Candida infection. Data on the development of resistance to antifungal proteins in Candida albicans are lacking. In this study, we provide evidence that NFAP2 has a low potential to trigger resistance in C. albicans in vitro, and the developed tolerance to NFAP2 is not associated with severe phenotypic changes compared with development of resistance to generic fluconazole. These results suggest the slow emergence of NFAP2-resistant Candida strains, and NFAP2 can reliably be used long-term in the clinic.
Collapse
Affiliation(s)
- Gábor Bende
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Nóra Zsindely
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Krisztián Laczi
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Plant Biology, HUN-REN Biological Research Center, Szeged, Hungary
| | - Zsolt Kristóffy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Csaba Papp
- Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Attila Farkas
- Institute of Plant Biology, HUN-REN Biological Research Center, Szeged, Hungary
| | - Liliána Tóth
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Szabolcs Sáringer
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gábor Rákhely
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biophysics, HUN-REN Biological Research Center, Szeged, Hungary
| | - Florentine Marx
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, HUN-REN Biological Research Center, Szeged, Hungary
| |
Collapse
|
5
|
Aguilar-Rodríguez J, Jakobson CM, Jarosz DF. The Hsp90 Molecular Chaperone as a Global Modifier of the Genotype-Phenotype-Fitness Map: An Evolutionary Perspective. J Mol Biol 2024; 436:168846. [PMID: 39481633 PMCID: PMC11608137 DOI: 10.1016/j.jmb.2024.168846] [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: 08/24/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024]
Abstract
Global modifier genes influence the mapping of genotypes onto phenotypes and fitness through their epistatic interactions with genetic variants on a massive scale. The first such factor to be identified, Hsp90, is a highly conserved molecular chaperone that plays a central role in protein homeostasis. Hsp90 is a "hub of hubs" that chaperones proteins engaged in many key cellular and developmental regulatory networks. These clients, which are enriched in kinases, transcription factors, and E3 ubiquitin ligases, drive diverse cellular functions and are themselves highly connected. By contrast to many other hub proteins, the abundance and activity of Hsp90 changes substantially in response to shifting environmental conditions. As a result, Hsp90 modifies the functional impact of many genetic variants simultaneously in a manner that depends on environmental stress. Studies in diverse organisms suggest that this coupling between Hsp90 function and challenging environments exerts a substantial impact on what parts of the genome are visible to natural selection, expanding adaptive opportunities when most needed. In this Perspective, we explore the multifaceted role of Hsp90 as global modifier of the genotype-phenotype-fitness map as well as its implications for evolution in nature and the clinic.
Collapse
Affiliation(s)
- José Aguilar-Rodríguez
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biology, Stanford University, Stanford, CA, USA
| | - Christopher M Jakobson
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Zheng L, Xu Y, Wang C, Dong Y, Guo L. Parallel evolution of fluconazole resistance and tolerance in Candida glabrata. Front Cell Infect Microbiol 2024; 14:1456907. [PMID: 39397866 PMCID: PMC11466938 DOI: 10.3389/fcimb.2024.1456907] [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: 06/29/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction With the growing population of immunocompromised individuals, opportunistic fungal pathogens pose a global health threat. Candida species, particularly C. albicans and non-albicans Candida species such as C. glabrata, are the most prevalent pathogenic fungi. Azoles, especially fluconazole, are widely used therapeutic options. Objective This study investigates how C. glabrata adapts to fluconazole, with a focus on understanding the factors regulating fluconazole tolerance and its relationship to resistance. Methods This study compared the factors regulating fluconazole tolerance between C. albicans and C. glabrata. We analyzed the impact of temperature on fluconazole tolerance, and requirement of calcineurin and Hsp90 for maintenance of fluconazole tolerance. We isolated colonies from edge, inside and outside of inhibition zone in disk diffusion assays. And we exposed C. glabrata strain to high concentrations of fluconazole and investigated the mutants for development of fluconazole resistance and tolerance. Results We found temperature modulated tolerance in the opposite way in C. albicans strain YJB-T1891 and C. glabrata strain CG4. Calcineurin and Hsp90 were required for maintenance of fluconazole tolerance in both species. Colonies from inside and outside of inhibition zones did not exhibited mutated phenotype, but colonies isolated from edge of inhibition zone exhibited diverse phenotype changes. Moreover, we discovered that high concentrations (16-128 μg/mL) of fluconazole induce the simultaneous but parallel development of tolerance and resistance in C. glabrata, unlike the sole development of tolerance in C. albicans. Conclusion This study highlights that while tolerance to fluconazole is a common response in Candida species, the specific molecular mechanisms and evolutionary pathways that lead to this response vary between species. Our findings emphasize the importance of understanding the regulation of fluconazole tolerance in different Candida species to develop effective therapeutic strategies.
Collapse
Affiliation(s)
- Lijun Zheng
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Xu
- Department of Pharmacy, The 960th Hospital of PLA, Jinan, China
| | - Chen Wang
- Department of Pharmacy, The 960th Hospital of PLA, Jinan, China
| | - Yubo Dong
- Department of Pharmacy, The 960th Hospital of PLA, Jinan, China
| | - Liangsheng Guo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Ball B, Sukumaran A, Pladwig S, Kazi S, Chan N, Honeywell E, Modrakova M, Geddes-McAlister J. Proteome signatures reveal homeostatic and adaptive oxidative responses by a putative co-chaperone, Wos2, to influence fungal virulence determinants in cryptococcosis. Microbiol Spectr 2024; 12:e0015224. [PMID: 38953322 PMCID: PMC11302251 DOI: 10.1128/spectrum.00152-24] [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: 01/16/2024] [Accepted: 05/27/2024] [Indexed: 07/04/2024] Open
Abstract
The increasing prevalence of invasive fungal pathogens is dramatically changing the clinical landscape of infectious diseases, posing an imminent threat to public health. Specifically, Cryptococcus neoformans, the human opportunistic pathogen, expresses elaborate virulence mechanisms and is equipped with sophisticated adaptation strategies to survive in harsh host environments. This study extensively characterizes Wos2, an Hsp90 co-chaperone homolog, featuring bilateral functioning for both cryptococcal adaptation and the resulting virulence response. In this study, we evaluated the proteome and secretome signatures associated with wos2 deletion in enriched and infection-mimicking conditions to reveal Wos2-dependent regulation of the oxidative stress response through global translational reprogramming. The wos2Δ strain demonstrates defective intracellular and extracellular antioxidant protection systems, measurable through a decreased abundance of critical antioxidant enzymes and reduced growth in the presence of peroxide stress. Additional Wos2-associated stress phenotypes were observed upon fungal challenge with heat shock, osmotic stress, and cell membrane stressors. We demonstrate the importance of Wos2 for intracellular lifestyle of C. neoformans during in vitro macrophage infection and provide evidence for reduced phagosomal replication levels associated with wos2Δ. Accordingly, wos2Δ featured significantly reduced virulence within impacting fungal burden in a murine model of cryptococcosis. Our study highlights a vulnerable point in the fungal chaperone network that offers a therapeutic opportunity to interfere with both fungal virulence and fitness.IMPORTANCEThe global impact of fungal pathogens, both emerging and emerged, is undeniable, and the alarming increase in antifungal resistance rates hampers our ability to protect the global population from deadly infections. For cryptococcal infections, a limited arsenal of antifungals and increasing rates of resistance demand alternative therapeutic strategies, including an anti-virulence approach, which disarms the pathogen of critical virulence factors, empowering the host to remove the pathogens and clear the infection. To this end, we apply state-of-the-art mass spectrometry-based proteomics to evaluate the impact of a recently defined novel co-chaperone, Wos2, toward cryptococcal virulence using in vitro and in vivo models of infection. We explore global proteome and secretome remodeling driven by the protein and uncover the novel role in modulating the fungal oxidative stress response. Complementation of proteome findings with in vitro infectivity assays demonstrated the protective role of Wos2 within the macrophage phagosome, influencing fungal replication and survival. These results underscore differential cryptococcal survivability and weakened patterns of dissemination in the absence of wos2. Overall, our study establishes Wos2 as an important contributor to fungal pathogenesis and warrants further research into critical proteins within global stress response networks as potential druggable targets to reduce fungal virulence and clear infection.
Collapse
Affiliation(s)
- Brianna Ball
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Arjun Sukumaran
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Samanta Pladwig
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Samiha Kazi
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Norris Chan
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Effie Honeywell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Manuela Modrakova
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
8
|
Yiu B, Robbins N, Cowen LE. Interdisciplinary approaches for the discovery of novel antifungals. Trends Mol Med 2024; 30:723-735. [PMID: 38777733 PMCID: PMC11987087 DOI: 10.1016/j.molmed.2024.04.018] [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: 02/16/2024] [Revised: 04/10/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Pathogenic fungi are an increasing public health concern. The emergence of antifungal resistance coupled with the scarce antifungal arsenal highlights the need for novel therapeutics. Fortunately, the past few years have witnessed breakthroughs in antifungal development. Here, we discuss pivotal interdisciplinary approaches for the discovery of novel compounds with efficacy against diverse fungal pathogens. We highlight breakthroughs in improving current antifungal scaffolds, as well as the utility of compound combinations to extend the lifespan of antifungals. Finally, we describe efforts to refine candidate chemical scaffolds by leveraging structure-guided approaches, and the use of functional genomics to expand our knowledge of druggable antifungal targets. Overall, we emphasize the importance of interdisciplinary collaborations in the endeavor to develop innovative antifungal strategies.
Collapse
Affiliation(s)
- Bonnie Yiu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada.
| |
Collapse
|
9
|
da Silva CM, de Lima Neto RG, de Carvalho AMR, Macêdo DPC, de Azevedo Melo AS, Neves RP. Taxonomy of Candida parapsilosis complex isolated from neonates and the role of Hsp90 inhibitors to enhanced the antifungal activity of micafungin. Lett Appl Microbiol 2024; 77:ovae044. [PMID: 38658187 DOI: 10.1093/lambio/ovae044] [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: 08/03/2023] [Revised: 03/25/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
Species from Candida parapsilosis complex are frequently found in neonatal candidemia. The antifungal agents to treat this infection are limited and the occurrence of low in vitro susceptibility to echinocandins such as micafungin has been observed. In this context, the chaperone Hsp90 could be a target to reduce resistance. Thus, the objective of this research was to identify isolates from the C. parapsilosis complex and verify the action of Hsp90 inhibitors associated with micafungin. The fungal identification was based on genetic sequencing and mass spectrometry. Minimal inhibitory concentrations were determined by broth microdilution method according to Clinical Laboratory and Standards Institute. The evaluation of the interaction between micafungin with Hsp90 inhibitors was realized using the checkerboard methodology. According to the polyphasic taxonomy, C. parapsilosis sensu stricto was the most frequently identified, followed by C. orthopsilosis and C. metapsilosis, and one isolate of Lodderomyces elongisporus was identified by genetic sequencing. The Hsp90 inhibitor geladanamycin associated with micafungin showed a synergic effect in 31.25% of the isolates, a better result was observed with radicicol, which shows synergic effect in 56.25% tested yeasts. The results obtained demonstrate that blocking Hsp90 could be effective to reduce antifungal resistance to echinocandins.
Collapse
Affiliation(s)
| | | | | | | | | | - Rejane Pereira Neves
- Federal University of Pernambuco, Mycology Department, Recife-PE, 50670-90, Brazil
| |
Collapse
|
10
|
Alabi PE, Gautier C, Murphy TP, Gu X, Lepas M, Aimanianda V, Sello JK, Ene IV. Small molecules restore azole activity against drug-tolerant and drug-resistant Candida isolates. mBio 2023; 14:e0047923. [PMID: 37326546 PMCID: PMC10470600 DOI: 10.1128/mbio.00479-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/13/2023] [Indexed: 06/17/2023] Open
Abstract
Each year, fungi cause more than 1.5 billion infections worldwide and have a devastating impact on human health, particularly in immunocompromised individuals or patients in intensive care units. The limited antifungal arsenal and emerging multidrug-resistant species necessitate the development of new therapies. One strategy for combating drug-resistant pathogens is the administration of molecules that restore fungal susceptibility to approved drugs. Accordingly, we carried out a screen to identify small molecules that could restore the susceptibility of pathogenic Candida species to azole antifungals. This screening effort led to the discovery of novel 1,4-benzodiazepines that restore fluconazole susceptibility in resistant isolates of Candida albicans, as evidenced by 100-1,000-fold potentiation of fluconazole activity. This potentiation effect was also observed in azole-tolerant strains of C. albicans and in other pathogenic Candida species. The 1,4-benzodiazepines selectively potentiated different azoles, but not other approved antifungals. A remarkable feature of the potentiation was that the combination of the compounds with fluconazole was fungicidal, whereas fluconazole alone is fungistatic. Interestingly, the potentiators were not toxic to C. albicans in the absence of fluconazole, but inhibited virulence-associated filamentation of the fungus. We found that the combination of the potentiators and fluconazole significantly enhanced host survival in a Galleria mellonella model of systemic fungal infection. Taken together, these observations validate a strategy wherein small molecules can restore the activity of highly used anti-infectives that have lost potency. IMPORTANCE In the last decade, we have been witnessing a higher incidence of fungal infections, due to an expansion of the fungal species capable of causing disease (e.g., Candida auris), as well as increased antifungal drug resistance. Among human fungal pathogens, Candida species are a leading cause of invasive infections and are associated with high mortality rates. Infections by these pathogens are commonly treated with azole antifungals, yet the expansion of drug-resistant isolates has reduced their clinical utility. In this work, we describe the discovery and characterization of small molecules that potentiate fluconazole and restore the susceptibility of azole-resistant and azole-tolerant Candida isolates. Interestingly, the potentiating 1,4-benzodiazepines were not toxic to fungal cells but inhibited their virulence-associated filamentous growth. Furthermore, combinations of the potentiators and fluconazole decreased fungal burdens and enhanced host survival in a Galleria mellonella model of systemic fungal infections. Accordingly, we propose the use of novel antifungal potentiators as a powerful strategy for addressing the growing resistance of fungi to clinically approved drugs.
Collapse
Affiliation(s)
- Philip E. Alabi
- Department of Chemistry, Brown University, Providence, Rhode Island, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Cécile Gautier
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
| | - Thomas P. Murphy
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Xilin Gu
- Department of Chemistry, Brown University, Providence, Rhode Island, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Mathieu Lepas
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Molecular Mycology Unit, Paris, France
| | - Vishukumar Aimanianda
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Molecular Mycology Unit, Paris, France
| | - Jason K. Sello
- Department of Chemistry, Brown University, Providence, Rhode Island, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Iuliana V. Ene
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
11
|
Metzner K, O’Meara MJ, Halligan B, Wotring JW, Sexton JZ, O’Meara TR. Imaging-Based Screening Identifies Modulators of the eIF3 Translation Initiation Factor Complex in Candida albicans. Antimicrob Agents Chemother 2023; 67:e0050323. [PMID: 37382550 PMCID: PMC10353439 DOI: 10.1128/aac.00503-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/07/2023] [Indexed: 06/30/2023] Open
Abstract
Fungal pathogens like Candida albicans can cause devastating human disease. Treatment of candidemia is complicated by the high rate of resistance to common antifungal therapies. Additionally, there is host toxicity associated with many antifungal compounds due to the conservation between essential mammalian and fungal proteins. An attractive new approach for antimicrobial development is to target virulence factors: non-essential processes that are required for the organism to cause disease in human hosts. This approach expands the potential target space while reducing the selective pressure toward resistance, as these targets are not essential for viability. In C. albicans, a key virulence factor is the ability to transition to hyphal morphology. We developed a high-throughput image analysis pipeline to distinguish between yeast and filamentous growth in C. albicans at the single cell level. Based on this phenotypic assay, we screened the FDA drug repurposing library of 2,017 compounds for their ability to inhibit filamentation and identified 33 compounds that block the hyphal transition in C. albicans with IC50 values ranging from 0.2 to 150 μM. Multiple compounds showed a phenyl sulfone chemotype, prompting further analysis. Of these phenyl sulfones, NSC 697923 displayed the most efficacy, and by selecting for resistant mutants, we identified eIF3 as the target of NSC 697923 in C. albicans.
Collapse
Affiliation(s)
- Katura Metzner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin Halligan
- University of Michigan Center for Drug Repurposing, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jesse W. Wotring
- University of Michigan Center for Drug Repurposing, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, Michigan, USA
| | - Jonathan Z. Sexton
- University of Michigan Center for Drug Repurposing, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, Michigan, USA
| | - Teresa R. O’Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Metzner K, O’Meara MJ, Halligan B, Wotring JW, Sexton JZ, O’Meara TR. Imaging-based screening identifies modulators of the eIF3 translation initiation factor complex in Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537517. [PMID: 37131825 PMCID: PMC10153179 DOI: 10.1101/2023.04.19.537517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Fungal pathogens like Candida albicans can cause devastating human disease. Treatment of candidemia is complicated by the high rate of resistance to common antifungal therapies. Additionally, there is host toxicity associated with many antifungal compounds due to the conservation between essential mammalian and fungal proteins. An attractive new approach for antimicrobial development is to target virulence factors: non-essential processes that are required for the organism to cause disease in human hosts. This approach expands the potential target space while reducing the selective pressure towards resistance, as these targets are not essential for viability. In C. albicans, a key virulence factor is the ability to transition to hyphal morphology. We developed a high-throughput image analysis pipeline to distinguish between yeast and filamentous growth in C. albicans at the single cell level. Based on this phenotypic assay, we screened the FDA drug repurposing library of 2,017 compounds for their ability to inhibit filamentation and identified 33 compounds that block the hyphal transition in C. albicans with IC 50 values ranging from 0.2 to 150 µM. Multiple compounds showed a phenyl vinyl sulfone chemotype, prompting further analysis. Of these phenyl vinyl sulfones, NSC 697923 displayed the most efficacy, and by selecting for resistant mutants, we identified eIF3 as the target of NSC 697923 in C. albicans .
Collapse
Affiliation(s)
- Katura Metzner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew J O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin Halligan
- University of Michigan Center for Drug Repurposing, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jesse W. Wotring
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, MI, USA
- University of Michigan Center for Drug Repurposing, USA
| | - Jonathan Z Sexton
- Department of Medicinal Chemistry, College of Pharmacy, Ann Arbor, MI, USA
- University of Michigan Center for Drug Repurposing, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Teresa R O’Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Liu N, Tu J, Huang Y, Yang W, Wang Q, Li Z, Sheng C. Target- and prodrug-based design for fungal diseases and cancer-associated fungal infections. Adv Drug Deliv Rev 2023; 197:114819. [PMID: 37024014 DOI: 10.1016/j.addr.2023.114819] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
Invasive fungal infections (IFIs) are emerging as a serious threat to public health and are associated with high incidence and mortality. IFIs also represent a frequent complication in patients with cancer who are undergoing chemotherapy. However, effective and safe antifungal agents remain limited, and the development of severe drug resistance further undermines the efficacy of antifungal therapy. Therefore, there is an urgent need for novel antifungal agents to treat life-threatening fungal diseases, especially those with new mode of action, favorable pharmacokinetic profiles, and anti-resistance activity. In this review, we summarize new antifungal targets and target-based inhibitor design, with a focus on their antifungal activity, selectivity, and mechanism. We also illustrate the prodrug design strategy used to improve the physicochemical and pharmacokinetic profiles of antifungal agents. Dual-targeting antifungal agents offer a new strategy for the treatment of resistant infections and cancer-associated fungal infections.
Collapse
|
14
|
Liu NN, Zhou J, Jiang T, Tarsio M, Yu F, Zheng X, Qi W, Liu L, Tan JC, Wei L, Ding J, Li J, Zeng L, Ren B, Huang X, Peng Y, Cao YB, Zhao Y, Zhang XY, Kane PM, Chen C, Wang H. A dual action small molecule enhances azoles and overcomes resistance through co-targeting Pdr5 and Vma1. Transl Res 2022; 247:39-57. [PMID: 35452875 DOI: 10.1016/j.trsl.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 02/26/2022] [Accepted: 04/12/2022] [Indexed: 11/19/2022]
Abstract
Fungal infection threatens human health worldwide due to the limited arsenal of antifungals and the rapid emergence of resistance. Epidermal growth factor receptor (EGFR) is demonstrated to mediate epithelial cell endocytosis of the leading human fungal pathogen, Candida albicans. However, whether EGFR inhibitors act on fungal cells remains unknown. Here, we discovered that the specific EGFR inhibitor osimertinib mesylate (OSI) potentiates azole efficacy against diverse fungal pathogens and overcomes azole resistance. Mechanistic investigation revealed a conserved activity of OSI by promoting intracellular fluconazole accumulation via inhibiting Pdr5 and disrupting V-ATPase function via targeting Vma1 at serine 274, eventually leading to inactivation of the global regulator TOR. Evaluation of the in vivo efficacy and toxicity of OSI demonstrated its potential clinical application in impeding fluconazole resistance. Thus, the identification of OSI as a dual action antifungal with co-targeting activity proposes a potentially effective therapeutic strategy to treat life-threatening fungal infection and overcome antifungal resistance.
Collapse
Affiliation(s)
- Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jia Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Jiang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Maureen Tarsio
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Feifei Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China
| | - Xuehan Zheng
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wanjun Qi
- Division of Infectious Diseases, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Lin Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Cong Tan
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luqi Wei
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ding
- Computational biology department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jingquan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingbing Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Yibing Peng
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Faculty of Medical Laboratory Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Bing Cao
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai TCM-Integrated Institute of Vascular Disease, Shanghai, China
| | - Yanbin Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, China
| | - Xin-Yu Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Changbin Chen
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Cui X, Wang L, Lü Y, Yue C. Development and research progress of anti-drug resistant fungal drugs. J Infect Public Health 2022; 15:986-1000. [PMID: 35981408 DOI: 10.1016/j.jiph.2022.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
With the widespread use of immunosuppressive agents and the increase in patients with severe infections, the incidence of fungal infections worldwide has increased year by year. The fungal pathogens Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus cause a total of more than 1 million deaths each year. Long-term use of antifungal drugs can easily lead to fungal resistance, and the prevalence of drug-resistant fungi is a major global health challenge. In order to effectively control global fungal infections, there is an urgent need for new drugs that can exert effective antifungal activity and overcome drug resistance. We must promote the discovery of new antifungal targets and drugs, and find effective ways to control drug-resistant fungi through different ways, so as to reduce the threat of drug-resistant fungi to human life, health and safety. In the past few years, certain progress has been made in the research and development of antifungal drugs. In addition to summarizing some of the antifungal drugs currently approved by the FDA, this review also focuses on potential antifungal drugs, the repositioned drugs, and drugs that can treat drug-resistant bacteria and fungal infections, and provide new ideas for the development of antifungal drugs in the future.
Collapse
Affiliation(s)
- Xiangyi Cui
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Lanlin Wang
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Yuhong Lü
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Changwu Yue
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| |
Collapse
|
16
|
Liu L, Sun Y, Gao Z, Yin W, Jiang H, Wu T, Sun Y, Qin Q, Zhao D, Cheng M. Design, synthesis, and evaluation of novel 3,4-isoxazolediamide derivatives for the combination treatment of azole-resistant candidiasis. Arch Pharm (Weinheim) 2022; 355:e2200266. [PMID: 36029272 DOI: 10.1002/ardp.202200266] [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: 05/20/2022] [Revised: 07/01/2022] [Accepted: 08/02/2022] [Indexed: 11/12/2022]
Abstract
Invasive fungal infections are emerging as serious infectious diseases worldwide. Due to the frequent emergence of resistance, the cure for invasive fungal infections is often unachievable. The molecular chaperone Hsp90 provides a promising target because it supports survival, virulence, and drug resistance in a variety of pathogens. Herein, we report on the structural optimization and structure-activity relationship studies of 3,4-isoxazolediamide analogs. As a new class of fungal Hsp90 inhibitor, compound B25 was found to have good synergistic effects with fluconazole and to avoid potential mammalian toxicity. It also showed remarkable metabolic stability in vitro. Collectively, B25 could be a promising lead compound for drug discovery targeting fungal Hsp90 and deserves further investigation.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Yixiang Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Zixuan Gao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenbo Yin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Hong Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianxiao Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Yin Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Qiaohua Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Dongmei Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
17
|
Iyer KR, Robbins N, Cowen LE. The role of Candida albicans stress response pathways in antifungal tolerance and resistance. iScience 2022; 25:103953. [PMID: 35281744 PMCID: PMC8905312 DOI: 10.1016/j.isci.2022.103953] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human fungal pathogens are the causative agents of devastating diseases across the globe, and the increasing prevalence of drug resistance threatens to undermine the already limited treatment options. One prominent pathogen is the opportunistic fungus Candida albicans, which can cause both superficial and serious systemic infections in immunocompromised individuals. C. albicans antifungal drug resistance and antifungal tolerance are supported by diverse and expansive cellular stress response pathways. Some of the major players are the Ca2+-calmodulin-activated phosphatase calcineurin, the protein kinase C cell wall integrity pathway, and the molecular chaperone heat shock protein 90. Beyond these core signal transducers, several other enzymes and transcription factors have been implicated in both tolerance and resistance. Here, we highlight some of the major stress response pathways, key advances in identifying chemical matter to inhibit these pathways, and implications for C. albicans persistence in the host. Candida albicans can cause superficial and serious systemic infections in humans Stress response pathways regulate C. albicans antifungal resistance and tolerance Stress response regulators include calcineurin, Pkc1, Hsp90, and many others Stress response inhibitors could reduce the likelihood of fungi persisting in humans
Collapse
Affiliation(s)
- Kali R. Iyer
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, West Tower, Room 1638, Toronto, ON M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, West Tower, Room 1638, Toronto, ON M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, West Tower, Room 1638, Toronto, ON M5G 1M1, Canada
- Corresponding author
| |
Collapse
|
18
|
Staniszewska M, Zdrojewski T, Gizińska M, Rogalska M, Kuryk Ł, Kowalkowska A, Łukowska-Chojnacka E. Tetrazole derivatives bearing benzodiazepine moiety—synthesis and action mode against virulence of Candida albicans. Eur J Med Chem 2022; 230:114060. [DOI: 10.1016/j.ejmech.2021.114060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
|
19
|
Li C, Tu J, Han G, Liu N, Sheng C. Heat shock protein 90 (Hsp90)/Histone deacetylase (HDAC) dual inhibitors for the treatment of azoles-resistant Candida albicans. Eur J Med Chem 2022; 227:113961. [PMID: 34742014 DOI: 10.1016/j.ejmech.2021.113961] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/22/2022]
Abstract
Clinical treatment of candidiasis has suffered from increasingly severe drug resistance and limited efficacy. Thus, novel strategies to deal with drug resistance are highly desired to develop effective therapeutic agents. Herein, dual inhibition of heat shock protein 90 (Hsp90) and histone deacetylase (HDAC) was validated as a new strategy to potentiate efficacy of fluconazole against resistant Candida albicans infections. The first generation of Hsp90/HDAC dual inhibitors were designed as synergistic enhancers to treat azoles-resistant candidiasis. In particular, compound J5 exhibited fungal-selective inhibitory effects on Hsp90 and HDACs, leading to low toxicity and excellent in vitro (FICI = 0.266) and in vivo synergistic antifungal potency to treat fluconazole resistant candidiasis. Antifungal-mechanistic investigation revealed that compound J5 suppressed important virulence factors and down-regulated expression of resistance-associated genes. Therefore, Hsp90/HDAC dual inhibitors represent a new strategy for the development of novel antifungal therapeutics to combat azole-resistant candidiasis.
Collapse
Affiliation(s)
- Chaochen Li
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jie Tu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Guiyan Han
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Na Liu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
20
|
Bouz G, Doležal M. Advances in Antifungal Drug Development: An Up-To-Date Mini Review. Pharmaceuticals (Basel) 2021; 14:1312. [PMID: 34959712 PMCID: PMC8706862 DOI: 10.3390/ph14121312] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
The utility of clinically available antifungals is limited by their narrow spectrum of activity, high toxicity, and emerging resistance. Antifungal drug discovery has always been a challenging area, since fungi and their human host are eukaryotes, making it difficult to identify unique targets for antifungals. Novel antifungals in clinical development include first-in-class agents, new structures for an established target, and formulation modifications to marketed antifungals, in addition to repurposed agents. Membrane interacting peptides and aromatherapy are gaining increased attention in the field. Immunotherapy is another promising treatment option, with antifungal antibodies advancing into clinical trials. Novel targets for antifungal therapy are also being discovered, allowing the design of new promising agents that may overcome the resistance issue. In this mini review, we will summarize the current status of antifungal drug pipelines in clinical stages, and the most recent advancements in preclinical antifungal drug development, with special focus on their chemistry.
Collapse
Affiliation(s)
- Ghada Bouz
- Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic
| | - Martin Doležal
- Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic
| |
Collapse
|
21
|
Uthayakumar D, Sharma J, Wensing L, Shapiro RS. CRISPR-Based Genetic Manipulation of Candida Species: Historical Perspectives and Current Approaches. Front Genome Ed 2021; 2:606281. [PMID: 34713231 PMCID: PMC8525362 DOI: 10.3389/fgeed.2020.606281] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022] Open
Abstract
The Candida genus encompasses a diverse group of ascomycete fungi that have captured the attention of the scientific community, due to both their role in pathogenesis and emerging applications in biotechnology; the development of gene editing tools such as CRISPR, to analyze fungal genetics and perform functional genomic studies in these organisms, is essential to fully understand and exploit this genus, to further advance antifungal drug discovery and industrial value. However, genetic manipulation of Candida species has been met with several distinctive barriers to progress, such as unconventional codon usage in some species, as well as the absence of a complete sexual cycle in its diploid members. Despite these challenges, the last few decades have witnessed an expansion of the Candida genetic toolbox, allowing for diverse genome editing applications that range from introducing a single point mutation to generating large-scale mutant libraries for functional genomic studies. Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 technology is among the most recent of these advancements, bringing unparalleled versatility and precision to genetic manipulation of Candida species. Since its initial applications in Candida albicans, CRISPR-Cas9 platforms are rapidly evolving to permit efficient gene editing in other members of the genus. The technology has proven useful in elucidating the pathogenesis and host-pathogen interactions of medically relevant Candida species, and has led to novel insights on antifungal drug susceptibility and resistance, as well as innovative treatment strategies. CRISPR-Cas9 tools have also been exploited to uncover potential applications of Candida species in industrial contexts. This review is intended to provide a historical overview of genetic approaches used to study the Candida genus and to discuss the state of the art of CRISPR-based genetic manipulation of Candida species, highlighting its contributions to deciphering the biology of this genus, as well as providing perspectives for the future of Candida genetics.
Collapse
Affiliation(s)
- Deeva Uthayakumar
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Jehoshua Sharma
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Lauren Wensing
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
22
|
Horianopoulos LC, Lee CWJ, Hu G, Caza M, Kronstad JW. Dnj1 Promotes Virulence in Cryptococcus neoformans by Maintaining Robust Endoplasmic Reticulum Homeostasis Under Temperature Stress. Front Microbiol 2021; 12:727039. [PMID: 34566931 PMCID: PMC8461255 DOI: 10.3389/fmicb.2021.727039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
The capacity of opportunistic fungal pathogens such as Cryptococcus neoformans to cause disease is dependent on their ability to overcome an onslaught of stresses including elevated temperature under mammalian host conditions. Protein chaperones and co-chaperones play key roles in thermotolerance. In this study, we characterized the role of the endoplasmic reticulum (ER) J-domain containing co-chaperone, Dnj1, in the virulence of C. neoformans. A strain expressing a Dnj1-GFP fusion protein was used to confirm localization to the ER, and a dnj1∆ deletion mutant was shown to be hypersensitive to the ER stress caused by tunicamycin (TM) or 4μ8C. Dnj1 and another ER chaperone, calnexin were found to coordinately maintain ER homeostasis and contribute to maintenance of cell wall architecture. Dnj1 also contributed to thermotolerance and increased in abundance at elevated temperatures representative of febrile patients (e.g., 39°C) thus highlighting its role as a temperature-responsive J domain protein. The elaboration of virulence factors such as the polysaccharide capsule and extracellular urease activity were also markedly impaired in the dnj1∆ mutant when induced at human body temperature (i.e., 37°C). These virulence factors are immunomodulatory and, indeed, infection with the dnj1∆ mutant revealed impaired induction of the cytokines IL-6, IL-10, and MCP-1 in the lungs of mice compared to infection with wild type or complemented strains. The dnj1∆ mutant also had attenuated virulence in an intranasal murine model of cryptococcosis. Altogether, our data indicate that Dnj1 is crucial for survival and virulence factor production at elevated temperatures. The characterization of this co-chaperone also highlights the importance of maintaining homeostasis in the ER for the pathogenesis of C. neoformans.
Collapse
Affiliation(s)
| | - Christopher W J Lee
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Guanggan Hu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Mélissa Caza
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - James W Kronstad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
23
|
Zhou J, Li J, Cheong I, Liu NN, Wang H. Evaluation of artemisinin derivative artemether as a fluconazole potentiator through inhibition of Pdr5. Bioorg Med Chem 2021; 44:116293. [PMID: 34243044 DOI: 10.1016/j.bmc.2021.116293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 01/08/2023]
Abstract
Antifungal development has gained increasing attention due to its limited armamentarium and drug resistance. Drug repurposing holds great potential in antifungal discovery. In this study, we explored the antifungal activity of artemisinin and its derivatives, dihydroartemisinin, artesunate and artemether. We identified that artemisinins can inhibit the growth of Candida albicans, and can enhance the activity of three commonly used antifungals, amphotericin B, micafungin and fluconazole (FLC), on Candida albicans growth and filamentation. Artemisinins possess stronger antifungal effect with FLC than with other antifungals. Among artemisinins, artemether exhibits the most potent antifungal activity with FLC and can recover the susceptibility of FLC-resistant clinical isolates to FLC treatment. The combinatorial antifungal activity of artemether and FLC is broad-spectrum, as it can inhibit the growth of Candida auris, Candida tropicalis, Candida parapsilosis, Saccharomyces cerevisiae and Cryptococcus neoformans. Mechanistic investigation revealed that artemether might enhance azole efficacy through disrupting the function of Pdr5, leading to intracellular accumulation of FLC. This study identified artemether as a novel FLC potentiator, providing potential therapeutic insights against fungal infection and antifungal resistance.
Collapse
Affiliation(s)
- Jia Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jinyang Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Iohong Cheong
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
24
|
Yuan R, Tu J, Sheng C, Chen X, Liu N. Effects of Hsp90 Inhibitor Ganetespib on Inhibition of Azole-Resistant Candida albicans. Front Microbiol 2021; 12:680382. [PMID: 34093502 PMCID: PMC8174564 DOI: 10.3389/fmicb.2021.680382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Candida albicans is the most common fungal pathogen. Recently, drug resistance of C. albicans is increasingly severe. Hsp90 is a promising antifungal target to overcome this problem. To evaluate the effects of Hsp90 inhibitor ganetespib on the inhibition of azole-resistant C. albicans, the microdilution checkerboard method was used to measure the in vitro synergistic efficacy of ganetespib. The XTT/menadione reduction assay, microscopic observation, and Rh6G efflux assay were established to investigate the effects of ganetespib on azole-resistant C. albicans biofilm formation, filamentation, and efflux pump. Real-time RT-PCR analysis was employed to clarify the mechanism of antagonizing drug resistance. The in vivo antifungal efficacy of ganetespib was determined by the infectious model of azole-resistant C. albicans. Ganetespib showed an excellent synergistic antifungal activity in vitro and significantly inhibited the fungal biofilm formation, whereas it had no inhibitory effect on fungal hypha formation. Expression of azole-targeting enzyme gene ERG11 and efflux pump genes CDR1, CDR2, and MDR1 was significantly down-regulated when ganetespib was used in combination with FLC. In a mouse model infected with FLC-resistant C. albicans, the combination of ganetespib and FLC effectively reversed the FLC resistance and significantly decreased the kidney fungal load of mouse.
Collapse
Affiliation(s)
- Rui Yuan
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, China
| | - Jie Tu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xi Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, China
| | - Na Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
25
|
Mechanistic Insight into Antimicrobial and Antioxidant Potential of Jasminum Species: A Herbal Approach for Disease Management. PLANTS 2021; 10:plants10061089. [PMID: 34071621 PMCID: PMC8227019 DOI: 10.3390/plants10061089] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 12/28/2022]
Abstract
Drug resistance among microbial pathogens and oxidative stress caused by reactive oxygen species are two of the most challenging global issues. Firstly, drug-resistant pathogens cause several fatalities every year. Secondly aging and a variety of diseases, such as cardiovascular disease and cancer, are associated with free radical generated oxidative stress. The treatments currently available are limited, ineffective, or less efficient, so there is an immediate need to tackle these issues by looking for new therapies to resolve resistance and neutralize the harmful effects of free radicals. In the 21st century, the best way to save humans from them could be by using plants as well as their bioactive constituents. In this specific context, Jasminum is a major plant genus that is used in the Ayurvedic system of medicine to treat a variety of ailments. The information in this review was gathered from a variety of sources, including books, websites, and databases such as Science Direct, PubMed, and Google Scholar. In this review, a total of 14 species of Jasminum have been found to be efficient and effective against a wide variety of microbial pathogens. In addition, 14 species were found to be active free radical scavengers. The review is also focused on the disorders related to oxidative stress, and it was concluded that Jasminum grandiflorum and J. sambac normalized various parameters that were elevated by free radical generation. Alkaloids, flavonoids (rutoside), terpenes, phenols, and iridoid glucosides are among the main phytoconstituents found in various Jasminum species. Furthermore, this review also provides insight into the mechanistic basis of drug resistance, the generation of free radicals, and the role of Jasminum plants in combating resistance and neutralizing free radicals.
Collapse
|
26
|
Lee Y, Puumala E, Robbins N, Cowen LE. Antifungal Drug Resistance: Molecular Mechanisms in Candida albicans and Beyond. Chem Rev 2021; 121:3390-3411. [PMID: 32441527 PMCID: PMC8519031 DOI: 10.1021/acs.chemrev.0c00199] [Citation(s) in RCA: 432] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fungal infections are a major contributor to infectious disease-related deaths across the globe. Candida species are among the most common causes of invasive mycotic disease, with Candida albicans reigning as the leading cause of invasive candidiasis. Given that fungi are eukaryotes like their human host, the number of unique molecular targets that can be exploited for antifungal development remains limited. Currently, there are only three major classes of drugs approved for the treatment of invasive mycoses, and the efficacy of these agents is compromised by the development of drug resistance in pathogen populations. Notably, the emergence of additional drug-resistant species, such as Candida auris and Candida glabrata, further threatens the limited armamentarium of antifungals available to treat these serious infections. Here, we describe our current arsenal of antifungals and elaborate on the resistance mechanisms Candida species possess that render them recalcitrant to therapeutic intervention. Finally, we highlight some of the most promising therapeutic strategies that may help combat antifungal resistance, including combination therapy, targeting fungal-virulence traits, and modulating host immunity. Overall, a thorough understanding of the mechanistic principles governing antifungal drug resistance is fundamental for the development of novel therapeutics to combat current and emerging fungal threats.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Emily Puumala
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| |
Collapse
|
27
|
Horianopoulos LC, Kronstad JW. Chaperone Networks in Fungal Pathogens of Humans. J Fungi (Basel) 2021; 7:209. [PMID: 33809191 PMCID: PMC7998936 DOI: 10.3390/jof7030209] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The heat shock proteins (HSPs) function as chaperones to facilitate proper folding and modification of proteins and are of particular importance when organisms are subjected to unfavourable conditions. The human fungal pathogens are subjected to such conditions within the context of infection as they are exposed to human body temperature as well as the host immune response. Herein, the roles of the major classes of HSPs are briefly reviewed and their known contributions in human fungal pathogens are described with a focus on Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. The Hsp90s and Hsp70s in human fungal pathogens broadly contribute to thermotolerance, morphological changes required for virulence, and tolerance to antifungal drugs. There are also examples of J domain co-chaperones and small HSPs influencing the elaboration of virulence factors in human fungal pathogens. However, there are diverse members in these groups of chaperones and there is still much to be uncovered about their contributions to pathogenesis. These HSPs do not act in isolation, but rather they form a network with one another. Interactions between chaperones define their specific roles and enhance their protein folding capabilities. Recent efforts to characterize these HSP networks in human fungal pathogens have revealed that there are unique interactions relevant to these pathogens, particularly under stress conditions. The chaperone networks in the fungal pathogens are also emerging as key coordinators of pathogenesis and antifungal drug tolerance, suggesting that their disruption is a promising strategy for the development of antifungal therapy.
Collapse
Affiliation(s)
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
| |
Collapse
|
28
|
He X, Liu D, Chen Q. Proteomic analysis on the regulation of DOPA-melanin synthesis in Talaromyces marneffei. Microb Pathog 2020; 150:104701. [PMID: 33340654 DOI: 10.1016/j.micpath.2020.104701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/17/2020] [Accepted: 12/06/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Yeast form of T.marneffei can produce DOPA-melanin which perform an important role in the pathogen surviving in macrophage. So far, the proteomic associated with melanin synthesis remain unclearly in T.marneffei. METHODS The whole yeast cell proteins were extracted from T.marneffei cultured with or without l-DOPA. Using two-dimensional gel electrophoresis combined with MALDI-TOF mass spectrometry, distinguished proteins were identified between T.marneffei cultured with or without l-DOPA. Furthermore, geldanamycin were used to assess the inhibition effect on T.marneffei melanin production in vitro. RESULTS 16 distinguished proteins were identified in DOPA-melanized yeast cells, as well as 15 triple-up-expressed proteins and 7 triple-down-expressed proteins in comparison with non DOPA-melanized yeast cells. Of note, proteins differentially expressed proteins were predominantly heat shock proteins. HSP90/60/70 genes expressions increased significantly demonstrated by q-RT-PCR, which was consistent with the proteomics changes. GO analysis showed that the majority of differentially expressed proteins including HSPs(especially HSP90) were found enriched in stress response, cellular process, protein folding, stimuli response and biological process. KEGG pathway analysis showed that proteins were enriched predominantly in phagosome. HSP90 inhibitor(Geldanamycin) inhibited the brown-black pigment production of T.marneffei yeast grown on brain heart infusion agar, as well as the inhibition effect was observed by transmission electron microscope. CONCLUSIONS The results demonstrates that HSP90 palys an essential role in T.marneffei DOPA-melanin synthesis pathway.
Collapse
Affiliation(s)
- Xiaoyue He
- Department of Dermatology and Venereology, First Affiliated Hospital,Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Donghua Liu
- Department of Dermatology and Venereology, First Affiliated Hospital,Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Key Laboratory of AIDS Prevention and Treatment,Nanning, Guangxi, 530021, China.
| | - Qicong Chen
- Institutes for Life Sciences School of Medicine South China University of Technology Guangzhou, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
29
|
Delarze E, Brandt L, Trachsel E, Patxot M, Pralong C, Maranzano F, Chauvel M, Legrand M, Znaidi S, Bougnoux ME, d’Enfert C, Sanglard D. Identification and Characterization of Mediators of Fluconazole Tolerance in Candida albicans. Front Microbiol 2020; 11:591140. [PMID: 33262748 PMCID: PMC7686038 DOI: 10.3389/fmicb.2020.591140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Candida albicans is an important human pathogen and a major concern in intensive care units around the world. C. albicans infections are associated with a high mortality despite the use of antifungal treatments. One of the causes of therapeutic failures is the acquisition of antifungal resistance by mutations in the C. albicans genome. Fluconazole (FLC) is one of the most widely used antifungal and mechanisms of FLC resistance occurring by mutations have been extensively investigated. However, some clinical isolates are known to be able to survive at high FLC concentrations without acquiring resistance mutations, a phenotype known as tolerance. Mechanisms behind FLC tolerance are not well studied, mainly due to the lack of a proper way to identify and quantify tolerance in clinical isolates. We proposed here culture conditions to investigate FLC tolerance as well as an easy and efficient method to identity and quantify tolerance to FLC. The screening of C. albicans strain collections revealed that FLC tolerance is pH- and strain-dependent, suggesting the involvement of multiple mechanisms. Here, we addressed the identification of FLC tolerance mediators in C. albicans by an overexpression strategy focusing on 572 C. albicans genes. This strategy led to the identification of two transcription factors, CRZ1 and GZF3. CRZ1 is a C2H2-type transcription factor that is part of the calcineurin-dependent pathway in C. albicans, while GZF3 is a GATA-type transcription factor of unknown function in C. albicans. Overexpression of each gene resulted in an increase of FLC tolerance, however, only the deletion of CRZ1 in clinical FLC-tolerant strains consistently decreased their FLC tolerance. Transcription profiling of clinical isolates with variable levels of FLC tolerance confirmed a calcineurin-dependent signature in these isolates when exposed to FLC.
Collapse
Affiliation(s)
- Eric Delarze
- Department of Laboratory, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Ludivine Brandt
- Department of Laboratory, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Emilie Trachsel
- Department of Laboratory, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Marion Patxot
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Claire Pralong
- Department of Laboratory, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Fabio Maranzano
- Department of Laboratory, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Murielle Chauvel
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, Paris, France
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, Paris, France
| | - Sadri Znaidi
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, Paris, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, Paris, France
- Unité de Parasitologie-Mycologie, Service de Microbiologie Clinique, Hôpital Necker-Enfants-Malades, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
- Université de Paris, Paris, France
| | - Christophe d’Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, Paris, France
| | - Dominique Sanglard
- Department of Laboratory, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
30
|
LeBlanc EV, Polvi EJ, Veri AO, Privé GG, Cowen LE. Structure-guided approaches to targeting stress responses in human fungal pathogens. J Biol Chem 2020; 295:14458-14472. [PMID: 32796038 DOI: 10.1074/jbc.rev120.013731] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/11/2020] [Indexed: 11/06/2022] Open
Abstract
Fungi inhabit extraordinarily diverse ecological niches, including the human body. Invasive fungal infections have a devastating impact on human health worldwide, killing ∼1.5 million individuals annually. The majority of these deaths are attributable to species of Candida, Cryptococcus, and Aspergillus Treating fungal infections is challenging, in part due to the emergence of resistance to our limited arsenal of antifungal agents, necessitating the development of novel therapeutic options. Whereas conventional antifungal strategies target proteins or cellular components essential for fungal growth, an attractive alternative strategy involves targeting proteins that regulate fungal virulence or antifungal drug resistance, such as regulators of fungal stress responses. Stress response networks enable fungi to adapt, grow, and cause disease in humans and include regulators that are highly conserved across eukaryotes as well as those that are fungal-specific. This review highlights recent developments in elucidating crystal structures of fungal stress response regulators and emphasizes how this knowledge can guide the design of fungal-selective inhibitors. We focus on the progress that has been made with highly conserved regulators, including the molecular chaperone Hsp90, the protein phosphatase calcineurin, and the small GTPase Ras1, as well as with divergent stress response regulators, including the cell wall kinase Yck2 and trehalose synthases. Exploring structures of these important fungal stress regulators will accelerate the design of selective antifungals that can be deployed to combat life-threatening fungal diseases.
Collapse
Affiliation(s)
- Emmanuelle V LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth J Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda O Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert G Privé
- Departments of Medical Biophysics and Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Tornadic Shear Stress Induces a Transient, Calcineurin-Dependent Hypervirulent Phenotype in Mucorales Molds. mBio 2020; 11:mBio.01414-20. [PMID: 32605990 PMCID: PMC7327176 DOI: 10.1128/mbio.01414-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Trauma-related necrotizing myocutaneous mucormycosis (NMM) has a high morbidity and mortality in victims of combat-related injuries, geometeorological disasters, and severe burns. Inspired by the observation that several recent clusters of NMM have been associated with extreme mechanical forces (e.g., during tornados), we studied the impact of mechanical stress on Mucoralean biology and virulence in a Drosophila melanogaster infection model. In contrast to other experimental procedures to exert mechanical stress, tornadic shear challenge (TSC) by magnetic stirring induced a hypervirulent phenotype in several clinically relevant Mucorales species but not in Aspergillus or Fusarium Whereas fungal growth rates, morphogenesis, and susceptibility to noxious environments or phagocytes were not altered by TSC, soluble factors released in the supernatant of shear-challenged R. arrhizus spores rendered static spores hypervirulent. Consistent with a rapid decay of TSC-induced hypervirulence, minimal transcriptional changes were revealed by comparative RNA sequencing analysis of static and shear-challenged Rhizopus arrhizus However, inhibition of the calcineurin/heat shock protein 90 (hsp90) stress response circuitry by cyclosporine and tanespimycin abrogated the increased pathogenicity of R. arrhizus spores following TSC. Similarly, calcineurin loss-of-function mutants of Mucor circinelloides displayed no increased virulence capacity in flies after undergoing TSC. Collectively, these results establish that TSC induces hypervirulence specifically in Mucorales and point out the calcineurin/hsp90 pathway as a key orchestrator of this phenotype. Our findings invite future studies of topical calcineurin inhibitor treatment of wounds as an adjunct mitigation strategy for NMM following high-energy trauma.IMPORTANCE Given the limited efficacy of current medical treatments in trauma-related necrotizing mucormycosis, there is a dire need to better understand the Mucoralean pathophysiology in order to develop novel strategies to counteract fungal tissue invasion following severe trauma. Here, we describe that tornadic shear stress challenge transiently induces a hypervirulent phenotype in various pathogenic Mucorales species but not in other molds known to cause wound infections. Pharmacological and genetic inhibition of calcineurin signaling abrogated hypervirulence in shear stress-challenged Mucorales, encouraging further evaluation of (topical) calcineurin inhibitors to improve therapeutic outcomes of NMM after combat-related blast injuries or violent storms.
Collapse
|
32
|
The Novel J-Domain Protein Mrj1 Is Required for Mitochondrial Respiration and Virulence in Cryptococcus neoformans. mBio 2020; 11:mBio.01127-20. [PMID: 32518190 PMCID: PMC7373193 DOI: 10.1128/mbio.01127-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cryptococcus neoformans is the causative agent of cryptococcal meningitis, a disease responsible for ∼15% of all HIV-related deaths. Unfortunately, development of antifungal drugs is challenging because potential targets are conserved between humans and C. neoformans. In this context, we characterized a unique J-domain protein, Mrj1, which lacks orthologs in humans. We showed that Mrj1 was required for normal mitochondrial respiration and that mutants lacking Mrj1 were deficient in growth, capsule elaboration, and virulence. Furthermore, we were able to phenocopy the defects in growth and capsule elaboration by inhibiting respiration. This result suggests that the role of Mrj1 in mitochondrial function was responsible for the observed virulence defects and reinforces the importance of mitochondria to fungal pathogenesis. Mitochondria are difficult to target, as their function is also key to human cells; however, Mrj1 presents an opportunity to target a unique fungal protein required for mitochondrial function and virulence in C. neoformans. The opportunistic fungal pathogen Cryptococcus neoformans must adapt to the mammalian environment to establish an infection. Proteins facilitating adaptation to novel environments, such as chaperones, may be required for virulence. In this study, we identified a novel mitochondrial co-chaperone, Mrj1 (mitochondrial respiration J-domain protein 1), necessary for virulence in C. neoformans. The mrj1Δ and J-domain-inactivated mutants had general growth defects at both routine laboratory and human body temperatures and were deficient in the major virulence factor of capsule elaboration. The latter phenotype was associated with cell wall changes and increased capsular polysaccharide shedding. Accordingly, the mrj1Δ mutant was avirulent in a murine model of cryptococcosis. Mrj1 has a mitochondrial localization and co-immunoprecipitated with Qcr2, a core component of complex III of the electron transport chain. The mrj1 mutants were deficient in mitochondrial functions, including growth on alternative carbon sources, growth without iron, and mitochondrial polarization. They were also insensitive to complex III inhibitors and hypersensitive to an alternative oxidase (AOX) inhibitor, suggesting that Mrj1 functions in respiration. In support of this conclusion, mrj1 mutants also had elevated basal oxygen consumption rates which were completely abolished by the addition of the AOX inhibitor, confirming that Mrj1 is required for mitochondrial respiration through complexes III and IV. Furthermore, inhibition of complex III phenocopied the capsule and cell wall defects of the mrj1 mutants. Taken together, these results indicate that Mrj1 is required for normal mitochondrial respiration, a key aspect of adaptation to the host environment and virulence.
Collapse
|
33
|
Abstract
Candida albicans has remained the main etiological agent of candidiasis, challenges clinicians with high mortality and morbidity. The emergence of resistance to antifungal drugs, toxicity and lower efficacy have all contributed to an urgent need to develop alternative drugs aiming at novel targets in C. albicans. Targeting the production of virulence factors, which are essential processes for infectious agents, represents an attractive substitute for the development of newer anti-infectives. The present review highlights the recent developments made in the understanding of the pathogenicity of C. albicans. Production of hydrolytic enzymes, morphogenesis and biofilm formation, along with their molecular and metabolic regulation in Candida are discussed with regard to the development of novel antipathogenic drugs against candidiasis. Over the last decade, candidiasis has remained a major problematic disease worldwide. In spite of the existence of many antifungal drugs, the treatment of such diseases has still remained unsuccessful due to drug inefficacy. Therefore, there is a need to discover antifungals with different modes of action, such as antipathogenic drugs against Candida albicans. Here, we describe how various types of virulence factors such as proteinase, phospholipase, hemolysin, adhesion, morphogenesis and biofilm formation, could be targeted to develop novel therapeutics. We can inhibit production of these virulence factors by controlling their molecular/metabolic regulation.
Collapse
|
34
|
Tripathi SK, Feng Q, Liu L, Levin DE, Roy KK, Doerksen RJ, Baerson SR, Shi X, Pan X, Xu WH, Li XC, Clark AM, Agarwal AK. Puupehenone, a Marine-Sponge-Derived Sesquiterpene Quinone, Potentiates the Antifungal Drug Caspofungin by Disrupting Hsp90 Activity and the Cell Wall Integrity Pathway. mSphere 2020; 5:e00818-19. [PMID: 31915228 PMCID: PMC6952202 DOI: 10.1128/msphere.00818-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/04/2019] [Indexed: 02/02/2023] Open
Abstract
The cell wall-targeting echinocandin antifungals, although potent and well tolerated, are inadequate in treating fungal infections due to their narrow spectrum of activity and their propensity to induce pathogen resistance. A promising strategy to overcome these drawbacks is to combine echinocandins with a molecule that improves their activity and also disrupts drug adaptation pathways. In this study, we show that puupehenone (PUUP), a marine-sponge-derived sesquiterpene quinone, potentiates the echinocandin drug caspofungin (CAS) in CAS-resistant fungal pathogens. We have conducted RNA sequencing (RNA-seq) analysis, followed by genetic and molecular studies, to elucidate PUUP's CAS-potentiating mechanism. We found that the combination of CAS and PUUP blocked the induction of CAS-responding genes required for the adaptation to cell wall stress through the cell wall integrity (CWI) pathway. Further analysis showed that PUUP inhibited the activation of Slt2 (Mpk1), the terminal mitogen-activated protein (MAP) kinase in this pathway. We also found that PUUP induced heat shock response genes and inhibited the activity of heat shock protein 90 (Hsp90). Molecular docking studies predicted that PUUP occupies a binding site on Hsp90 required for the interaction between Hsp90 and its cochaperone Cdc37. Thus, we show that PUUP potentiates CAS activity by a previously undescribed mechanism which involves a disruption of Hsp90 activity and the CWI pathway. Given the requirement of the Hsp90-Cdc37 complex in Slt2 activation, we suggest that inhibitors of this complex would disrupt the CWI pathway and synergize with echinocandins. Therefore, the identification of PUUP's CAS-potentiating mechanism has important implications in the development of new antifungal combination therapies.IMPORTANCE Fungal infections cause more fatalities worldwide each year than malaria or tuberculosis. Currently available antifungal drugs have various limitations, including host toxicity, narrow spectrum of activity, and pathogen resistance. Combining these drugs with small molecules that can overcome these limitations is a useful strategy for extending their clinical use. We have investigated the molecular mechanism by which a marine-derived compound potentiates the activity of the antifungal echinocandin caspofungin. Our findings revealed a mechanism, different from previously reported caspofungin potentiators, in which potentiation is achieved by the disruption of Hsp90 activity and signaling through the cell wall integrity pathway, processes that play important roles in the adaptation to caspofungin in fungal pathogens. Given the importance of stress adaptation in the development of echinocandin resistance, this work will serve as a starting point in the development of new combination therapies that will likely be more effective and less prone to pathogen resistance.
Collapse
Affiliation(s)
- Siddharth K Tripathi
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Qin Feng
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Li Liu
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - David E Levin
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Kuldeep K Roy
- Division of Medicinal Chemistry, Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Robert J Doerksen
- Division of Medicinal Chemistry, Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Scott R Baerson
- Natural Products Utilization Research Unit, U.S. Department of Agriculture, Agricultural Research Service, Oxford, Mississippi, USA
| | - Xiaomin Shi
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Xuewen Pan
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Wen-Hui Xu
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Xing-Cong Li
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
- Division of Pharmacognosy, Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Alice M Clark
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
- Division of Pharmacognosy, Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| | - Ameeta K Agarwal
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
- Division of Pharmacology, Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi, USA
| |
Collapse
|
35
|
Daniyan MO, Przyborski JM, Shonhai A. Partners in Mischief: Functional Networks of Heat Shock Proteins of Plasmodium falciparum and Their Influence on Parasite Virulence. Biomolecules 2019; 9:E295. [PMID: 31340488 PMCID: PMC6681276 DOI: 10.3390/biom9070295] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
The survival of the human malaria parasite Plasmodium falciparum under the physiologically distinct environments associated with their development in the cold-blooded invertebrate mosquito vectors and the warm-blooded vertebrate human host requires a genome that caters to adaptability. To this end, a robust stress response system coupled to an efficient protein quality control system are essential features of the parasite. Heat shock proteins constitute the main molecular chaperone system of the cell, accounting for approximately two percent of the malaria genome. Some heat shock proteins of parasites constitute a large part (5%) of the 'exportome' (parasite proteins that are exported to the infected host erythrocyte) that modify the host cell, promoting its cyto-adherence. In light of their importance in protein folding and refolding, and thus the survival of the parasite, heat shock proteins of P. falciparum have been a major subject of study. Emerging evidence points to their role not only being cyto-protection of the parasite, as they are also implicated in regulating parasite virulence. In undertaking their roles, heat shock proteins operate in networks that involve not only partners of parasite origin, but also potentially functionally associate with human proteins to facilitate parasite survival and pathogenicity. This review seeks to highlight these interplays and their roles in parasite pathogenicity. We further discuss the prospects of targeting the parasite heat shock protein network towards the developments of alternative antimalarial chemotherapies.
Collapse
Affiliation(s)
- Michael O Daniyan
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State 220005, Nigeria.
| | - Jude M Przyborski
- Center of Infectious Diseases, Parasitology, University of Heidelberg Medical School, INF324, 69120 Heidelberg, Germany
| | - Addmore Shonhai
- Department of Biochemistry, School of Mathematical & Natural Sciences, University of Venda, P. Bag X5050, Thohoyandou 0950, South Africa.
| |
Collapse
|
36
|
O’Meara TR, O’Meara MJ, Polvi EJ, Pourhaghighi MR, Liston SD, Lin ZY, Veri AO, Emili A, Gingras AC, Cowen LE. Global proteomic analyses define an environmentally contingent Hsp90 interactome and reveal chaperone-dependent regulation of stress granule proteins and the R2TP complex in a fungal pathogen. PLoS Biol 2019; 17:e3000358. [PMID: 31283755 PMCID: PMC6638986 DOI: 10.1371/journal.pbio.3000358] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 07/18/2019] [Accepted: 06/19/2019] [Indexed: 11/18/2022] Open
Abstract
Hsp90 is a conserved molecular chaperone that assists in the folding and function of diverse cellular regulators, with a profound impact on biology, disease, and evolution. As a central hub of protein interaction networks, Hsp90 engages with hundreds of protein-protein interactions within eukaryotic cells. These interactions include client proteins, which physically interact with Hsp90 and depend on the chaperone for stability or function, as well as co-chaperones and partner proteins that modulate chaperone function. Currently, there are no methods to accurately predict Hsp90 interactors and there has been considerable network rewiring over evolutionary time, necessitating experimental approaches to define the Hsp90 network in the species of interest. This is a pressing challenge for fungal pathogens, for which Hsp90 is a key regulator of stress tolerance, drug resistance, and virulence traits. To address this challenge, we applied a novel biochemical fractionation and quantitative proteomic approach to examine alterations to the proteome upon perturbation of Hsp90 in a leading human fungal pathogen, Candida albicans. In parallel, we performed affinity purification coupled to mass spectrometry to define physical interacting partners for Hsp90 and the Hsp90 co-chaperones and identified 164 Hsp90-interacting proteins, including 111 that are specific to the pathogen. We performed the first analysis of the Hsp90 interactome upon antifungal drug stress and demonstrated that Hsp90 stabilizes processing body (P-body) and stress granule proteins that contribute to drug tolerance. We also describe novel roles for Hsp90 in regulating posttranslational modification of the Rvb1-Rvb2-Tah1-Pih1 (R2TP) complex and the formation of protein aggregates in response to thermal stress. This study provides a global view of the Hsp90 interactome in a fungal pathogen, demonstrates the dynamic role of Hsp90 in response to environmental perturbations, and highlights a novel connection between Hsp90 and the regulation of mRNA-associated protein granules.
Collapse
Affiliation(s)
- Teresa R. O’Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Matthew J. O’Meara
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
| | - Elizabeth J. Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - M. Reza Pourhaghighi
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Sean D. Liston
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Zhen-Yuan Lin
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Amanda O. Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Andrew Emili
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
- Department of Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
37
|
Genomic Identification of the TOR Signaling Pathway as a Target of the Plant Alkaloid Antofine in the Phytopathogen Fusarium graminearum. mBio 2019; 10:mBio.00792-19. [PMID: 31186319 PMCID: PMC6561021 DOI: 10.1128/mbio.00792-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Fusarium head blight caused by the fungal pathogen Fusarium graminearum is a devastating disease of cereal crops worldwide, with limited effective chemical treatments available. Here we show that the natural alkaloid compound antofine can inhibit fusarium head blight in wheat. Using yeast genomic screening, we identified the TOR pathway component RRD2 as a target of antofine that is also required for F. graminearum pathogenicity. Antofine, a phenanthroindolizidine alkaloid, is a bioactive natural product isolated from milkweeds that exhibits numerous biological activities, including anticancer, antimicrobial, antiviral, and anti-inflammatory properties. However, the direct targets and mode of action of antofine have not been determined. In this report, we show that antofine displays antifungal properties against the phytopathogen Fusarium graminearum, the cause of Fusarium head blight disease (FHB). FHB does devastating damage to agriculture, causing billions of dollars in economic losses annually. We therefore sought to understand the mode of action of antofine in F. graminearum using insights from yeast chemical genomic screens. We used haploinsufficiency profiling (HIP) to identify putative targets of antofine in yeast and identified three candidate targets, two of which had homologs in F. graminearum. The Fusarium homologues of two targets, glutamate dehydrogenase (FgGDH) and resistance to rapamycin deletion 2 (FgRRD2), can bind antofine. Of the two genes, only the Fgrrd2 knockout displayed a loss of virulence in wheat, indicating that RRD2 is an antivirulence target of antofine in F. graminearum. Mechanistically, we demonstrate that antofine disrupts the interaction between FgRRD2 and FgTap42, which is part of the Tap42-phosphatase complex in the target of rapamycin (TOR) signaling pathway, a central regulator of cell growth in eukaryotes and a pathway of extensive study for controlling numerous pathologies.
Collapse
|
38
|
Genetic Analysis of Candida auris Implicates Hsp90 in Morphogenesis and Azole Tolerance and Cdr1 in Azole Resistance. mBio 2019; 10:mBio.02529-18. [PMID: 30696744 PMCID: PMC6355988 DOI: 10.1128/mbio.02529-18] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida auris is an emerging fungal pathogen and a serious global health threat as the majority of clinical isolates display elevated resistance to currently available antifungal drugs. Despite the increased prevalence of C. auris infections, the mechanisms governing drug resistance remain largely elusive. In diverse fungi, the evolution of drug resistance is enabled by the essential molecular chaperone Hsp90, which stabilizes key regulators of cellular responses to drug-induced stress. Hsp90 also orchestrates temperature-dependent morphogenesis in Candida albicans, a key virulence trait. However, the role of Hsp90 in the pathobiology of C. auris remains unknown. In order to study regulatory functions of Hsp90 in C. auris, we placed HSP90 under the control of a doxycycline-repressible promoter to enable transcriptional repression. We found that Hsp90 is essential for growth in C. auris and that it enables tolerance of clinical isolates with respect to the azoles, which inhibit biosynthesis of the membrane sterol ergosterol. High-level azole resistance was independent of Hsp90 but dependent on the ABC transporter CDR1, deletion of which resulted in abrogated resistance. Strikingly, we discovered that C. auris undergoes a morphogenetic transition from yeast to filamentous growth in response to HSP90 depletion or cell cycle arrest but not in response to other cues that induce C. albicans filamentation. Finally, we observed that this developmental transition is associated with global transcriptional changes, including the induction of cell wall-related genes. Overall, this report provides a novel insight into mechanisms of drug tolerance and resistance in C. auris and describes a developmental transition in response to perturbation of a core regulator of protein homeostasis.IMPORTANCE Fungal pathogens pose a serious threat to public health. Candida auris is an emerging fungal pathogen that is often resistant to commonly used antifungal drugs. However, the mechanisms governing drug resistance and virulence in this organism remain largely unexplored. In this study, we adapted a conditional expression system to modulate the transcription of an essential gene, HSP90, which regulates antifungal resistance and virulence in diverse fungal pathogens. We showed that Hsp90 is essential for growth in C. auris and is important for tolerance of the clinically important azole antifungals, which block ergosterol biosynthesis. Further, we established that the Cdr1 efflux transporter regulates azole resistance. Finally, we discovered that C. auris transitions from yeast to filamentous growth in response to Hsp90 inhibition, accompanied by global transcriptional remodeling. Overall, this work provides a novel insight into mechanisms regulating azole resistance in C. auris and uncovers a distinct developmental program regulated by Hsp90.
Collapse
|
39
|
Whitesell L, Robbins N, Huang DS, McLellan CA, Shekhar-Guturja T, LeBlanc EV, Nation CS, Hui R, Hutchinson A, Collins C, Chatterjee S, Trilles R, Xie JL, Krysan DJ, Lindquist S, Porco JA, Tatu U, Brown LE, Pizarro J, Cowen LE. Structural basis for species-selective targeting of Hsp90 in a pathogenic fungus. Nat Commun 2019; 10:402. [PMID: 30679438 PMCID: PMC6345968 DOI: 10.1038/s41467-018-08248-w] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022] Open
Abstract
New strategies are needed to counter the escalating threat posed by drug-resistant fungi. The molecular chaperone Hsp90 affords a promising target because it supports survival, virulence and drug-resistance across diverse pathogens. Inhibitors of human Hsp90 under development as anticancer therapeutics, however, exert host toxicities that preclude their use as antifungals. Seeking a route to species-selectivity, we investigate the nucleotide-binding domain (NBD) of Hsp90 from the most common human fungal pathogen, Candida albicans. Here we report structures for this NBD alone, in complex with ADP or in complex with known Hsp90 inhibitors. Encouraged by the conformational flexibility revealed by these structures, we synthesize an inhibitor with >25-fold binding-selectivity for fungal Hsp90 NBD. Comparing co-crystals occupied by this probe vs. anticancer Hsp90 inhibitors revealed major, previously unreported conformational rearrangements. These insights and our probe's species-selectivity in culture support the feasibility of targeting Hsp90 as a promising antifungal strategy.
Collapse
Affiliation(s)
- Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - David S Huang
- Department of Chemistry, Center for Molecular Discovery, Boston University, Boston, MA, 02215, USA
| | | | - Tanvi Shekhar-Guturja
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Emmanuelle V LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Catherine S Nation
- Department of Tropical Medicine, School of Public Health and Tropical Medicine and Vector-Borne Infectious Disease Research Center, Tulane University, New Orleans, LA, 70112, USA
| | - Raymond Hui
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Ashley Hutchinson
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Cathy Collins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Sharanya Chatterjee
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Richard Trilles
- Department of Chemistry, Center for Molecular Discovery, Boston University, Boston, MA, 02215, USA
| | - Jinglin L Xie
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Damian J Krysan
- Departments of Pediatrics and Microbiology/Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - John A Porco
- Department of Chemistry, Center for Molecular Discovery, Boston University, Boston, MA, 02215, USA
| | - Utpal Tatu
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Lauren E Brown
- Department of Chemistry, Center for Molecular Discovery, Boston University, Boston, MA, 02215, USA
| | - Juan Pizarro
- Department of Tropical Medicine, School of Public Health and Tropical Medicine and Vector-Borne Infectious Disease Research Center, Tulane University, New Orleans, LA, 70112, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
40
|
Ramirez-Garcia A, Pellon A, Rementeria A, Buldain I, Barreto-Bergter E, Rollin-Pinheiro R, de Meirelles JV, Xisto MIDS, Ranque S, Havlicek V, Vandeputte P, Govic YL, Bouchara JP, Giraud S, Chen S, Rainer J, Alastruey-Izquierdo A, Martin-Gomez MT, López-Soria LM, Peman J, Schwarz C, Bernhardt A, Tintelnot K, Capilla J, Martin-Vicente A, Cano-Lira J, Nagl M, Lackner M, Irinyi L, Meyer W, de Hoog S, Hernando FL. Scedosporium and Lomentospora: an updated overview of underrated opportunists. Med Mycol 2018. [PMID: 29538735 DOI: 10.1093/mmy/myx113] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Species of Scedosporium and Lomentospora are considered as emerging opportunists, affecting immunosuppressed and otherwise debilitated patients, although classically they are known from causing trauma-associated infections in healthy individuals. Clinical manifestations range from local infection to pulmonary colonization and severe invasive disease, in which mortality rates may be over 80%. These unacceptably high rates are due to the clinical status of patients, diagnostic difficulties, and to intrinsic antifungal resistance of these fungi. In consequence, several consortia have been founded to increase research efforts on these orphan fungi. The current review presents recent findings and summarizes the most relevant points, including the Scedosporium/Lomentospora taxonomy, environmental distribution, epidemiology, pathology, virulence factors, immunology, diagnostic methods, and therapeutic strategies.
Collapse
Affiliation(s)
- Andoni Ramirez-Garcia
- Fungal and Bacterial Biomics Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Aize Pellon
- Fungal and Bacterial Biomics Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Aitor Rementeria
- Fungal and Bacterial Biomics Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Idoia Buldain
- Fungal and Bacterial Biomics Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | | | | | | | - Stephane Ranque
- Laboratoire de Parasitologie-Mycologie, AP-HM / CHU Timone, Marseille, France
| | - Vladimir Havlicek
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Patrick Vandeputte
- Laboratoire de Parasitologie-Mycologie, CHU, Angers, France.,Host-Pathogen Interaction Study Group (EA 3142), UNIV Angers, UNIV Brest, Angers, France
| | - Yohann Le Govic
- Laboratoire de Parasitologie-Mycologie, CHU, Angers, France.,Host-Pathogen Interaction Study Group (EA 3142), UNIV Angers, UNIV Brest, Angers, France
| | - Jean-Philippe Bouchara
- Laboratoire de Parasitologie-Mycologie, CHU, Angers, France.,Host-Pathogen Interaction Study Group (EA 3142), UNIV Angers, UNIV Brest, Angers, France
| | - Sandrine Giraud
- Host-Pathogen Interaction Study Group (EA 3142), UNIV Angers, UNIV Brest, Angers, France
| | - Sharon Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, ICPMR, Westmead Hospital, The University of Sydney, New South Wales, Australia
| | - Johannes Rainer
- Institute of Microbiology, Leopold-Franzens University Innsbruck, Austria
| | - Ana Alastruey-Izquierdo
- Mycology Reference Laboratory, National Centre for Microbiology. Instituto de Salud Carlos III. Majadahonda, Madrid, Spain
| | | | | | - Javier Peman
- Microbiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Carsten Schwarz
- Cystic Fibrosis Centre Berlin/Charité-Universitätsmedizin Berlin, Germany
| | - Anne Bernhardt
- Mycotic and Parasitic Agents and Mycobacteria, Robert Koch Institute, Berlin, Germany
| | - Kathrin Tintelnot
- Mycotic and Parasitic Agents and Mycobacteria, Robert Koch Institute, Berlin, Germany
| | - Javier Capilla
- Mycology Unit, Medical School and IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - Adela Martin-Vicente
- Mycology Unit, Medical School and IISPV, Universitat Rovira i Virgili, Reus, Spain.,Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN USA
| | - Jose Cano-Lira
- Mycology Unit, Medical School and IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - Markus Nagl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Lackner
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Laszlo Irinyi
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Westmead Clinical School, Sydney Medical School - Westmead Hospital, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Institute for Medical Research, Sydney, New South Wales, Australia
| | - Wieland Meyer
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Westmead Clinical School, Sydney Medical School - Westmead Hospital, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Institute for Medical Research, Sydney, New South Wales, Australia
| | - Sybren de Hoog
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Fernando L Hernando
- Fungal and Bacterial Biomics Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
41
|
Rocha MC, Santos CA, Malavazi I. The Regulatory Function of the Molecular Chaperone Hsp90 in the Cell Wall Integrity of Pathogenic Fungi. CURR PROTEOMICS 2018. [DOI: 10.2174/1570164615666180820155807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Different signaling cascades including the Cell Wall Integrity (CWI), the High Osmolarity Glycerol (HOG) and the Ca2+/calcineurin pathways control the cell wall biosynthesis and remodeling in fungi. Pathogenic fungi, such as Aspergillus fumigatus and Candida albicans, greatly rely on these signaling circuits to cope with different sources of stress, including the cell wall stress evoked by antifungal drugs and the host’s response during infection. Hsp90 has been proposed as an important regulatory protein and an attractive target for antifungal therapy since it stabilizes major effector proteins that act in the CWI, HOG and Ca2+/calcineurin pathways. Data from the human pathogen C. albicans have provided solid evidence that loss-of-function of Hsp90 impairs the evolution of resistance to azoles and echinocandin drugs. In A. fumigatus, Hsp90 is also required for cell wall integrity maintenance, reinforcing a coordinated function of the CWI pathway and this essential molecular chaperone. In this review, we focus on the current information about how Hsp90 impacts the aforementioned signaling pathways and consequently the homeostasis and maintenance of the cell wall, highlighting this cellular event as a key mechanism underlying antifungal therapy based on Hsp90 inhibition.
Collapse
Affiliation(s)
- Marina Campos Rocha
- Departmento de Genetica e Evolucao, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sao Carlos, Sao Carlos, Brazil
| | - Camilla Alves Santos
- Departmento de Genetica e Evolucao, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sao Carlos, Sao Carlos, Brazil
| | - Iran Malavazi
- Departmento de Genetica e Evolucao, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sao Carlos, Sao Carlos, Brazil
| |
Collapse
|
42
|
Tome M, Zupan J, Tomičić Z, Matos T, Raspor P. Synergistic and antagonistic effects of immunomodulatory drugs on the action of antifungals against Candida glabrata and Saccharomyces cerevisiae. PeerJ 2018; 6:e4999. [PMID: 29915703 PMCID: PMC6004109 DOI: 10.7717/peerj.4999] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/29/2018] [Indexed: 12/16/2022] Open
Abstract
Candidemia and other forms of invasive fungal infections caused by Candida glabrata and to a lesser extent Saccharomyces cerevisiae are a serious health problem, especially if their steadily rising resistance to the limited range of antifungal drugs is taken into consideration. Various drug combinations are an attractive solution to the resistance problem, and some drug combinations are already common in the clinical environment due to the nature of diseases or therapies. We tested a few of the common antifungal-immunomodulatory drug combinations and evaluated their effect on selected strains of C. glabrata and S. cerevisiae. The combinations were performed using the checkerboard microdilution assay and interpreted using the Loewe additivity model and a model based on the Bliss independence criterion. A synergistic interaction was confirmed between calcineurin inhibitors (Fk506 and cyclosporine A) and antifungals (fluconazole, itraconazole, and amphotericin B). A new antagonistic interaction between mycophenolic acid (MPA) and azole antifungals was discovered in non-resistant strains. A possible mechanism that explains this is induction of the Cdr1 efflux pump by MPA in C. glabrata ATCC 2001. The Pdr1 regulatory cascade plays a role in overall resistance to fluconazole, but it is not essential for the antagonistic interaction. This was confirmed by the Cgpdr1Δ mutant still displaying the antagonistic interaction between the drugs, although at lower concentrations of fluconazole. This antagonism calls into question the use of simultaneous therapy with MPA and azoles in the clinical environment.
Collapse
Affiliation(s)
- Miha Tome
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Zupan
- Biotechnology, Microbiology, and Food Safety, Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia.,Lek d.d., Ljubljana, Slovenia
| | - Zorica Tomičić
- Faculty of Technology, University of Novi Sad, Novi Sad, Serbia
| | - Tadeja Matos
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Raspor
- Biotechnology, Microbiology, and Food Safety, Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia.,Retired from University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
43
|
Geddes-McAlister J, Shapiro RS. New pathogens, new tricks: emerging, drug-resistant fungal pathogens and future prospects for antifungal therapeutics. Ann N Y Acad Sci 2018; 1435:57-78. [DOI: 10.1111/nyas.13739] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/19/2018] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Jennifer Geddes-McAlister
- Department of Molecular and Cellular Biology; University of Guelph; Guelph Ontario Canada
- Department of Proteomics and Signal Transduction; Max Planck Institute of Biochemistry; Munich Germany
| | - Rebecca S. Shapiro
- Department of Molecular and Cellular Biology; University of Guelph; Guelph Ontario Canada
| |
Collapse
|
44
|
Abstract
The fungal pathogens Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus have transitioned from a rare curiosity to a leading cause of human mortality. The management of infections caused by these organisms is intimately dependent on the efficacy of antifungal agents; however, fungi that are resistant to these treatments are regularly isolated in the clinic, impeding our ability to control infections. Given the significant impact fungal pathogens have on human health, it is imperative to understand the molecular mechanisms that govern antifungal drug resistance. This review describes our current knowledge of the mechanisms by which antifungal drug resistance evolves in experimental populations and clinical settings. We explore current antifungal treatment options and discuss promising strategies to impede the evolution of drug resistance. By tackling antifungal drug resistance as an evolutionary problem, there is potential to improve the utility of current treatments and accelerate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; , ,
| | - Tavia Caplan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; , ,
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; , ,
| |
Collapse
|
45
|
Mount HO, Revie NM, Todd RT, Anstett K, Collins C, Costanzo M, Boone C, Robbins N, Selmecki A, Cowen LE. Global analysis of genetic circuitry and adaptive mechanisms enabling resistance to the azole antifungal drugs. PLoS Genet 2018; 14:e1007319. [PMID: 29702647 PMCID: PMC5922528 DOI: 10.1371/journal.pgen.1007319] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
Invasive fungal infections caused by the pathogen Candida albicans have transitioned from a rare curiosity to a major cause of human mortality. This is in part due to the emergence of resistance to the limited number of antifungals available to treat fungal infections. Azoles function by targeting the biosynthesis of ergosterol, a key component of the fungal cell membrane. Loss-of-function mutations in the ergosterol biosynthetic gene ERG3 mitigate azole toxicity and enable resistance that depends upon fungal stress responses. Here, we performed a genome-wide synthetic genetic array screen in Saccharomyces cerevisiae to map ERG3 genetic interactors and uncover novel circuitry important for azole resistance. We identified nine genes that enabled erg3-mediated azole resistance in the model yeast and found that only two of these genes had a conserved impact on resistance in C. albicans. Further, we screened a C. albicans homozygous deletion mutant library and identified 13 genes for which deletion enhances azole susceptibility. Two of the genes, RGD1 and PEP8, were also important for azole resistance acquired by diverse mechanisms. We discovered that loss of function of retrograde transport protein Pep8 overwhelms the functional capacity of the stress response regulator calcineurin, thereby abrogating azole resistance. To identify the mechanism through which the GTPase activator protein Rgd1 enables azole resistance, we selected for mutations that restore resistance in strains lacking Rgd1. Whole genome sequencing uncovered parallel adaptive mechanisms involving amplification of both chromosome 7 and a large segment of chromosome 3. Overexpression of a transporter gene on the right portion of chromosome 3, NPR2, was sufficient to enable azole resistance in the absence of Rgd1. Thus, we establish a novel mechanism of adaptation to drug-induced stress, define genetic circuitry underpinning azole resistance, and illustrate divergence in resistance circuitry over evolutionary time. Fungal infections caused by the pathogen Candida albicans pose a serious threat to human health. Treating these infections relies heavily on the azole antifungals, however, resistance to these drugs develops readily demanding novel therapeutic strategies. We performed large-scale systematic screens in both C. albicans and the model yeast Saccharomyces cerevisiae to identify genes that enable azole resistance. Our genome-wide screen in S. cerevisiae identified nine determinants of azole resistance, only two of which were important for resistance in C. albicans. Our screen of C. albicans mutants identified 13 genes for which deletion enhances susceptibility to azoles, including RGD1 and PEP8. We found that loss of Pep8 overwhelms the functional capacity of a key stress response regulator, calcineurin. In contrast, amplification of chromosome 7 and the right portion of chromosome 3 can restore resistance in strains lacking Rgd1, suggesting that Rgd1 may enable azole resistance by inducing genes in these amplified regions. Specifically, overexpression of a gene involved in transport on chromosome 3, NPR2, was sufficient to restore azole resistance in the absence of Rgd1. Thus, we establish novel circuitry important for antifungal drug resistance, and uncover adaptive mechanisms involving genomic plasticity that occur in response to drug induced stress.
Collapse
Affiliation(s)
| | - Nicole M. Revie
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Robert T. Todd
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Kaitlin Anstett
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Cathy Collins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Costanzo
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Anna Selmecki
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
46
|
Veri AO, Miao Z, Shapiro RS, Tebbji F, O’Meara TR, Kim SH, Colazo J, Tan K, Vyas VK, Whiteway M, Robbins N, Wong KH, Cowen LE. Tuning Hsf1 levels drives distinct fungal morphogenetic programs with depletion impairing Hsp90 function and overexpression expanding the target space. PLoS Genet 2018; 14:e1007270. [PMID: 29590106 PMCID: PMC5873724 DOI: 10.1371/journal.pgen.1007270] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/22/2018] [Indexed: 12/24/2022] Open
Abstract
The capacity to respond to temperature fluctuations is critical for microorganisms to survive within mammalian hosts, and temperature modulates virulence traits of diverse pathogens. One key temperature-dependent virulence trait of the fungal pathogen Candida albicans is its ability to transition from yeast to filamentous growth, which is induced by environmental cues at host physiological temperature. A key regulator of temperature-dependent morphogenesis is the molecular chaperone Hsp90, which has complex functional relationships with the transcription factor Hsf1. Although Hsf1 controls global transcriptional remodeling in response to heat shock, its impact on morphogenesis remains unknown. Here, we establish an intriguing paradigm whereby overexpression or depletion of C. albicans HSF1 induces morphogenesis in the absence of external cues. HSF1 depletion compromises Hsp90 function, thereby driving filamentation. HSF1 overexpression does not impact Hsp90 function, but rather induces a dose-dependent expansion of Hsf1 direct targets that drives overexpression of positive regulators of filamentation, including Brg1 and Ume6, thereby bypassing the requirement for elevated temperature during morphogenesis. This work provides new insight into Hsf1-mediated environmentally contingent transcriptional control, implicates Hsf1 in regulation of a key virulence trait, and highlights fascinating biology whereby either overexpression or depletion of a single cellular regulator induces a profound developmental transition. For human pathogens, the capacity to respond to elevated temperature is required for survival, with elevated temperature in the form of fever as a conserved host response to defend against infection. One of the leading fungal pathogens of humans in Candida albicans, which is capable of growing in both a yeast and filamentous state. The ability to transition between these forms is a key virulence trait, and one that is highly temperature-dependent. A pivotal regulator of filamentous growth is the temperature-responsive molecular chaperone Hsp90, which has complex relationships with the transcription factor Hsf1. Although Hsf1 regulates changes in gene expression in response to heat shock, its impact on morphogenesis remains unknown. Here, we uncover an intriguing phenomenon whereby overexpression or depletion of C. albicans HSF1 induces morphogenesis. We observe that HSF1 depletion compromises Hsp90 function, thereby driving filamentation. In contrast, HSF1 overexpression induces a dose-dependent expansion of its transcriptional targets that drives overexpression of positive regulators of filamentous growth. This work illuminates novel mechanisms through which tuning the levels of an environmentally contingent transcription factor drives a key developmental program.
Collapse
Affiliation(s)
- Amanda O. Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Zhengqiang Miao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Rebecca S. Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Faiza Tebbji
- Infectious Disease Research Centre, Université Laval, Quebec City, Quebec, Canada
| | - Teresa R. O’Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sang Hu Kim
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Juan Colazo
- Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Kaeling Tan
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Valmik K. Vyas
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montréal, Quebec, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Koon Ho Wong
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
47
|
The Rim Pathway Mediates Antifungal Tolerance in Candida albicans through Newly Identified Rim101 Transcriptional Targets, Including Hsp90 and Ipt1. Antimicrob Agents Chemother 2018; 62:AAC.01785-17. [PMID: 29311085 DOI: 10.1128/aac.01785-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/19/2017] [Indexed: 12/13/2022] Open
Abstract
Invasive candidiasis (IC) is a major cause of morbidity and mortality despite antifungal treatment. Azoles and echinocandins are used as first-line therapies for IC. However, their efficacy is limited by yeast tolerance and the emergence of acquired resistance. Tolerance is a reversible stage created due to the yeast's capacity to counter antifungal drug exposure, leading to persistent growth. For Candida albicans, multiple stress signaling pathways have been shown to contribute to this adaptation. Among them, the pH-responsive Rim pathway, through its transcription factor Rim101p, was shown to mediate azole and echinocandin tolerance. The Rim pathway is fungus specific, is conserved among the members of the fungal kingdom, and plays a key role in pathogenesis and virulence. The present study aimed at confirming the role of Rim101p and investigating the implication of the other Rim proteins in antifungal tolerance in C. albicans, as well as the mechanisms underlying it. Time-kill curve experiments and colony formation tests showed that genetic inhibition of all the Rim factors enhances echinocandin and azole antifungal activity. Through RNA sequencing analysis of a rim101-/- mutant, a strain constitutively overexpressing RIM101, and control strains, we discovered novel Rim-dependent genes involved in tolerance, including HSP90, encoding a major molecular chaperone, and IPT1, involved in sphingolipid biosynthesis. Rim mutants were also hypersensitive to pharmacological inhibition of Hsp90. Taken together, these data suggest that Rim101 acts upstream of Hsp90 and that targeting the Rim pathway in combination with existing antifungal drugs may represent a promising antifungal strategy to indirectly but specifically target Hsp90 in yeasts.
Collapse
|
48
|
Liu N, Tu J, Dong G, Wang Y, Sheng C. Emerging New Targets for the Treatment of Resistant Fungal Infections. J Med Chem 2018; 61:5484-5511. [DOI: 10.1021/acs.jmedchem.7b01413] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Na Liu
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Jie Tu
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Yan Wang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| |
Collapse
|
49
|
Khandelwal NK, Chauhan N, Sarkar P, Esquivel BD, Coccetti P, Singh A, Coste AT, Gupta M, Sanglard D, White TC, Chauvel M, d'Enfert C, Chattopadhyay A, Gaur NA, Mondal AK, Prasad R. Azole resistance in a Candida albicans mutant lacking the ABC transporter CDR6/ROA1 depends on TOR signaling. J Biol Chem 2017; 293:412-432. [PMID: 29158264 DOI: 10.1074/jbc.m117.807032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
ATP-binding cassette (ABC) transporters help export various substrates across the cell membrane and significantly contribute to drug resistance. However, a recent study reported an unusual case in which the loss of an ABC transporter in Candida albicans, orf19.4531 (previously named ROA1), increases resistance against antifungal azoles, which was attributed to an altered membrane potential in the mutant strain. To obtain further mechanistic insights into this phenomenon, here we confirmed that the plasma membrane-localized transporter (renamed CDR6/ROA1 for consistency with C. albicans nomenclature) could efflux xenobiotics such as berberine, rhodamine 123, and paraquat. Moreover, a CDR6/ROA1 null mutant, NKKY101, displayed increased susceptibility to these xenobiotics. Interestingly, fluorescence recovery after photobleaching (FRAP) results indicated that NKKY101 mutant cells exhibited increased plasma membrane rigidity, resulting in reduced azole accumulation and contributing to azole resistance. Transcriptional profiling revealed that ribosome biogenesis genes were significantly up-regulated in the NKKY101 mutant. As ribosome biogenesis is a well-known downstream phenomenon of target of rapamycin (TOR1) signaling, we suspected a link between ribosome biogenesis and TOR1 signaling in NKKY101. Therefore, we grew NKKY101 cells on rapamycin and observed TOR1 hyperactivation, which leads to Hsp90-dependent calcineurin stabilization and thereby increased azole resistance. This in vitro finding was supported by in vivo data from a mouse model of systemic infection in which NKKY101 cells led to higher fungal load after fluconazole challenge than wild-type cells. Taken together, our study uncovers a mechanism of azole resistance in C. albicans, involving increased membrane rigidity and TOR signaling.
Collapse
Affiliation(s)
- Nitesh Kumar Khandelwal
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.,the International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India, and
| | - Neeraj Chauhan
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07103
| | - Parijat Sarkar
- the CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| | - Brooke D Esquivel
- the School of Biological Sciences, Cell Biology, and Biophysics, University of Missouri, Kansas City, Missouri 64110
| | - Paola Coccetti
- the Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy.,SYSBIO, Centre of Systems Biology, 20126 Milan, Italy
| | - Ashutosh Singh
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.,the Department of Biochemistry, Lucknow University, Lucknow 226024, Uttar Pradesh, India
| | - Alix T Coste
- the Institute of Microbiology, University of Lausanne and University Hospital Center, Rue du Bugnon 48, Lausanne, CH-1011, Switzerland
| | - Meghna Gupta
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.,the Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158
| | - Dominique Sanglard
- the Institute of Microbiology, University of Lausanne and University Hospital Center, Rue du Bugnon 48, Lausanne, CH-1011, Switzerland
| | - Theodore C White
- the School of Biological Sciences, Cell Biology, and Biophysics, University of Missouri, Kansas City, Missouri 64110
| | - Murielle Chauvel
- the Département Génomes et Génétique, Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, INRA, 75015 Paris, France
| | - Christophe d'Enfert
- the Département Génomes et Génétique, Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, INRA, 75015 Paris, France
| | | | - Naseem A Gaur
- the International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India, and
| | - Alok Kumar Mondal
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajendra Prasad
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India, .,the Amity Institute of Integrative Sciences and Health, Amity University Haryana, Amity Education Valley Gurgaon-122413, India
| |
Collapse
|
50
|
Xie JL, Qin L, Miao Z, Grys BT, Diaz JDLC, Ting K, Krieger JR, Tong J, Tan K, Leach MD, Ketela T, Moran MF, Krysan DJ, Boone C, Andrews BJ, Selmecki A, Ho Wong K, Robbins N, Cowen LE. The Candida albicans transcription factor Cas5 couples stress responses, drug resistance and cell cycle regulation. Nat Commun 2017; 8:499. [PMID: 28894103 PMCID: PMC5593949 DOI: 10.1038/s41467-017-00547-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/06/2017] [Indexed: 12/16/2022] Open
Abstract
The capacity to coordinate environmental sensing with initiation of cellular responses underpins microbial survival and is crucial for virulence and stress responses in microbial pathogens. Here we define circuitry that enables the fungal pathogen Candida albicans to couple cell cycle dynamics with responses to cell wall stress induced by echinocandins, a front-line class of antifungal drugs. We discover that the C. albicans transcription factor Cas5 is crucial for proper cell cycle dynamics and responses to echinocandins, which inhibit β-1,3-glucan synthesis. Cas5 has distinct transcriptional targets under basal and stress conditions, is activated by the phosphatase Glc7, and can regulate the expression of target genes in concert with the transcriptional regulators Swi4 and Swi6. Thus, we illuminate a mechanism of transcriptional control that couples cell wall integrity with cell cycle regulation, and uncover circuitry governing antifungal drug resistance.Cas5 is a transcriptional regulator of responses to cell wall stress in the fungal pathogen Candida albicans. Here, Xie et al. show that Cas5 also modulates cell cycle dynamics and responses to antifungal drugs.
Collapse
Affiliation(s)
- Jinglin L Xie
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
| | - Longguang Qin
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Zhengqiang Miao
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Ben T Grys
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada, M5S 3E1
| | - Jacinto De La Cruz Diaz
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, 14642, USA
| | - Kenneth Ting
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
| | - Jonathan R Krieger
- The Hospital for Sick Children, SPARC Biocentre, Toronto, ON, Canada, M5G 0A4
| | - Jiefei Tong
- The Hospital for Sick Children, Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada, M5G 0A4
| | - Kaeling Tan
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Michelle D Leach
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
- Aberdeen Fungal Group, School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Abderdeen, AB252ZD, UK
| | - Troy Ketela
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
| | - Michael F Moran
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
- The Hospital for Sick Children, SPARC Biocentre, Toronto, ON, Canada, M5G 0A4
- The Hospital for Sick Children, Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada, M5G 0A4
| | - Damian J Krysan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, 14642, USA
- Department of Pediatrics and Microbiology/Immunology, University of Rochester, Rochester, NY, 14642, USA
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada, M5S 3E1
| | - Brenda J Andrews
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada, M5S 3E1
| | - Anna Selmecki
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Koon Ho Wong
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5G 1M1.
| |
Collapse
|