1
|
Zajac C, Scott NE, Kline S, Erayil SE, Selmecki A. Hotspot gene conversion between FKS1 and FKS2 in echinocandin resistant Candida glabrata serial isolates. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:31. [PMID: 40247099 PMCID: PMC12006411 DOI: 10.1038/s44259-025-00102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025]
Abstract
Candida glabrata (Nakaseomyces glabratus) is the most common cause of drug-resistant candidemia and is associated with a high mortality rate. Only a few mechanisms of drug resistance are known in C. glabrata, predominantly involving recurrent single nucleotide polymorphisms. The importance of structural variation in acquired drug resistance is not understood. We performed comparative phenotypic and genomic analyses of six serial bloodstream isolates of C. glabrata and identified novel mutations associated with resistance to echinocandins. Critically, we identified a novel gene conversion event between the hotspot 2 regions of FKS1 and FKS2 that was associated with increased resistance to micafungin. We further analyzed 621 publicly available C. glabrata genomes and found three additional examples of structural variation involving FKS1 and FKS2. Ultimately, drug resistance in C. glabrata involves structural variants that are missed with current diagnostic methods and need to be considered when designing and implementing more effective antifungal management strategies.
Collapse
Affiliation(s)
- Christopher Zajac
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Nancy E Scott
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- University of Minnesota, Bioinformatics and Computational Biology Program, Minneapolis, MN, USA
| | - Susan Kline
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Serin E Erayil
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA.
- University of Minnesota, Bioinformatics and Computational Biology Program, Minneapolis, MN, USA.
| |
Collapse
|
2
|
Winkler ML, Deshpande L, Kimbrough JH, Karr M, Rhomberg P, Klauer AL, Castanheira M. Anidulafungin is a useful surrogate marker for predicting in vitro susceptibility to rezafungin among five Candida species using CLSI methods and interpretive criteria. J Clin Microbiol 2025; 63:e0112924. [PMID: 39840977 PMCID: PMC11837505 DOI: 10.1128/jcm.01129-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/23/2024] [Indexed: 01/23/2025] Open
Abstract
This study addresses the use of other echinocandins as surrogate markers to predict the susceptibility of rezafungin against the six most common Candida spp. The Clinical Laboratory Standards Institute (CLSI) reference broth microdilution method was performed to test 5,720 clinical isolates of six different Candida species. Species-specific interpretative criteria by CLSI breakpoints or epidemiological cutoff values were applied. Essential agreement was 100% within two doubling dilutions for all species and comparisons. The categorical agreement of rezafungin using anidulafungin against all Candida spp. was 97.6% (2.9% very major errors [VMEs], 0.2% major errors [MEs], and 2.2% minor errors [miEs]); for caspofungin, it was 99.6% (11.4% VME, 0.09% ME, and 0.19% miE); and for micafungin, it was 99.6% (14.3% VME, 0.15% ME, and 0.17% miE). There were species-specific differences that led to unacceptably high VME for Candida dubliniensis with all agents and for Candida parapsilosis when caspofungin or micafungin but not anidulafungin was used as the comparator. Genetic analysis showed rezafungin nonsusceptibility correlated well with FKS hotspot mutations. The best-performing surrogate was anidulafungin, which can be used to predict rezafungin susceptible or nonsusceptible in Candida albicans, Candida glabrata, Candida parapsilosis, Candida tropicalis, and Candida krusei with low error rates and ≥90% essential and categorical agreement. Micafungin or caspofungin can also be used as a surrogate marker for predicting rezafungin susceptible or nonsusceptible in C. albicans, C. glabrata, C. tropicalis, and C. krusei. No surrogate performs appropriately to determine rezafungin susceptibility for C. dubliniensis.
Collapse
Affiliation(s)
| | | | | | - Maura Karr
- Element Iowa City (JMI Laboratories), North Liberty, Iowa, USA
| | - Paul Rhomberg
- Element Iowa City (JMI Laboratories), North Liberty, Iowa, USA
| | - Abby L. Klauer
- Element Iowa City (JMI Laboratories), North Liberty, Iowa, USA
| | | |
Collapse
|
3
|
Doorley LA, Meza-Perez V, Jones SJ, Rybak JM. A Candidozyma ( Candida ) auris -optimized Episomal Plasmid Induced Cas9-editing system reveals the direct impact of the S639F encoding FKS1 mutation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638356. [PMID: 39990403 PMCID: PMC11844485 DOI: 10.1101/2025.02.14.638356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Objectives Mutations in the Candidozyma ( Candida ) auris β-glucan synthase gene ( FKS1 ) altering S639 are frequently associated with clinical echinocandin resistance. However, the direct impact of these mutations remains uncharacterized. We have developed a novel C. auris- optimized E pisomal P lasmid- I nduced C as9 (EPIC) gene-editing system capable of recyclable precision genome editing and demonstrate the contribution of FKS1 S639F mutation to echinocandin resistance. Methods The EPIC gene-editing system was generated for optimized use in C. auris , and ADE2 modification was evaluated in 5 C. auris clades. Mutations leading to Fks1 S639F and Fks1 WT were placed into echinocandin-susceptible and echinocandin-resistant isolates from Clade-III and -I, respectively, using the EPIC system. Echinocandin susceptibility was determined by CLSI broth microdilution, and cell wall abundance of chitin and β-glucan was assessed by staining with Calcofluor White (CFW) and Aniline Blue (AB). Results The EPIC system was capable of targeted ADE2 editing in C. auris isolates from 5 genetic clades and shown to be precise by confirmatory sequencing of 50 transformants. A single nucleotide change in FKS1 resulting in either the S639F substitution or a silent synonymous mutation was introduced in an echinocandin-susceptible Clade-III isolate. Precision FKS1 editing by the EPIC system was confirmed by whole genome sequencing. Subsequent susceptibility testing demonstrated introduction of the FKS1 S639F mutation to increase resistance to echinocandins. Moreover, introduction of a wildtype Fks1 sequence to an echinocandin-resistant Clade-I clinical isolate, correcting the FKS1 S639F mutation, resulted in a restoration of echinocandin sensitivity. Evaluation of cell wall composition showed isolates or strains harboring FKS1 S639F to contain significantly elevated β-glucan and chitin content relative to isogenic comparators. Conclusions These data demonstrate the potential of our EPIC system in its ability to introduce single nucleotide substitutions in multiple C. auris clade backgrounds while revealing the direct impact of the S639F encoding FKS1 mutation on echinocandin resistance.
Collapse
|
4
|
Scott NE, Wash E, Zajac C, Erayil SE, Kline SE, Selmecki A. Heterogeneity of Candida bloodstream isolates in an academic medical center and affiliated hospitals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636768. [PMID: 39975022 PMCID: PMC11839140 DOI: 10.1101/2025.02.05.636768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Invasive Candida bloodstream infections (candidemia) are a deadly global health threat. Rare Candida species are increasingly important causes of candidemia and phenotypic data, including patterns of antifungal drug resistance, is limited. There is geographic variation in the distribution of Candida species and frequency of antifungal drug resistance, which means that collecting and reporting regional data can have significant clinical value. Here, we report the first survey of species distribution, frequency of antifungal drug resistance, and phenotypic variability of Candida bloodstream isolates from an academic medical center and 5 affiliated hospitals in the Minneapolis-Saint Paul region of Minnesota, collected during an 18-month period from 2019 to 2021. We collected 288 isolates spanning 11 species from 119 patients. C. albicans was the most frequently recovered species, followed by C. glabrata and C. parapsilosis, with 10% of cases representing additional, rare species. We performed antifungal drug susceptibility for the three major drug classes and, concerningly, we identified fluconazole, micafungin and multidrug resistance rates in C. glabrata that were ~ 2 times higher than that reported in other regions of the United States. We report some of the first phenotypic data in rare non-albicans Candida species. Through analysis of serial isolates from individual patients, we identified clinically relevant within-patient differences of MIC values in multiple drug classes. Our results provide valuable clinical data relevant to antifungal stewardship efforts and highlight important areas of future research, including within-patient dynamics of infection and the mechanisms of drug resistance in rare Candida species.
Collapse
Affiliation(s)
- Nancy E. Scott
- University of Minnesota, Bioinformatics and Computational Biology Program
- University of Minnesota, Department of Microbiology and Immunology
| | - Elizabeth Wash
- University of Minnesota, Department of Microbiology and Immunology
- University of Minnesota, Molecular, Cellular, Developmental Biology and Genetics Program
| | | | - Serin E. Erayil
- University of Minnesota, Department of Medicine, Division of Infectious Diseases and International Medicine
| | - Susan E. Kline
- University of Minnesota, Department of Medicine, Division of Infectious Diseases and International Medicine
| | - Anna Selmecki
- University of Minnesota, Bioinformatics and Computational Biology Program
- University of Minnesota, Department of Microbiology and Immunology
- University of Minnesota, Molecular, Cellular, Developmental Biology and Genetics Program
| |
Collapse
|
5
|
Belay WY, Getachew M, Tegegne BA, Teffera ZH, Dagne A, Zeleke TK, Wondm SA, Abebe RB, Gedif AA, Fenta A, Yirdaw G, Tilahun A, Aschale Y. Antimicrobial resistance with a focus on antibacterial, antifungal, antimalarial, and antiviral drugs resistance, its threat, global priority pathogens, prevention, and control strategies: a review. Ther Adv Infect Dis 2025; 12:20499361251340144. [PMID: 40416942 PMCID: PMC12103682 DOI: 10.1177/20499361251340144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Antimicrobial resistance (AMR) poses a significant global health threat by diminishing the effectiveness of once-powerful antimicrobial agents, leading to higher rates of illness and death, along with escalating healthcare costs. While bacterial resistance is a primary concern, resistance is also increasing against antifungal, antiparasitic, and antiviral drugs. Many of the last-resort drugs are becoming less effective due to AMR. Projections indicate that by 2050, AMR could cause up to 10 million deaths annually, making it the leading cause of death worldwide, a situation that could result in a post-antibiotic era with substantially increased morbidity and mortality. This review aims to raise awareness about the dangers of AMR and its potential to become a silent global pandemic. It begins by examining antimicrobial drugs, followed by a discussion on AMR, focusing on resistance to antibacterial, antifungal, antimalarial, and antiviral drugs, along with its effects on health, and the economy, and prioritized global pathogens. Finally, it explores preventive measures and innovative strategies to combat AMR.
Collapse
Affiliation(s)
- Wubetu Yihunie Belay
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Samuel Agegnew Wondm
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of Clinical Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Abie Gedif
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getasew Yirdaw
- Department of Environmental Health Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Tilahun
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
6
|
Cartledge K, Short FL, Hall A, Lambert K, McDonald MJ, Lithgow T. Ethical bioprospecting and microbial assessments for sustainable solutions to the AMR crisis. IUBMB Life 2025; 77:e2931. [PMID: 39718471 PMCID: PMC11668235 DOI: 10.1002/iub.2931] [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: 08/29/2024] [Accepted: 11/01/2024] [Indexed: 12/25/2024]
Abstract
Antimicrobial resistance (AMR) has been declared one of the top 10 global public health challenges of our age by the World Health Organization, and the World Bank describes AMR as a crisis affecting the finance, health, and agriculture sectors and a major threat to the attainment of Sustainable Development Goals. But what is AMR? It is a phenotype that evolves in microbes exposed to antimicrobial molecules and causes dangerous infections. This suggests that scientists and healthcare workers should be on the frontline in the search for sustainable solutions to AMR. Yet AMR is also a societal problem to be understood by everyone. This review aims to explore the need to address the problem of AMR through a coherent, international strategy with buy-in from all sectors of society. As reviewed here, the sustainable solutions to AMR will be driven by better understanding of AMR biology but will require more than this alone to succeed. Some advances on the horizon, such as the use of bacteriophage (phage) to treat AMR infections. However, many of the new technologies and new therapeutics to address AMR require access to biodiversity, where the custodians of that biodiversity-and the traditional knowledge required to access it-are needed as key partners in the scientific, clinical, biotechnological, and international ventures that would treat the problem of AMR and ultimately prevent its further evolution. Many of these advances will be built on microbial assessments to understand the extent of AMR in our environments and bioprospecting to identify microbes that may have beneficial uses. Genuine partnerships for access to this biodiversity and sharing of benefits accrued require a consideration of ethical practice and behavior. Behavior change is needed across all sectors of culturally diverse societies so that rapid deployment of solutions can be implemented for maximum effect against the impacts of AMR.
Collapse
Affiliation(s)
| | - Francesca L. Short
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- Infection Program, Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityMelbourneAustralia
| | - Alex Hall
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- Infection Program, Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityMelbourneAustralia
| | - Karen Lambert
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- School of Curriculum, Teaching and Inclusive EducationMonash UniversityMelbourneAustralia
| | - Michael J. McDonald
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- School of Biological SciencesMonash UniversityMelbourneAustralia
| | - Trevor Lithgow
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- Infection Program, Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityMelbourneAustralia
| |
Collapse
|
7
|
Islam T, Danishuddin, Tamanna NT, Matin MN, Barai HR, Haque MA. Resistance Mechanisms of Plant Pathogenic Fungi to Fungicide, Environmental Impacts of Fungicides, and Sustainable Solutions. PLANTS (BASEL, SWITZERLAND) 2024; 13:2737. [PMID: 39409607 PMCID: PMC11478979 DOI: 10.3390/plants13192737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024]
Abstract
The significant reduction in agricultural output and the decline in product quality are two of the most glaring negative impacts caused by plant pathogenic fungi (PPF). Furthermore, contaminated food or transit might introduce mycotoxins produced by PPF directly into the food chain. Eating food tainted with mycotoxin is extremely dangerous for both human and animal health. Using fungicides is the first choice to control PPF or their toxins in food. Fungicide resistance and its effects on the environment and public health are becoming more and more of a concern, despite the fact that chemical fungicides are used to limit PPF toxicity and control growth in crops. Fungicides induce target site alteration and efflux pump activation, and mutations in PPF result in resistance. As a result, global trends are shifting away from chemically manufactured pesticides and toward managing fungal plant diseases using various biocontrol techniques, tactics, and approaches. However, surveillance programs to monitor fungicide resistance and their environmental impact are much fewer compared to bacterial antibiotic resistance surveillance programs. In this review, we discuss the PPF that contributes to disease development in plants, the fungicides used against them, factors causing the spread of PPF and the emergence of new strains, the antifungal resistance mechanisms of PPF, health, the environmental impacts of fungicides, and the use of biocontrol agents (BCAs), antimicrobial peptides (AMPs), and nanotechnologies to control PPF as a safe and eco-friendly alternative to fungicides.
Collapse
Affiliation(s)
- Tarequl Islam
- Department of Microbiology, Noakhali Science and Technology University, Noakhali 3814, Bangladesh;
| | - Danishuddin
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.N.M.)
| | - Noshin Tabassum Tamanna
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali 3814, Bangladesh;
| | - Muhammad Nurul Matin
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.N.M.)
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Hasi Rani Barai
- School of Mechanical and IT Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Md Azizul Haque
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.N.M.)
| |
Collapse
|
8
|
Nickels TJ, Gale AN, Harrington AA, Timp W, Cunningham KW. Transposon-sequencing (Tn-seq) of the Candida glabrata reference strain CBS138 reveals epigenetic plasticity, structural variation, and intrinsic mechanisms of resistance to micafungin. G3 (BETHESDA, MD.) 2024; 14:jkae173. [PMID: 39047065 PMCID: PMC11373651 DOI: 10.1093/g3journal/jkae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Candida glabrata (also called Nakaseomyces glabratus) is an opportunistic pathogen that can resist common antifungals and rapidly acquire multidrug resistance. A large amount of genetic variation exists between isolates, which complicates generalizations. Portable transposon-sequencing (Tn-seq) methods can efficiently provide genome-wide information on strain differences and genetic mechanisms. Using the Hermes transposon, the CBS138 reference strain and a commonly studied derivative termed 2001 were subjected to Tn-seq in control conditions and after exposure to varying doses of the clinical antifungal micafungin. The approach revealed large differences between these strains, including a 131-kb tandem duplication and a variety of fitness differences. Additionally, both strains exhibited up to 1,000-fold increased transposon accessibility in subtelomeric regions relative to the BG2 strain, indicative of open subtelomeric chromatin in these isolates and large epigenetic variation within the species. Unexpectedly, the Pdr1 transcription factor conferred resistance to micafungin through targets other than CDR1. Other micafungin resistance pathways were also revealed including mannosyltransferase activity and biosynthesis of the lipid precursor sphingosine, the inhibition of which by SDZ 90-215 and myriocin enhanced the potency of micafungin in vitro. These findings provide insights into the complexity of the C. glabrata species as well as strategies for improving antifungal efficacy.
Collapse
Affiliation(s)
- Timothy J Nickels
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Andrew N Gale
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kyle W Cunningham
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
9
|
Dakalbab S, Hamdy R, Holigová P, Abuzaid EJ, Abu-Qiyas A, Lashine Y, Mohammad MG, Soliman SSM. Uniqueness of Candida auris cell wall in morphogenesis, virulence, resistance, and immune evasion. Microbiol Res 2024; 286:127797. [PMID: 38851008 DOI: 10.1016/j.micres.2024.127797] [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: 04/19/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Candida auris has drawn global attention due to its alarming multidrug resistance and the emergence of pan resistant strains. C. auris poses a significant risk in nosocomial candidemia especially among immunocompromised patients. C. auris showed unique virulence characteristics associated with cell wall including cell polymorphism, adaptation, endurance on inanimate surfaces, tolerance to external conditions, and immune evasion. Notably, it possesses a distinctive cell wall composition, with an outer mannan layer shielding the inner 1,3-β glucan from immune recognition, thereby enabling immune evasion and drug resistance. This review aimed to comprehend the association between unique characteristics of C. auris's cell wall and virulence, resistance mechanisms, and immune evasion. This is particularly relevant since the fungal cell wall has no human homology, providing a potential therapeutic target. Understanding the complex interactions between the cell wall and the host immune system is essential for devising effective treatment strategies, such as the use of repurposed medications, novel therapeutic agents, and immunotherapy like monoclonal antibodies. This therapeutic targeting strategy of C. auris holds promise for effective eradication of this resilient pathogen.
Collapse
Affiliation(s)
- Salam Dakalbab
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Rania Hamdy
- Research Institute for Science and Engineering (RISE), University of Sharjah, Sharjah 27272, United Arab Emirates; Faculty of Pharmacy, Zagazig University, P.O. Box 44519, Egypt
| | | | - Eman J Abuzaid
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box, Sharjah 27272, United Arab Emirates
| | - Ameera Abu-Qiyas
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box, Sharjah 27272, United Arab Emirates
| | - Yasmina Lashine
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box, Sharjah 27272, United Arab Emirates; Faculty of Pharmacy, Zagazig University, P.O. Box 44519, Egypt
| | - Mohammad G Mohammad
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box, Sharjah 27272, United Arab Emirates; Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Sameh S M Soliman
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box, Sharjah 27272, United Arab Emirates; Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
10
|
Bisen AC, Sanap SN, Agrawal S, Biswas A, Mishra A, Verma SK, Singh V, Bhatta RS. Etiopathology, Epidemiology, Diagnosis, and Treatment of Fungal Keratitis. ACS Infect Dis 2024; 10:2356-2380. [PMID: 38847789 DOI: 10.1021/acsinfecdis.4c00203] [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] [Indexed: 07/13/2024]
Abstract
Fungal keratitis (FK) is a severe ocular condition resulting from corneal infection that is prevalent in tropical countries, particularly in developing regions of Asia and Africa. Factors like corneal lens misuse, inappropriate steroid use, and diagnostic challenges have provoked the epidemic. FK causes significant vision impairment, scarring, and ocular deformities. Accurate pathological diagnosis is crucial for effective therapeutic intervention. Topical antifungal therapy with surface healing medications proves effective in preventing fungal-borne ulcers. Managing FK requires a comprehensive understanding of fungal pathogenesis, guiding formulation strategies and preventive measures to curb global ocular blindness. This review provides in-depth insights into FK, covering etiology, epidemiology, pathogenesis, therapeutic interventions, antifungal resistance, limitations, prevention, and future perspectives on ocular surface disease management.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anjali Mishra
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sarvesh Kumar Verma
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Vaishali Singh
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
11
|
Kaden T, Alonso-Roman R, Akbarimoghaddam P, Mosig AS, Graf K, Raasch M, Hoffmann B, Figge MT, Hube B, Gresnigt MS. Modeling of intravenous caspofungin administration using an intestine-on-chip reveals altered Candida albicans microcolonies and pathogenicity. Biomaterials 2024; 307:122525. [PMID: 38489910 DOI: 10.1016/j.biomaterials.2024.122525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/21/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
Candida albicans is a commensal yeast of the human intestinal microbiota that, under predisposing conditions, can become pathogenic and cause life-threatening systemic infections (candidiasis). Fungal-host interactions during candidiasis are commonly studied using conventional 2D in vitro models, which have provided critical insights into the pathogenicity. However, microphysiological models with a higher biological complexity may be more suitable to mimic in vivo-like infection processes and antifungal drug efficacy. Therefore, a 3D intestine-on-chip model was used to investigate fungal-host interactions during the onset of invasive candidiasis and evaluate antifungal treatment under clinically relevant conditions. By combining microbiological and image-based analyses we quantified infection processes such as invasiveness and fungal translocation across the epithelial barrier. Additionally, we obtained novel insights into fungal microcolony morphology and association with the tissue. Our results demonstrate that C. albicans microcolonies induce injury to the epithelial tissue by disrupting apical cell-cell contacts and causing inflammation. Caspofungin treatment effectively reduced the fungal biomass and induced substantial alterations in microcolony morphology during infection with a wild-type strain. However, caspofungin showed limited effects after infection with an echinocandin-resistant clinical isolate. Collectively, this organ-on-chip model can be leveraged for in-depth characterization of pathogen-host interactions and alterations due to antimicrobial treatment.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH, Jena, Germany; Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Raquel Alonso-Roman
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany; Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Parastoo Akbarimoghaddam
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Applied Systems Biology, HKI-Center for Systems Biology of Infection, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany; Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany; Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | | | | | - Bianca Hoffmann
- Applied Systems Biology, HKI-Center for Systems Biology of Infection, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Marc T Figge
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Applied Systems Biology, HKI-Center for Systems Biology of Infection, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany.
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany; Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany.
| | - Mark S Gresnigt
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany.
| |
Collapse
|
12
|
Mathuria A, Ali N, Kataria N, Mani I. Drug repurposing for fungal infections. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:59-78. [PMID: 38942545 DOI: 10.1016/bs.pmbts.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
The rise of multidrug-resistant bacteria is a well-recognized threat to world health, necessitating the implementation of effective treatments. This issue has been identified as a top priority on the global agenda by the World Health Organization. Certain strains, such as Candida glabrata, Candida krusei, Candida lusitaniae, Candida auris, select cryptococcal species, and opportunistic Aspergillus or Fusarium species, have significant intrinsic resistance to numerous antifungal medicines. This inherent resistance and subsequent suboptimal clinical outcomes underscore the critical imperative for enhanced therapeutic alternatives and management protocols. The challenge of effectively treating fungal infections, compounded by the protracted timelines involved in developing novel drugs, underscores the pressing need to explore alternative therapeutic avenues. Among these, drug repurposing emerges as a particularly promising and expeditious solution, providing cost-effective solutions and safety benefits. In the fight against life-threatening resistant fungal infections, the idea of repurposing existing medications has encouraged research into both established and new compounds as a last-resort therapy. This chapter seeks to provide a comprehensive overview of contemporary antifungal drugs, as well as their key resistance mechanisms. Additionally, it seeks to provide insight into the antimicrobial properties of non-traditional drugs, thereby offering a holistic perspective on the evolving landscape of antifungal therapeutics.
Collapse
Affiliation(s)
- Anshu Mathuria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Namra Ali
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India
| | - Naina Kataria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
13
|
Nickels TJ, Gale AP, Harrington AA, Timp W, Cunningham KW. Tn-seq of the Candida glabrata reference strain CBS138 reveals epigenetic plasticity, structural variation, and intrinsic mechanisms of resistance to micafungin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592251. [PMID: 38746084 PMCID: PMC11092758 DOI: 10.1101/2024.05.02.592251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
C. glabrata is an opportunistic pathogen that can resist common antifungals and rapidly acquire multidrug resistance. A large amount of genetic variation exists between isolates, which complicates generalizations. Portable Tn-seq methods can efficiently provide genome-wide information on strain differences and genetic mechanisms. Using the Hermes transposon, the CBS138 reference strain and a commonly studied derivative termed 2001 were subjected to Tn-seq in control conditions and after exposure to varying doses of the clinical antifungal micafungin. The approach revealed large differences between these strains, including a 131 kb tandem duplication and a variety of fitness differences. Additionally, both strains exhibited up to 1000-fold increased transposon accessibility in subtelomeric regions relative to the BG2 strain, indicative of open subtelomeric chromatin in these isolates and large epigenetic variation within the species. Unexpectedly, the Pdr1 transcription factor conferred resistance to micafungin through targets other than CDR1 . Other micafungin resistance pathways were also revealed including mannosyltransferase activity and biosynthesis of the lipid precursor sphingosine, the drugging of which by SDZ 90-215 or myriocin enhanced the potency of micafungin in vitro . These findings provide insights into complexity of the C. glabrata species as well as strategies for improving antifungal efficacy. Summary Candida glabrata is an emerging pathogen with large genetic diversity and genome plasticity. The type strain CBS138 and a laboratory derivative were mutagenized with the Hermes transposon and profiled using Tn-seq. Numerous genes that regulate innate and acquired resistance to an important clinical antifungal were uncovered, including a pleiotropic drug resistance gene (PDR1) and a duplication of part of one chromosome. Compounds that target PDR1 and other genes may augment the potency of existing antifungals.
Collapse
|
14
|
Ahmady L, Gothwal M, Mukkoli MM, Bari VK. Antifungal drug resistance in Candida: a special emphasis on amphotericin B. APMIS 2024; 132:291-316. [PMID: 38465406 DOI: 10.1111/apm.13389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
Invasive fungal infections in humans caused by several Candida species, increased considerably in immunocompromised or critically ill patients, resulting in substantial morbidity and mortality. Candida albicans is the most prevalent species, although the frequency of these organisms varies greatly according to geographic region. Infections with C. albicans and non-albicans Candida species have become more common, especially in the past 20 years, as a result of aging, immunosuppressive medication use, endocrine disorders, malnourishment, extended use of medical equipment, and an increase in immunogenic diseases. Despite C. albicans being the species most frequently associated with human infections, C. glabrata, C. parapsilosis, C. tropicalis, and C. krusei also have been identified. Several antifungal drugs with different modes of action are approved for use in clinical settings to treat fungal infections. However, due to the common eukaryotic structure of humans and fungi, only a limited number of antifungal drugs are available for therapeutic use. Furthermore, drug resistance in Candida species has emerged as a result of the growing use of currently available antifungal drugs against fungal infections. Amphotericin B (AmB), a polyene class of antifungal drugs, is mainly used for the treatment of serious systemic fungal infections. AmB interacts with fungal plasma membrane ergosterol, triggering cellular ion leakage via pore formation, or extracting the ergosterol from the plasma membrane inducing cellular death. AmB resistance is primarily caused by changes in the content or structure of ergosterol. This review summarizes the antifungal drug resistance exhibited by Candida species, with a special focus on AmB.
Collapse
Affiliation(s)
- Lailema Ahmady
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | - Manisha Gothwal
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | | | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
15
|
Puumala E, Sychantha D, Lach E, Reeves S, Nabeela S, Fogal M, Nigam A, Johnson JW, Aspuru-Guzik A, Shapiro RS, Uppuluri P, Kalyaanamoorthy S, Magolan J, Whitesell L, Robbins N, Wright GD, Cowen LE. Allosteric inhibition of tRNA synthetase Gln4 by N-pyrimidinyl-β-thiophenylacrylamides exerts highly selective antifungal activity. Cell Chem Biol 2024; 31:760-775.e17. [PMID: 38402621 PMCID: PMC11031294 DOI: 10.1016/j.chembiol.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
Candida species are among the most prevalent causes of systemic fungal infections, which account for ∼1.5 million annual fatalities. Here, we build on a compound screen that identified the molecule N-pyrimidinyl-β-thiophenylacrylamide (NP-BTA), which strongly inhibits Candida albicans growth. NP-BTA was hypothesized to target C. albicans glutaminyl-tRNA synthetase, Gln4. Here, we confirmed through in vitro amino-acylation assays NP-BTA is a potent inhibitor of Gln4, and we defined how NP-BTA arrests Gln4's transferase activity using co-crystallography. This analysis also uncovered Met496 as a critical residue for the compound's species-selective target engagement and potency. Structure-activity relationship (SAR) studies demonstrated the NP-BTA scaffold is subject to oxidative and non-oxidative metabolism, making it unsuitable for systemic administration. In a mouse dermatomycosis model, however, topical application of the compound provided significant therapeutic benefit. This work expands the repertoire of antifungal protein synthesis target mechanisms and provides a path to develop Gln4 inhibitors.
Collapse
Affiliation(s)
- Emily Puumala
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David Sychantha
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Elizabeth Lach
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shawn Reeves
- Department of Chemistry, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Sunna Nabeela
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance, CA 90502, USA
| | - Meea Fogal
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - AkshatKumar Nigam
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Jarrod W Johnson
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Alán Aspuru-Guzik
- Chemical Physics Theory Group, Department of Chemistry, University of Toronto Toronto, ON M5S 3H6, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5S 2E4, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada; Department of Materials Science & Engineering, University of Toronto, Toronto, ON M5S 3E4, Canada; Vector Institute for Artificial Intelligence, Toronto, ON M5G 1M1, Canada; Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada; Acceleration Consortium, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Priya Uppuluri
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance, CA 90502, USA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | | | - Jakob Magolan
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gerard D Wright
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
16
|
Colombo SA, de Mello DMD, Morais BRM, Salvato LA, Dorella FA, Tavares GC, da Silva VMF, de Azevedo MI. CHARACTERIZATION OF THE FUNGAL MICROBIOTA IN THE NOSTRILS AND RECTUM OF AMAZONIAN MANATEES ( TRICHECHUS INUNGUIS) FROM A REHABILITATION PROGRAM IN BRAZIL. J Zoo Wildl Med 2024; 55:125-135. [PMID: 38453495 DOI: 10.1638/2022-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2023] [Indexed: 03/09/2024] Open
Abstract
The present study characterized the filamentous and yeast-like fungal microbiota of the nasal cavity and rectum of Amazonian manatees (Trichechus inunguis) undergoing rehabilitation at the Laboratory of Aquatic Mammals, National Institute of Amazonian Research, Manaus, Amazonas, and determined the antifungal susceptibility of these organisms. Nasal and rectal swabs were collected from 22 calves and three juveniles. The samples were seeded in Sabouraud agar supplemented with chloramphenicol 10%, incubated at 26°C, and observed daily for up to 7 d. The growth of different filamentous and yeast-like fungi was observed among the two anatomical sites. Filamentous fungi were categorized by macro- and microscopic characteristics of the colonies. Representatives of each group were selected for molecular identification based on the internal transcribed spacer region. Yeast identification was performed using MALDI-TOF MS and molecular analyses. Thirteen genera of filamentous fungi and six genera of yeasts were isolated and identified. The dominant filamentous species were Fusarium spp., Aspergillus spp., and Cochliobolus lunatus in the nostril samples and Aspergillus melleus in the rectal samples. Candida was the dominant genus among the identified yeasts at both anatomical sites. In the antifungal susceptibility test, 28 isolates showed resistance to fluconazole (78%), itraconazole (39%), and nystatin (42%). The knowledge of fungal microbiota composition of Amazonian manatees provides information that assists in monitoring the health status of individuals maintained in captivity, as these organisms can behave either as opportunists or as primary pathogens. Moreover, the composition and resistance of these organisms may vary among different rehabilitation institutions or different time frames of search, reinforcing the importance of constant in loco surveillance of these microorganisms. This study provides new perspectives on the fungal diversity in the microbiota of manatees and supports future studies concerning the clinical and epidemiological aspects and the impacts of these agents on the health of Amazonian manatees undergoing rehabilitation.
Collapse
Affiliation(s)
- Salene A Colombo
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Daniela M D de Mello
- Laboratory of Aquatic Mammals, National Institute of Amazonian Research-INPA, Manaus, AM 69060-001, Brazil
| | - Bruna R M Morais
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Lauranne A Salvato
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Fernanda A Dorella
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Guilherme C Tavares
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Vera M F da Silva
- Laboratory of Aquatic Mammals, National Institute of Amazonian Research-INPA, Manaus, AM 69060-001, Brazil
| | - Maria I de Azevedo
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil,
| |
Collapse
|
17
|
Jospe-Kaufman M, Ben-Zeev E, Mottola A, Dukhovny A, Berman J, Carmeli S, Fridman M. Reshaping Echinocandin Antifungal Drugs To Circumvent Glucan Synthase Point-Mutation-Mediated Resistance. Angew Chem Int Ed Engl 2024; 63:e202314728. [PMID: 38161189 DOI: 10.1002/anie.202314728] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Echinocandins are a class of antifungal drugs that inhibit the activity of the β-(1,3)-glucan synthase complex, which synthesizes fungal cell wall β-(1,3)-glucan. Echinocandin resistance is linked to mutations in the FKS gene, which encodes the catalytic subunit of the glucan synthase complex. We present a molecular-docking-based model that provides insight into how echinocandins interact with the target Fks protein: echinocandins form a ternary complex with both Fks and membrane lipids. We used reductive dehydration of alcohols to generate dehydroxylated echinocandin derivatives and evaluated their potency against a panel of Candida pathogens constructed by introducing resistance-conferring mutations in the FKS gene. We found that removing the hemiaminal alcohol, which drives significant conformational alterations in the modified echinocandins, reduced their efficacy. Conversely, eliminating the benzylic alcohol of echinocandins enhanced potency by up to two orders of magnitude, in a manner dependent upon the resistance-conferring mutation. Strains that have developed resistance to either rezafungin, the most recently clinically approved echinocandin, or its dehydroxylated derivative RZF-1, exhibit high resistance to rezafungin while demonstrating moderate resistance to RZF-1. These findings provide valuable insight for combating echinocandin resistance through chemical modifications.
Collapse
Affiliation(s)
- Moriah Jospe-Kaufman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Efrat Ben-Zeev
- The Whol Drug Discovery institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Austin Mottola
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Anna Dukhovny
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Shmuel Carmeli
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| |
Collapse
|
18
|
Domán M, Kaszab E, Laczkó L, Bali K, Makrai L, Kovács R, Majoros L, Bányai K. Genomic epidemiology of antifungal resistance in human and avian isolates of Candida albicans: a pilot study from the One Health perspective. Front Vet Sci 2024; 11:1345877. [PMID: 38435368 PMCID: PMC10904516 DOI: 10.3389/fvets.2024.1345877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Stress-induced genomic changes in Candida albicans contribute to the adaptation of this species to various environmental conditions. Variations of the genome composition of animal-origin C. albicans strains are largely unexplored and drug resistance or other selective pressures driving the evolution of these yeasts remained an intriguing question. Comparative genome analysis was carried out to uncover chromosomal aneuploidies and regions with loss of heterozygosity (LOH), two mechanisms that manage genome plasticity. We detected aneuploidy only in human isolates. Bird-derived isolates showed LOH in genes commonly associated with antifungal drug resistance similar to human isolates. Our study suggests that environmental fungicide usage might exert selective pressure on C. albicans infecting animals, thus contributing to the spread of potentially resistant strains between different hosts.
Collapse
Affiliation(s)
- Marianna Domán
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | - Eszter Kaszab
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
- One Health Institute, University of Debrecen, Debrecen, Hungary
| | - Levente Laczkó
- One Health Institute, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Conservation Biology Research Group, University of Debrecen, Debrecen, Hungary
| | - Krisztina Bali
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | | | - Renátó Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Majoros
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Krisztián Bányai
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
19
|
Zheng L, Xu Y, Wang C, Yang F, Dong Y, Guo L. Susceptibility to caspofungin is regulated by temperature and is dependent on calcineurin in Candida albicans. Microbiol Spectr 2023; 11:e0179023. [PMID: 37966204 PMCID: PMC10715083 DOI: 10.1128/spectrum.01790-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 10/06/2023] [Indexed: 11/16/2023] Open
Abstract
IMPORTANCE Echinocandins are the newest antifungal drugs and are first-line treatment option for life-threatening systemic infections. Due to lack of consensus regarding what temperature should be used when evaluating susceptibility of yeasts to echinocandins, typically either 30°C, 35°C, or 37°C is used. However, the impact of temperature on antifungal efficacy of echinocandins is unexplored. In the current study, we demonstrated that Candida albicans laboratory strain SC5314 was more susceptible to caspofungin at 37°C than at 30°C. We also found that calcineurin was required for temperature-modulated caspofungin susceptibility. Surprisingly, the altered caspofungin susceptibility was not due to differential expression of some canonical genes such as FKS, CHS, or CHT genes. The molecular mechanism of temperature-modulated caspofungin susceptibility is undetermined and deserves further investigations.
Collapse
Affiliation(s)
- Lijun Zheng
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Xu
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Chen Wang
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Feng Yang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yubo Dong
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Liangsheng Guo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Czajka KM, Venkataraman K, Brabant-Kirwan D, Santi SA, Verschoor C, Appanna VD, Singh R, Saunders DP, Tharmalingam S. Molecular Mechanisms Associated with Antifungal Resistance in Pathogenic Candida Species. Cells 2023; 12:2655. [PMID: 37998390 PMCID: PMC10670235 DOI: 10.3390/cells12222655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
Candidiasis is a highly pervasive infection posing major health risks, especially for immunocompromised populations. Pathogenic Candida species have evolved intrinsic and acquired resistance to a variety of antifungal medications. The primary goal of this literature review is to summarize the molecular mechanisms associated with antifungal resistance in Candida species. Resistance can be conferred via gain-of-function mutations in target pathway genes or their transcriptional regulators. Therefore, an overview of the known gene mutations is presented for the following antifungals: azoles (fluconazole, voriconazole, posaconazole and itraconazole), echinocandins (caspofungin, anidulafungin and micafungin), polyenes (amphotericin B and nystatin) and 5-fluorocytosine (5-FC). The following mutation hot spots were identified: (1) ergosterol biosynthesis pathway mutations (ERG11 and UPC2), resulting in azole resistance; (2) overexpression of the efflux pumps, promoting azole resistance (transcription factor genes: tac1 and mrr1; transporter genes: CDR1, CDR2, MDR1, PDR16 and SNQ2); (3) cell wall biosynthesis mutations (FKS1, FKS2 and PDR1), conferring resistance to echinocandins; (4) mutations of nucleic acid synthesis/repair genes (FCY1, FCY2 and FUR1), resulting in 5-FC resistance; and (5) biofilm production, promoting general antifungal resistance. This review also provides a summary of standardized inhibitory breakpoints obtained from international guidelines for prominent Candida species. Notably, N. glabrata, P. kudriavzevii and C. auris demonstrate fluconazole resistance.
Collapse
Affiliation(s)
- Karolina M. Czajka
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
| | - Krishnan Venkataraman
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | | | - Stacey A. Santi
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Chris Verschoor
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Vasu D. Appanna
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Ravi Singh
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Deborah P. Saunders
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| |
Collapse
|
21
|
Boyce KJ. The Microevolution of Antifungal Drug Resistance in Pathogenic Fungi. Microorganisms 2023; 11:2757. [PMID: 38004768 PMCID: PMC10673521 DOI: 10.3390/microorganisms11112757] [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: 10/11/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
The mortality rates of invasive fungal infections remain high because of the limited number of antifungal drugs available and antifungal drug resistance, which can rapidly evolve during treatment. Mutations in key resistance genes such as ERG11 were postulated to be the predominant cause of antifungal drug resistance in the clinic. However, recent advances in whole genome sequencing have revealed that there are multiple mechanisms leading to the microevolution of resistance. In many fungal species, resistance can emerge through ERG11-independent mechanisms and through the accumulation of mutations in many genes to generate a polygenic resistance phenotype. In addition, genome sequencing has revealed that full or partial aneuploidy commonly occurs in clinical or microevolved in vitro isolates to confer antifungal resistance. This review will provide an overview of the mutations known to be selected during the adaptive microevolution of antifungal drug resistance and focus on how recent advances in genome sequencing technology have enhanced our understanding of this process.
Collapse
Affiliation(s)
- Kylie J Boyce
- School of Science, RMIT University, Melbourne, VIC 3085, Australia
| |
Collapse
|
22
|
Silva LN, Ramos LS, Oliveira SSC, Magalhães LB, Cypriano J, Abreu F, Macedo AJ, Branquinha MH, Santos ALS. Development of Echinocandin Resistance in Candida haemulonii: An Emergent, Widespread, and Opportunistic Fungal Pathogen. J Fungi (Basel) 2023; 9:859. [PMID: 37623630 PMCID: PMC10455776 DOI: 10.3390/jof9080859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
Echinocandins, used for the prevention and treatment of invasive fungal infections, have led to a rise in breakthrough infections caused by resistant Candida species. Among these species, those belonging to the Candida haemulonii complex are rare multidrug-resistant (MDR) yeasts that are frequently misidentified but have emerged as significant healthcare-associated pathogens causing invasive infections. The objectives of this study were to investigate the evolutionary pathways of echinocandin resistance in C. haemulonii by identifying mutations in the FKS1 gene and evaluating the impact of resistance on fitness. After subjecting a MDR clinical isolate of C. haemulonii (named Ch4) to direct selection using increasing caspofungin concentrations, we successfully obtained an isolate (designated Ch4'r) that exhibited a high level of resistance, with MIC values exceeding 16 mg/L for all tested echinocandin drugs (caspofungin, micafungin, and anidulafungin). Sequence analysis revealed a specific mutation in the resistant Ch4'r strain, leading to an arginine-histidine amino acid substitution (R1354H), occurring at the G4061A position of the HS2 region of the FKS1 gene. Compared to the wild-type strain, Ch4'r exhibited significantly reduced growth proliferation, biofilm formation capability, and phagocytosis ratio, indicating a decrease in fitness. Transmission electron microscopy analysis revealed alterations in cell wall components, with a notable increase in cell wall thickness. The resistant strain also exhibited higher amounts (2.5-fold) of chitin, a cell wall-located molecule, compared to the wild-type strain. Furthermore, the resistant strain demonstrated attenuated virulence in the Galleria mellonella larval model. The evolved strain Ch4'r maintained its resistance profile in vivo since the treatment with either caspofungin or micafungin did not improve larval survival or reduce the fungal load. Taken together, our findings suggest that the acquisition of pan-echinocandin resistance occurred rapidly after drug exposure and was associated with a significant fitness cost in C. haemulonii. This is particularly concerning as echinocandins are often the first-line treatment option for MDR Candida species.
Collapse
Affiliation(s)
- Laura N. Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.N.S.); (L.S.R.); (S.S.C.O.); (L.B.M.); (M.H.B.)
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.N.S.); (L.S.R.); (S.S.C.O.); (L.B.M.); (M.H.B.)
| | - Simone S. C. Oliveira
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.N.S.); (L.S.R.); (S.S.C.O.); (L.B.M.); (M.H.B.)
| | - Lucas B. Magalhães
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.N.S.); (L.S.R.); (S.S.C.O.); (L.B.M.); (M.H.B.)
| | - Jefferson Cypriano
- Laboratório de Biologia Celular e Magnetotaxia & Unidade de Microscopia Multiusuário, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.C.); (F.A.)
| | - Fernanda Abreu
- Laboratório de Biologia Celular e Magnetotaxia & Unidade de Microscopia Multiusuário, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.C.); (F.A.)
| | - Alexandre J. Macedo
- Laboratório de Biofilmes e Diversidade Microbiana, Centro de Biotecnologia e Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90010-150, Brazil;
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.N.S.); (L.S.R.); (S.S.C.O.); (L.B.M.); (M.H.B.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-853, Brazil
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.N.S.); (L.S.R.); (S.S.C.O.); (L.B.M.); (M.H.B.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-853, Brazil
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
23
|
Scott NE, Edwin Erayil S, Kline SE, Selmecki A. Rapid Evolution of Multidrug Resistance in a Candida lusitaniae Infection during Micafungin Monotherapy. Antimicrob Agents Chemother 2023; 67:e0054323. [PMID: 37428075 PMCID: PMC10433866 DOI: 10.1128/aac.00543-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023] Open
Abstract
Candida (Clavispora) lusitaniae is a rare, emerging non-albicans Candida species that can cause life-threatening invasive infections, spread within hospital settings, and rapidly acquire antifungal drug resistance, including multidrug resistance. The frequency and spectrum of mutations causing antifungal drug resistance in C. lusitaniae are poorly understood. Analyses of serial clinical isolates of any Candida species are uncommon and often analyze a limited number of samples collected over months of antifungal therapy with multiple drug classes, limiting the ability to understand relationships between drug classes and specific mutations. Here, we performed comparative genomic and phenotypic analysis of 20 serial C. lusitaniae bloodstream isolates collected daily from an individual patient treated with micafungin monotherapy during a single 11-day hospital admission. We identified isolates with decreased micafungin susceptibility 4 days after initiation of antifungal therapy and a single isolate with increased cross-resistance to micafungin and fluconazole, despite no history of azole therapy in this patient. Only 14 unique single nucleotide polymorphisms (SNPs) were identified between all 20 samples, including three different FKS1 alleles among isolates with decreased micafungin susceptibility and an ERG3 missense mutation found only in the isolate with increased cross-resistance to both micafungin and fluconazole. This is the first clinical evidence of an ERG3 mutation in C. lusitaniae that occurred during echinocandin monotherapy and is associated with cross-resistance to multiple drug classes. Overall, the evolution of multidrug resistance in C. lusitaniae is rapid and can emerge during treatment with only first-line antifungal therapy.
Collapse
Affiliation(s)
- Nancy E. Scott
- University of Minnesota, Bioinformatics and Computational Biology Program, Minneapolis, Minnesota, USA
- University of Minnesota, Department of Microbiology and Immunology, Minneapolis, Minnesota, USA
| | - Serin Edwin Erayil
- University of Minnesota Medical School, Department of Medicine, Division of Infectious Diseases and International Medicine, Minneapolis, Minnesota, USA
| | - Susan E. Kline
- University of Minnesota Medical School, Department of Medicine, Division of Infectious Diseases and International Medicine, Minneapolis, Minnesota, USA
| | - Anna Selmecki
- University of Minnesota, Bioinformatics and Computational Biology Program, Minneapolis, Minnesota, USA
- University of Minnesota, Department of Microbiology and Immunology, Minneapolis, Minnesota, USA
| |
Collapse
|
24
|
Franconi I, Rizzato C, Poma N, Tavanti A, Lupetti A. Candida parapsilosis sensu stricto Antifungal Resistance Mechanisms and Associated Epidemiology. J Fungi (Basel) 2023; 9:798. [PMID: 37623569 PMCID: PMC10456088 DOI: 10.3390/jof9080798] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
Fungal diseases cause millions of deaths per year worldwide. Antifungal resistance has become a matter of great concern in public health. In recent years rates of non-albicans species have risen dramatically. Candida parapsilosis is now reported to be the second most frequent species causing candidemia in several countries in Europe, Latin America, South Africa and Asia. Rates of acquired azole resistance are reaching a worrisome threshold from multiple reports as in vitro susceptibility testing is now starting also to explore tolerance and heteroresistance to antifungal compounds. With this review, the authors seek to evaluate known antifungal resistance mechanisms and their worldwide distribution in Candida species infections with a specific focus on C. parapsilosis.
Collapse
Affiliation(s)
- Iacopo Franconi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno, 37, 56127 Pisa, Italy; (I.F.); (C.R.)
| | - Cosmeri Rizzato
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno, 37, 56127 Pisa, Italy; (I.F.); (C.R.)
| | - Noemi Poma
- Department of Biology, University of Pisa, Via San Zeno, 37, 56127 Pisa, Italy; (N.P.); (A.T.)
| | - Arianna Tavanti
- Department of Biology, University of Pisa, Via San Zeno, 37, 56127 Pisa, Italy; (N.P.); (A.T.)
| | - Antonella Lupetti
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno, 37, 56127 Pisa, Italy; (I.F.); (C.R.)
| |
Collapse
|
25
|
Zuber J, Sah SK, Mathews DH, Rustchenko E. Genome-Wide DNA Changes Acquired by Candida albicans Caspofungin-Adapted Mutants. Microorganisms 2023; 11:1870. [PMID: 37630430 PMCID: PMC10458384 DOI: 10.3390/microorganisms11081870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/22/2023] [Indexed: 08/27/2023] Open
Abstract
Drugs from the echinocandin (ECN) class are now recommended 'front-line' treatments of infections caused by a prevailing fungal pathogen, C. albicans. However, the increased use of ECNs is associated with a rising resistance to ECNs. As the acquisition of ECN resistance in C. albicans is viewed as a multistep evolution, determining factors that are associated with the decreased ECN susceptibility is of importance. We have recently identified two cohorts of genes that are either up- or downregulated in concert in order to control remodeling of cell wall, an organelle targeted by ECNs, in laboratory mutants with decreased ECN susceptibility. Here, we profiled the global DNA sequence of four of these adapted mutants in search of DNA changes that are associated with decreased ECN susceptibility. We find a limited number of 112 unique mutations representing two alternative mutational pathways. Approximately half of the mutations occurred as hotspots. Approximately half of mutations and hotspots were shared by ECN-adapted mutants despite the mutants arising as independent events and differing in some of their phenotypes, as well as in condition of chromosome 5. A total of 88 mutations are associated with 43 open reading frames (ORFs) and occurred inside of an ORF or within 1 kb of an ORF, predominantly as single-nucleotide substitution. Mutations occurred more often in the 5'-UTR than in the 3'-UTR by a 1.67:1 ratio. A total of 16 mutations mapped to eight genomic features that were not ORFs: Tca4-4 retrotransposon; Tca2-7 retrotransposon; lambda-4a long terminal repeat; mu-Ra long terminal repeat; MRS-7b Major Repeat Sequence; MRS-R Major Repeat Sequence; RB2-5a repeat sequence; and tL (CAA) leucine tRNA. Finally, eight mutations are not associated with any ORF or other genomic feature. Repeated occurrence of single-nucleotide substitutions in non-related drug-adapted mutants strongly indicates that these DNA changes are accompanying drug adaptation and could possibly influence ECN susceptibility, thus serving as factors facilitating evolution of ECN drug resistance due to classical mutations in FKS1.
Collapse
Affiliation(s)
| | | | | | - Elena Rustchenko
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Z.); (S.K.S.); (D.H.M.)
| |
Collapse
|
26
|
Lee Y, Robbins N, Cowen LE. Molecular mechanisms governing antifungal drug resistance. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:5. [PMID: 38686214 PMCID: PMC11057204 DOI: 10.1038/s44259-023-00007-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/17/2023] [Indexed: 05/02/2024]
Abstract
Fungal pathogens are a severe public health problem. The leading causative agents of systemic fungal infections include species from the Candida, Cryptococcus, and Aspergillus genera. As opportunistic pathogens, these fungi are generally harmless in healthy hosts; however, they can cause significant morbidity and mortality in immunocompromised patients. Despite the profound impact of pathogenic fungi on global human health, the current antifungal armamentarium is limited to only three major classes of drugs, all of which face complications, including host toxicity, unfavourable pharmacokinetics, or limited spectrum of activity. Further exacerbating this issue is the growing prevalence of antifungal-resistant infections and the emergence of multidrug-resistant pathogens. In this review, we discuss the diverse strategies employed by leading fungal pathogens to evolve antifungal resistance, including drug target alterations, enhanced drug efflux, and induction of cellular stress response pathways. Such mechanisms of resistance occur through diverse genetic alterations, including point mutations, aneuploidy formation, and epigenetic changes given the significant plasticity observed in many fungal genomes. Additionally, we highlight recent literature surrounding the mechanisms governing resistance in emerging multidrug-resistant pathogens including Candida auris and Candida glabrata. Advancing our knowledge of the molecular mechanisms by which fungi adapt to the challenge of antifungal exposure is imperative for designing therapeutic strategies to tackle the emerging threat of antifungal resistance.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| |
Collapse
|
27
|
Desrini S, Girardot M, Imbert C, Mustofa M, Nuryastuti T. Screening antibiofilm activity of invasive plants growing at the Slope Merapi Mountain, Central Java, against Candida albicans. BMC Complement Med Ther 2023; 23:232. [PMID: 37438777 DOI: 10.1186/s12906-023-04044-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/18/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Candida albicans causes high-mortality candidiasis. Antifungal drug resistance demands the development of virulence factor-targeting drugs, particularly antibiofilm. This study screened the effects of five invasive plants growing in Indonesia (Mimosa pudica, Lantana camara, Acacia mangium, Ageratina riparia, and Mikania micrantha) against C. albicans biofilms. Antifungal activity, antiphospholipase activity, biofilm morphology of C. albicans, and cytotoxic capacity were also evaluated. METHODS Maceration was used to extract the plants, and the most active extract inhibiting the biofilms was fractionated using liquid-liquid fractionation. Antibiofilm activity was determined by a colorimetric assay, MTT. Antifungal activity was tested using the broth microdilution method. A phospholipase assay was performed using the egg-yolk agar method. Influence on the C. albicans morphology was assessed using scanning electron microscopy (SEM). The cytotoxic effect was carried out against Vero and HeLa cell lines. RESULTS M. pudica extracts showed the most potent antifungal efficacy with minimum inhibitory concentration (MIC) of 15.62 µg/mL and 7.81 µg/mL for aerial parts and roots, respectively. At high concentrations (500 µg/mL and 250 µg/mL), ethanol extract of M. pudica aerial parts strongly inhibited the phospholipase activity. Ethyl-acetate fraction of M. pudica aerial parts demonstrated the most potent antibiofilm activity against 24 h old biofilm of C. albicans with an inhibitory concentration (53.89%) of 62.5 µg/mL showed no cytotoxicity in both Vero and HeLa cells. This fraction affected the morphology of C. albicans and contained promising compounds for inhibiting the 24 h old biofilm of C. albicans. CONCLUSIONS Invasive M. pudica plant inhibited the growth of planktonic C. albicans cells and its ethyl acetate fraction decreased the metabolic activity of C. albicans biofilms. This result demonstrates the potential of invasive M. pudica plant to reduce biofilm-associated candida infection.
Collapse
Affiliation(s)
- Sufi Desrini
- Department of Pharmacology, Faculty of Medicine, Universitas Islam Indonesia, Yogyakarta, Indonesia
- Doctoral Program of Faculty Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Laboratoire Ecologie Et Biologie Des Interactions - UMR CNRS 7267, Université de Poitiers, Poitiers, France
| | - Marion Girardot
- Laboratoire Ecologie Et Biologie Des Interactions - UMR CNRS 7267, Université de Poitiers, Poitiers, France
| | - Christine Imbert
- Laboratoire Ecologie Et Biologie Des Interactions - UMR CNRS 7267, Université de Poitiers, Poitiers, France
| | - Mustofa Mustofa
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Indonesia Biofilm Research Collaboration Center UGM-BRIN, Yogyakarta, Indonesia
| | - Titik Nuryastuti
- Indonesia Biofilm Research Collaboration Center UGM-BRIN, Yogyakarta, Indonesia.
- Department of Microbiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
28
|
Osset-Trénor P, Pascual-Ahuir A, Proft M. Fungal Drug Response and Antimicrobial Resistance. J Fungi (Basel) 2023; 9:jof9050565. [PMID: 37233275 DOI: 10.3390/jof9050565] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
Antifungal resistance is a growing concern as it poses a significant threat to public health. Fungal infections are a significant cause of morbidity and mortality, especially in immunocompromised individuals. The limited number of antifungal agents and the emergence of resistance have led to a critical need to understand the mechanisms of antifungal drug resistance. This review provides an overview of the importance of antifungal resistance, the classes of antifungal agents, and their mode of action. It highlights the molecular mechanisms of antifungal drug resistance, including alterations in drug modification, activation, and availability. In addition, the review discusses the response to drugs via the regulation of multidrug efflux systems and antifungal drug-target interactions. We emphasize the importance of understanding the molecular mechanisms of antifungal drug resistance to develop effective strategies to combat the emergence of resistance and highlight the need for continued research to identify new targets for antifungal drug development and explore alternative therapeutic options to overcome resistance. Overall, an understanding of antifungal drug resistance and its mechanisms will be indispensable for the field of antifungal drug development and clinical management of fungal infections.
Collapse
Affiliation(s)
- Paloma Osset-Trénor
- Department of Biotechnology, Instituto de Biología Molecular y Celular de Plantas IBMCP, Universidad Politécnica de Valencia, 46022 Valencia, Spain
| | - Amparo Pascual-Ahuir
- Department of Biotechnology, Instituto de Biología Molecular y Celular de Plantas IBMCP, Universidad Politécnica de Valencia, 46022 Valencia, Spain
| | - Markus Proft
- Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia IBV-CSIC, Consejo Superior de Investigaciones Científicas CSIC, 46010 Valencia, Spain
| |
Collapse
|
29
|
Kiyohara M, Miyazaki T, Okamoto M, Hirayama T, Makimura K, Chibana H, Nakada N, Ito Y, Sumiyoshi M, Ashizawa N, Takeda K, Iwanaga N, Takazono T, Izumikawa K, Yanagihara K, Kohno S, Mukae H. Evaluation of a Novel FKS1 R1354H Mutation Associated with Caspofungin Resistance in Candida auris Using the CRISPR-Cas9 System. J Fungi (Basel) 2023; 9:jof9050529. [PMID: 37233240 DOI: 10.3390/jof9050529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Outbreaks of invasive infections, with high mortality rates, caused by multidrug-resistant Candida auris have been reported worldwide. Although hotspot mutations in FKS1 are an established cause of echinocandin resistance, the actual contribution of these mutations to echinocandin resistance remains unknown. Here, we sequenced the FKS1 gene of a caspofungin-resistant clinical isolate (clade I) and identified a novel resistance mutation (G4061A inducing R1354H). We applied the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system to generate a recovered strain (H1354R) in which only this single nucleotide mutation was reverted to its wild-type sequence. We also generated mutant strains with only the R1354H mutation introduced into C. auris wild-type strains (clade I and II) and analyzed their antifungal susceptibility. Compared to their parental strains, the R1354H mutants exhibited a 4- to 16-fold increase in caspofungin minimum inhibitory concentration (MIC) while the H1354R reverted strain exhibited a 4-fold decrease in caspofungin MIC. In a mouse model of disseminated candidiasis, the in vivo therapeutic effect of caspofungin was more closely related to the FKS1 R1354H mutation and the virulence of the strain than its in vitro MIC. The CRISPR-Cas9 system could thus aid in elucidating the mechanism underlying drug resistance in C. auris.
Collapse
Affiliation(s)
- Maiko Kiyohara
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Taiga Miyazaki
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Michiyo Okamoto
- Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Tatsuro Hirayama
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
- Department of Pharmacotherapeutics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Koichi Makimura
- Teikyo University Institute of Medical Mycology, Tokyo 192-0395, Japan
| | - Hiroji Chibana
- Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Nana Nakada
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yuya Ito
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Makoto Sumiyoshi
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Nobuyuki Ashizawa
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kazuaki Takeda
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Naoki Iwanaga
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Shigeru Kohno
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
30
|
Lim HJ, Choi MJ, Byun SA, Won EJ, Park JH, Choi YJ, Choi HJ, Choi HW, Kee SJ, Kim SH, Shin MG, Lee SY, Kim MN, Shin JH. Whole-Genome Sequence Analysis of Candida glabrata Isolates from a Patient with Persistent Fungemia and Determination of the Molecular Mechanisms of Multidrug Resistance. J Fungi (Basel) 2023; 9:jof9050515. [PMID: 37233226 DOI: 10.3390/jof9050515] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Whole-genome sequencing (WGS) was used to determine the molecular mechanisms of multidrug resistance for 10 serial Candida glabrata bloodstream isolates obtained from a neutropenic patient during 82 days of amphotericin B (AMB) or echinocandin therapy. For WGS, a library was prepared and sequenced using a Nextera DNA Flex Kit (Illumina) and the MiseqDx (Illumina) instrument. All isolates harbored the same Msh2p substitution, V239L, associated with multilocus sequence type 7 and a Pdr1p substitution, L825P, that caused azole resistance. Of six isolates with increased AMB MICs (≥2 mg/L), three harboring the Erg6p A158fs mutation had AMB MICs ≥ 8 mg/L, and three harboring the Erg6p R314K, Erg3p G236D, or Erg3p F226fs mutation had AMB MICs of 2-3 mg/L. Four isolates harboring the Erg6p A158fs or R314K mutation had fluconazole MICs of 4-8 mg/L while the remaining six had fluconazole MICs ≥ 256 mg/L. Two isolates with micafungin MICs > 8 mg/L harbored Fks2p (I661_L662insF) and Fks1p (C499fs) mutations, while six isolates with micafungin MICs of 0.25-2 mg/L harbored an Fks2p K1357E substitution. Using WGS, we detected novel mechanisms of AMB and echinocandin resistance; we explored mechanisms that may explain the complex relationship between AMB and azole resistance.
Collapse
Affiliation(s)
- Ha Jin Lim
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Min Ji Choi
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Seung A Byun
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Eun Jeong Won
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Joo Heon Park
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Yong Jun Choi
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Hyun-Jung Choi
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Hyun-Woo Choi
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Soo Hyun Kim
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Myung Geun Shin
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Seung Yeob Lee
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Mi-Na Kim
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jong Hee Shin
- Department of Laboratory Medicine, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| |
Collapse
|
31
|
Hu X, Yang P, Chai C, Liu J, Sun H, Wu Y, Zhang M, Zhang M, Liu X, Yu H. Structural and mechanistic insights into fungal β-1,3-glucan synthase FKS1. Nature 2023; 616:190-198. [PMID: 36949198 PMCID: PMC10032269 DOI: 10.1038/s41586-023-05856-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 02/16/2023] [Indexed: 03/24/2023]
Abstract
The membrane-integrated synthase FKS is involved in the biosynthesis of β-1,3-glucan, the core component of the fungal cell wall1,2. FKS is the target of widely prescribed antifungal drugs, including echinocandin and ibrexafungerp3,4. Unfortunately, the mechanism of action of FKS remains enigmatic and this has hampered development of more effective medicines targeting the enzyme. Here we present the cryo-electron microscopy structures of Saccharomyces cerevisiae FKS1 and the echinocandin-resistant mutant FKS1(S643P). These structures reveal the active site of the enzyme at the membrane-cytoplasm interface and a glucan translocation path spanning the membrane bilayer. Multiple bound lipids and notable membrane distortions are observed in the FKS1 structures, suggesting active FKS1-membrane interactions. Echinocandin-resistant mutations are clustered at a region near TM5-6 and TM8 of FKS1. The structure of FKS1(S643P) reveals altered lipid arrangements in this region, suggesting a drug-resistant mechanism of the mutant enzyme. The structures, the catalytic mechanism and the molecular insights into drug-resistant mutations of FKS1 revealed in this study advance the mechanistic understanding of fungal β-1,3-glucan biosynthesis and establish a foundation for developing new antifungal drugs by targeting FKS.
Collapse
Affiliation(s)
- Xinlin Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changdong Chai
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huanhuan Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingjie Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Min Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaotian Liu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Hongjun Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Chakraborty S, Rahate K, Kumar C, Idicula-Thomas S. Expanding the therapeutic options for Candida infections using novel inhibitors of secreted aspartyl proteases. Drug Dev Res 2023; 84:96-109. [PMID: 36435973 DOI: 10.1002/ddr.22015] [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: 09/04/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/28/2022]
Abstract
For widening the therapeutic options for Candida management, the druggability of Candida proteome was systematically investigated using an innovative pipeline of high-throughput data mining algorithms, followed by in vitro validation of the observations. Through this exercise, HIV-1 protease was found to share structural similarity with secreted aspartyl protease-3 (SAP3), a virulence protein of Candida. Using the molecular fingerprint of HIV-1 protease inhibitor GRL-09510, we performed virtual screening of peptidomimetic library followed by high-precision docking and MD simulations for discovery of SAP inhibitors. Wet-lab validation of the four shortlisted peptidomimetics revealed that two molecules, when used in combination with fluconazole, could significantly reduce the dosage of fluconazole required for 50% inhibition of Candida albicans. The SAP inhibitory activity of these peptidomimetics was confirmed through SAP assays and found to be on par with pepstatin A, a known peptidomimetic inhibitor of aspartyl proteases.
Collapse
Affiliation(s)
- Shuvechha Chakraborty
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Kshitija Rahate
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Chandan Kumar
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
33
|
Branco J, Miranda IM, Rodrigues AG. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. J Fungi (Basel) 2023; 9:jof9010080. [PMID: 36675901 PMCID: PMC9862255 DOI: 10.3390/jof9010080] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Candida parapsilosis is the second most common Candida species isolated in Asia, Southern Europe, and Latin America and is often involved in invasive infections that seriously impact human health. This pathogen is part of the psilosis complex, which also includes Candida orthopsilosis and Candida metapsilosis. C. parapsilosis infections are particularly prevalent among neonates with low birth weights, individuals who are immunocompromised, and patients who require prolonged use of a central venous catheter or other indwelling devices, whose surfaces C. parapsilosis exhibits an enhanced capacity to adhere to and form biofilms. Despite this well-acknowledged prevalence, the biology of C. parapsilosis has not been as extensively explored as that of Candida albicans. In this paper, we describe the molecular mechanistic pathways of virulence in C. parapsilosis and show how they differ from those of C. albicans. We also describe the mode of action of antifungal drugs used for the treatment of Candida infections, namely, polyenes, echinocandins, and azoles, as well as the resistance mechanisms developed by C. parapsilosis to overcome them. Finally, we stress the importance of the ongoing search for species-specific features that may aid the development of effective control strategies and thus reduce the burden on patients and healthcare costs.
Collapse
Affiliation(s)
- Joana Branco
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Correspondence: ; Tel./Fax: +351-225513662
| | - Isabel M. Miranda
- Cardiovascular Research & Development Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Acácio G. Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
34
|
Husain F, Yadav A, Sah SK, Hayes JJ, Rustchenko E. Candida albicans Strains Adapted to Caspofungin Due to Aneuploidy Become Highly Tolerant under Continued Drug Pressure. Microorganisms 2022; 11:23. [PMID: 36677315 PMCID: PMC9866909 DOI: 10.3390/microorganisms11010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/17/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Candida albicans is a prevalent fungal pathogen of humans. Understanding the development of decreased susceptibility to ECN drugs of this microbe is of substantial interest, as it is viewed as an intermediate step allowing the formation of FKS1 resistance mutations. We used six previously characterized mutants that decreased caspofungin susceptibility either by acquiring aneuploidy of chromosome 5 (Ch5) or by aneuploidy-independent mechanisms. When we exposed these caspofungin-adapted mutants to caspofungin again, we obtained 60 evolved mutants with further decreases in caspofungin susceptibility, as determined with CLSI method. We show that the initial adaptation to caspofungin is coupled with the adaptation to other ECNs, such as micafungin and anidulafungin, in mutants with no ploidy change, but not in aneuploid mutants, which become more susceptible to micafungin and anidulafungin. Furthermore, we find that the initial mechanism of caspofungin adaptation determines the pattern of further adaptation as parentals with no ploidy change further adapt to all ECNs by relatively small decreases in susceptibility, whereas aneuploid parentals adapt to all ECNs, primarily by large decrease in susceptibilities. Our data suggest that either distinct or common mechanisms can govern adaptation to different ECNs.
Collapse
Affiliation(s)
| | | | | | | | - Elena Rustchenko
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
35
|
De Cesare GB, Hafez A, Stead D, Llorens C, Munro CA. Biomarkers of caspofungin resistance in Candida albicans isolates: A proteomic approach. Virulence 2022; 13:1005-1018. [PMID: 35730400 PMCID: PMC9225221 DOI: 10.1080/21505594.2022.2081291] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/17/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Candida albicans is a clinically important polymorphic fungal pathogen that causes life-threatening invasive infections in immunocompromised patients. Antifungal therapy failure is a substantial clinical problem, due to the emergence of an increasing number of drug-resistant isolates. Caspofungin is a common antifungal drug, often used as first-line therapy that inhibits cell wall β-(1,3)-glucan synthesis. In this work, the cell surface of different echinocandin-resistant C. albicans clinical isolates was compared with sensitive isolates and their responses to echinocandin treatment analyzed. Proteomic analysis detected changes in the repertoire of proteins involved in cell wall organization and maintenance, in drug-resistant strains compared to susceptible isolates and after incubation with caspofungin. Moreover, an interaction network was created from the differential expression results. Our findings suggest drug resistance may involve not only a different cell wall architecture, but also a different response to drugs.
Collapse
Affiliation(s)
- Giuseppe Buda De Cesare
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, UK
| | - Ahmed Hafez
- Biotechvana, Parc Científic Universitat de València, Valencia, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Faculty of Computer and Information, Minia University, Minia, Egypt
| | - David Stead
- Aberdeen Proteomics, Rowett Institute ofNutrition and Health, University of Aberdeen, Foresterhill, UK
| | - Carlos Llorens
- Biotechvana, Parc Científic Universitat de València, Valencia, Spain
| | - Carol A. Munro
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, UK
| |
Collapse
|
36
|
Ghasemi R, Lotfali E, Rezaei K, Madinehzad SA, Tafti MF, Aliabadi N, Kouhsari E, Fattahi M. Meyerozyma guilliermondii species complex: review of current epidemiology, antifungal resistance, and mechanisms. Braz J Microbiol 2022; 53:1761-1779. [PMID: 36306113 PMCID: PMC9679122 DOI: 10.1007/s42770-022-00813-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 06/30/2022] [Indexed: 01/13/2023] Open
Abstract
Meyerozyma guilliermondii has been accepted as a complex composed of Meyerozyma guilliermondii, Meyerozyma carpophila, and Meyerozyma caribbica. M. guilliermondii is a saprophyte detected on human mucosa and skin. It can lead to serious infections in patients with risk factors like chemotherapy, immunodeficiency, gastrointestinal or cardiovascular surgery, and oncology disorders. Most deaths related to M. guilliermondii infections occur in individuals with malignancy. In recent decades, incidence of M. guilliermondii infections is increased. Sensitivity of this microorganism to conventional antifungals (e.g., amphotericin B, fluconazole, micafungin and anidulafungin) was reduced. Prophylactic and empirical uses of these drugs are linked to elevated minimal inhibitory concentrations (MICs) of M. guilliermondii. Drug resistance has concerned many researchers across the world. They are attempting to discover appropriate solution to combat this challenge. This study reviews the most important mechanisms of resistance to antifungals developed by in M. guilliermondii species complex.
Collapse
Affiliation(s)
- Reza Ghasemi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Lotfali
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rezaei
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ataollah Madinehzad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Falah Tafti
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikta Aliabadi
- Microbiology Department Islamic, Azad University Tehran Branch, Tehran, Iran
| | - Ebrahim Kouhsari
- Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahsa Fattahi
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Jenull S, Shivarathri R, Tsymala I, Penninger P, Trinh PC, Nogueira F, Chauhan M, Singh A, Petryshyn A, Stoiber A, Chowdhary A, Chauhan N, Kuchler K. Transcriptomics and Phenotyping Define Genetic Signatures Associated with Echinocandin Resistance in Candida auris. mBio 2022; 13:e0079922. [PMID: 35968956 PMCID: PMC9426441 DOI: 10.1128/mbio.00799-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Candida auris emerged as a human fungal pathogen only during the past decade. Remarkably, C. auris displays high degrees of genomic diversity and phenotypic plasticity, with four major clades causing hospital outbreaks with high mortality and morbidity rates. C. auris can show clinical resistance to all classes of antifungal drugs, including echinocandins that are usually recommended as first-line therapies for invasive candidiasis. Here, we exploit transcriptomics coupled with phenotypic profiling to characterize a set of clinical C. auris isolates displaying pronounced echinocandin resistance (ECN-R). A hot spot mutation in the echinocandin FKS1 target gene is present in all resistant isolates. Moreover, ECN-R strains share a core signature set of 362 genes differentially expressed in ECN-R isolates. Among others, mitochondrial gene expression and genes affecting cell wall function appear to be the most prominent, with the latter correlating well with enhanced adhesive traits, increased cell wall mannan content, and altered sensitivity to cell wall stress of ECN-R isolates. Moreover, ECN-R phenotypic signatures were also linked to pathogen recognition and interaction with immune cells. Hence, transcriptomics paired with phenotyping is a suitable tool to predict resistance and fitness traits as well as treatment outcomes in pathogen populations with complex phenotypic diversity. IMPORTANCE The surge in antimicrobial drug resistance in some bacterial and fungal pathogens constitutes a significant challenge to health care facilities. The emerging human fungal pathogen Candida auris has been particularly concerning, as isolates can display pan-antifungal resistance traits against all drugs, including echinocandins. However, the mechanisms underlying this phenotypic diversity remain poorly understood. We identify transcriptomic signatures in C. auris isolates resistant to otherwise fungicidal echinocandins. We identify a set of differentially expressed genes shared by resistant strains compared to unrelated susceptible isolates. Moreover, phenotyping demonstrates that resistant strains show distinct behaviors, with implications for host-pathogen interactions. Hence, this work provides a solid basis to identify the mechanistic links between antifungal multidrug resistance and fitness costs that affect the interaction of C. auris with host immune defenses.
Collapse
Affiliation(s)
- Sabrina Jenull
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Raju Shivarathri
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Irina Tsymala
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Philipp Penninger
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Phan-Canh Trinh
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Filomena Nogueira
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
- CCRI-St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Manju Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Ashutosh Singh
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Andriy Petryshyn
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anton Stoiber
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anuradha Chowdhary
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Neeraj Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Karl Kuchler
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| |
Collapse
|
38
|
Wiederhold NP. Pharmacodynamics, Mechanisms of Action and Resistance, and Spectrum of Activity of New Antifungal Agents. J Fungi (Basel) 2022; 8:jof8080857. [PMID: 36012845 PMCID: PMC9410397 DOI: 10.3390/jof8080857] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/07/2022] [Accepted: 08/14/2022] [Indexed: 12/21/2022] Open
Abstract
Several new antifungals are currently in late-stage development, including those with novel pharmacodynamics/mechanisms of action that represent new antifungal classes (manogepix, olorofim, ATI-2307, GR-2397). Others include new agents within established classes or with mechanisms of action similar to clinically available antifungals (ibrexafungerp, rezafungin, oteseconazole, opelconazole, MAT2203) that have been modified in order to improve certain characteristics, including enhanced pharmacokinetics and greater specificity for fungal targets. Many of the antifungals under development also have activity against Candida and Aspergillus strains that have reduced susceptibility or acquired resistance to azoles and echinocandins, whereas others demonstrate activity against species that are intrinsically resistant to most clinically available antifungals. The tolerability and drug–drug interaction profiles of these new agents also appear to be promising, although the number of human subjects that have been exposed to many of these agents remains relatively small. Overall, these agents have the potential for expanding our antifungal armamentarium and improving clinical outcomes in patients with invasive mycoses.
Collapse
Affiliation(s)
- Nathan P Wiederhold
- Fungus Testing Laboratory, Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
39
|
Lee Y, Liston SD, Lee D, Robbins N, Cowen LE. Functional analysis of the Candida albicans kinome reveals Hrr25 as a regulator of antifungal susceptibility. iScience 2022; 25:104432. [PMID: 35663022 PMCID: PMC9160768 DOI: 10.1016/j.isci.2022.104432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/05/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022] Open
Abstract
Candida albicans is a leading cause of death due to systemic fungal infections. Poor patient outcomes are attributable to the limited number of antifungal classes and the increasing prevalence of drug resistance. Protein kinases have emerged as rewarding targets in the development of drugs for diverse diseases, yet kinases remain untapped in the quest for new antifungals. Here, we performed a comprehensive analysis of the C. albicans kinome to identify genes for which loss-of-function confers hypersensitivity to the two most widely deployed antifungals, echinocandins and azoles. Through this analysis, we found a role for the casein kinase 1 (CK1) homologue Hrr25 in regulating tolerance to both antifungals as well as target-mediated echinocandin resistance. Follow-up investigations established that Hrr25 regulates these responses through its interaction with the SBF transcription factor. Thus, we provide insights into the circuitry governing cellular responses to antifungals and implicate Hrr25 as a key mediator of drug resistance. Screening Candida albicans kinase mutants reveals 47 regulators of antifungal tolerance Hrr25 is important for growth and cell wall/membrane stress tolerance Hrr25 enables target-mediated echinocandin resistance Hrr25 interacts with the SBF transcription factor complex
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sean D Liston
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Dongyeob Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
40
|
Lotfali E, Erami M, Fattahi M, Nemati H, Ghasemi Z, Mahdavi E. Analysis of molecular resistance to azole and echinocandin in Candida species in patients with vulvovaginal candidiasis. Curr Med Mycol 2022; 8:1-7. [PMID: 36654793 PMCID: PMC9825790 DOI: 10.18502/cmm.8.2.10326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/29/2022] [Accepted: 03/19/2022] [Indexed: 11/15/2022] Open
Abstract
Background and Purpose Vulvovaginal candidiasis (VVC) is considered the most common mucosal infection caused by Candida species. Azoles were considered the first-line treatment for VVC or recurrent vulvovaginal candidiasis (RVVC) in both healthy and immunocompromised populations. Recently, azole-resistant isolates, especially among non-albicans Candida samples have been encountered. This study aimed to evaluate the antifungal susceptibility profile of Candida spp. isolated from VVC or RVVC patients and assess the molecular resistance mechanism of Candida spp. to azole and echinocandin. Materials and Methods Point mutation analysis was performed on the ERG11 and FKS candidate genes of azole- and caspofungin-resistant Candida albicans and Candida glabrata isolates. Real-time polymerase chain reaction was performed to gain insight into the differential expression of ERG11 mRNA. Results Variations in the amino acid D116E were observed in fluconazole- and itraconazole-resistant C. albicans strains, and changes in amino acid E517Q were observed only in fluconazole-resistant C. albicans strains. No polymorphisms were observed in the complete sequence alignment of the ERG11 gene in one azole-resistant C. glabrata isolate. The mutation triggered the changes in the amino acid serine in the reference gene FKS1 by the leucine at position 642 (S642L) of the isolates. Conclusion In patients with persistent or recurrent infection, the choice of an antifungal agent is often challenging and requires monitoring of the antifungal susceptibility of the colonizing strain. C. albicans and C. glabrata isolates can be resistant to azole and caspofungin antifungal agents without mutations in the ERG 11 and HS1 regions of the FKS1 gene.
Collapse
Affiliation(s)
- Ensieh Lotfali
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahzad Erami
- Kashan Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahsa Fattahi
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Houshang Nemati
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Ghasemi
- Razi Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Elham Mahdavi
- Department of Medical Parasitology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
41
|
Jaber QZ, Logviniuk D, Yona A, Fridman M. Echinocandins Localized to the Target-Harboring Cell Surface Are Not Degraded but Those Entering the Vacuole Are. ACS Chem Biol 2022; 17:1155-1163. [PMID: 35404573 PMCID: PMC9127807 DOI: 10.1021/acschembio.2c00060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Echinocandin antifungal drugs have a broad spectrum of activities and excellent safety profiles. These agents noncompetitively inhibit the formation of the major polysaccharide component of the fungal cell wall, a reaction catalyzed by the membrane-bound β-glucan synthase (GS) protein complex. We have developed fluorescent probes of three echinocandin drugs: caspofungin (CSF), anidulafungin (ANF), and rezafungin (RZF). Fluorescent echinocandins had the same spectrum of activities as the parent echinocandins, supporting the fact that conjugation of the dye did not alter their mode of action. Of the three echinocandins, ANF has the most potent in vitro activity. Investigation of the subcellular distribution of the fluorescent echinocandins in live Candida yeast cells revealed that despite their high structural similarity, each of the drug probes had a unique subcellular distribution pattern. Fluorescent CSF, which is the least potent of the three echinocandins, accumulated in Candida vacuoles; fluorescent ANF localized in the extracellular environment and on the yeast cell surface where the target GS resides; and fluorescent RZF was partitioned between the surface and the vacuole over time. Recovery of fluorescent CSF from Candida cells revealed substantial degradation over time; functional vacuoles were necessary for this degradation. Under the same conditions, fluorescent ANF was not degraded. This study supports the "target-oriented drug subcellular localization" principle. In the case of echinocandins, localization to the cell surface can contribute to improved potency and accumulation in vacuoles induces degradation leading to drug deactivation.
Collapse
|
42
|
Echinocandins Susceptibility Patterns of 2,787 Yeast Isolates: Importance of the Thresholds for the Detection of FKS Mutations. Antimicrob Agents Chemother 2022; 66:e0172521. [PMID: 35412354 DOI: 10.1128/aac.01725-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Since echinocandins are recommended as first line therapy for invasive candidiasis, detection of resistance, mainly due to alteration in FKS protein, is of main interest. EUCAST AFST recommends testing both MIC of anidulafungin and micafungin, and breakpoints (BPs) have been proposed to detect echinocandin-resistant isolates. We analyzed MIC distribution for all three available echinocandins of 2,787 clinical yeast isolates corresponding to 5 common and 16 rare yeast species, using the standardized EUCAST method for anidulafungin and modified for caspofungin and micafungin (AM3-MIC). In our database, 64 isolates of common pathogenic species were resistant to anidulafungin, according to the EUCAST BP, and/or to caspofungin, using our previously published threshold (AM3-MIC ≥ 0.5 mg/L). Among these 64 isolates, 50 exhibited 21 different FKS mutations. We analyzed the capacity of caspofungin AM3-MIC and anidulafungin MIC determination in detecting isolates with FKS mutation. They were always identified using caspofungin AM3-MIC and the local threshold while some isolates were misclassified using anidulafungin MIC and EUCAST threshold. However, both methods misclassified four wild-type C. glabrata as resistant. Based on a large data set from a single center, the use of AM3-MIC testing for caspofungin looks promising in identifying non-wild-type C. albicans, C. tropicalis and P. kudiravzevii isolates, but additional multicenter comparison is mandatory to conclude on the possible superiority of AM3-MIC testing compared to the EUCAST method.
Collapse
|
43
|
Logviniuk D, Jaber QZ, Dobrovetsky R, Kozer N, Ksiezopolska E, Gabaldón T, Carmeli S, Fridman M. Benzylic Dehydroxylation of Echinocandin Antifungal Drugs Restores Efficacy against Resistance Conferred by Mutated Glucan Synthase. J Am Chem Soc 2022; 144:5965-5975. [PMID: 35347986 PMCID: PMC8991007 DOI: 10.1021/jacs.2c00269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Each year, infections caused by fungal pathogens claim the lives of about 1.6 million people and affect the health of over a billion people worldwide. Among the most recently developed antifungal drugs are the echinocandins, which noncompetitively inhibit β-glucan synthase, a membrane-bound protein complex that catalyzes the formation of the main polysaccharide component of the fungal cell wall. Resistance to echinocandins is conferred by mutations in FKS genes, which encode the catalytic subunit of the β-glucan synthase complex. Here, we report that selective removal of the benzylic alcohol of the nonproteinogenic amino acid 3S,4S-dihydroxy-l-homotyrosine of the echinocandins anidulafungin and rezafungin, restored their efficacy against a large panel of echinocandin-resistant Candida strains. The dehydroxylated compounds did not significantly affect the viability of human-derived cell culture lines. An analysis of the efficacy of the dehydroxylated echinocandins against resistant Candida strains, which contain mutations in the FKS1 and/or FKS2 genes of the parental strains, identified amino acids of the Fks proteins that are likely to reside in proximity to the l-homotyrosine residue of the bound drug. This study describes the first example of a chemical modification strategy to restore the efficacy of echinocandin drugs, which have a critical place in the arsenal of antifungal drugs, against resistant fungal pathogens.
Collapse
Affiliation(s)
- Dana Logviniuk
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Qais Z Jaber
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Roman Dobrovetsky
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Noga Kozer
- The Wohl Drug Discovery institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ewa Ksiezopolska
- Barcelona Supercomputing Centre (BSC-CNS), Jordi Girona, 29, Barcelona 08034, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, Barcelona 08028, Spain
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Jordi Girona, 29, Barcelona 08034, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, Barcelona 08028, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Passeig de Lluís Companys, 23, Barcelona 08010, Spain.,Centro Investigación Biomédica En Red de Enfermedades Infecciosas, Madrid 28029, Spain
| | - Shmuel Carmeli
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
44
|
The novel antifungal agent AB-22 displays in vitro activity against hyphal growth and biofilm formation in Candida albicans and potency for treating systemic candidiasis. J Microbiol 2022; 60:438-443. [DOI: 10.1007/s12275-022-2016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 10/18/2022]
|
45
|
Perrine-Walker F. Caspofungin resistance in Candida albicans: genetic factors and synergistic compounds for combination therapies. Braz J Microbiol 2022; 53:1101-1113. [PMID: 35352319 PMCID: PMC9433586 DOI: 10.1007/s42770-022-00739-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/25/2022] [Indexed: 11/25/2022] Open
Abstract
Caspofungin and other echinocandins have been used for the treatment of human infections by the opportunistic yeast pathogen, Candida albicans. There has been an increase in infections by non-albicans Candida species such as Candida glabrata, Candida parapsilosis, Candida tropicalis, Candida krusei, and Candida auris in clinical or hospital settings. This is problematic to public health due to the increasing prevalence of echinocandin resistant species/strains. This review will present a summary on various studies that investigated the inhibitory action of caspofungin on 1,3-β-D-glucan synthesis, on cell wall structure, and biofilm formation of C. albicans. It will highlight some of the issues linked to caspofungin resistance or reduced caspofungin sensitivity in various Candida species and the potential benefits of antimicrobial peptides and other compounds in synergy with caspofungin.
Collapse
Affiliation(s)
- Francine Perrine-Walker
- Department of Biochemistry and Genetics, La Trobe Institute For Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
46
|
Espinel-Ingroff A. Commercial Methods for Antifungal Susceptibility Testing of Yeasts: Strengths and Limitations as Predictors of Resistance. J Fungi (Basel) 2022; 8:309. [PMID: 35330310 PMCID: PMC8954760 DOI: 10.3390/jof8030309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 02/01/2023] Open
Abstract
Susceptibility testing can yield variable results because it is method (commercial or reference), agent, and species dependent. Therefore, in order for results to be clinically relevant, MICs (minimal inhibitory concentrations) or MECs (minimal effective concentrations) should help in selecting the best treatment agent in the clinical setting. This is accomplished by categorical endpoints, ideally, breakpoints (BPs) and/or ECVs/ECOFFs (epidemiological cutoff values). BPs and ECVs are available by the reference methods (CLSI [Clinical and Laboratory Standards Institute] and EUCAST [European Committee on Antifungal Susceptibility Testing]) for a variety of species/agent combinations. The lack of clinical data precludes establishment of BPs for susceptibility testing by the commercial methods and ECVs have only been calculated for the Etest and SYO assays. The goal of this review is to summarize the variety of commercial methods for antifungal susceptibility testing and the potential value of Etest and SYO ECVs for detecting mutants/non-wild type (NWT) Candida isolates. Therefore, the literature search focused on publications where the commercial method, meaning MICs and ECVs, were reported for specific NWT isolates; genetic mutations have also been listed. For the Etest, the best performers recognizing the NWT were anidulafungin ECVs: 92% for the common species; 97% for C. glabrata and fluconazole ECVs, mostly for C. parapsilosis (45 NWT isolates). By the SYO, posaconazole ECVs recognized 93% of the C. albicans and 96% of the C. parapsilosis NWT isolates and micafungin ECVs 94% (mostly C. albicans and C. glabrata). Smaller sets, some with clinical data, were also listed. These are promising results for the use of both commercial methods to identify antifungal resistance (NWT isolates). However, ECVs for other species and methods need to be defined, including the C. neoformans complex and emerging species.
Collapse
|
47
|
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
|
48
|
Sharma D, Paul RA, Rudramurthy SM, Kashyap N, Bhattacharya S, Soman R, Shankarnarayan SA, Chavan D, Singh S, Das P, Kaur H, Ghosh AK, Prasad R, Sanyal K, Chakrabarti A. Impact of FKS1 Genotype on Echinocandin In Vitro Susceptibility in Candida auris and In Vivo Response in a Murine Model of Infection. Antimicrob Agents Chemother 2022; 66:e0165221. [PMID: 34780273 PMCID: PMC8765266 DOI: 10.1128/aac.01652-21] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022] Open
Abstract
Echinocandins are frontline antifungal agents in the management of invasive infections due to multidrug resistant Candida auris. The study aimed to evaluate echinocandin resistance in C. auris isolates of multicentric origin, identify the resistance mechanism, and analyze the pharmacodynamic response to caspofungin in a neutropenic mouse model of infection. A total of 199 C. auris isolates originating from 30 centers across India were tested for susceptibility to echinocandins. Isolates with reduced susceptibility were evaluated for FKS1 mutations and in vivo response to caspofungin in a murine model of disseminated candidiasis. In addition, the response to echinocandins was assessed in light of in vitro growth kinetics, chitin content; and transcript levels of chitin synthase and FKS1 genes. We report 10 resistant C. auris isolates with four FKS1 mutations: F635Y (n = 2), F635L (n = 4), S639F (n = 3), and R1354S (n = 1). Of these, F635Y and R1354S exhibited the most profound resistance in mouse model of disseminated infection. S639F and F635L mutations conferred a moderate in vivo resistance, whereas wild-type isolates exhibiting borderline MIC were susceptible in vivo. FKS1 genotype was more accurate predictor of in vivo response than the MIC of the isolates. Isolates with high basal or inducible chitin content exhibited higher in vitro MIC in FKS1 mutant compared to wild type. FKS1 mutations play a major role in clinically relevant echinocandin resistance in C. auris with differential in vivo outcomes. This study could have implications for clinical practice and, therefore, warrants further studies.
Collapse
Affiliation(s)
- Dipti Sharma
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Raees A. Paul
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | | | - Nisha Kashyap
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | | | | | | | | | - Shreya Singh
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | | | - Harsimran Kaur
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Anup K. Ghosh
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology & Integrative Sciences and Health, Amity University Haryana, Gurgaon, India
| | - Kaustuv Sanyal
- Jawaharlal Nehru Center for Advanced Scientific Research, Bengaluru, India
| | | |
Collapse
|
49
|
da Silva Dantas A, Nogueira F, Lee KK, Walker LA, Edmondson M, Brand AC, Lenardon MD, Gow NAR. Crosstalk between the calcineurin and cell wall integrity pathways prevents chitin overexpression in Candida albicans. J Cell Sci 2021; 134:jcs258889. [PMID: 34792152 PMCID: PMC8729787 DOI: 10.1242/jcs.258889] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/09/2021] [Indexed: 10/26/2022] Open
Abstract
Echinocandins such as caspofungin are frontline antifungal drugs that compromise β-1,3 glucan synthesis in the cell wall. Recent reports have shown that fungal cells can resist killing by caspofungin by upregulation of chitin synthesis, thereby sustaining cell wall integrity (CWI). When echinocandins are removed, the chitin content of cells quickly returns to basal levels, suggesting that there is a fitness cost associated with having elevated levels of chitin in the cell wall. We show here that simultaneous activation of the calcineurin and CWI pathways generates a subpopulation of Candida albicans yeast cells that have supra-normal chitin levels interspersed throughout the inner and outer cell wall, and that these cells are non-viable, perhaps due to loss of wall elasticity required for cell expansion and growth. Mutations in the Ca2+-calcineurin pathway prevented the formation of these non-viable supra-high chitin cells by negatively regulating chitin synthesis driven by the CWI pathway. The Ca2+-calcineurin pathway may therefore act as an attenuator that prevents the overproduction of chitin by coordinating both chitin upregulation and negative regulation of the CWI signaling pathway. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Alessandra da Silva Dantas
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter EX4 4QD, UK
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Filomena Nogueira
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Children's Cancer Research Institute, Labdia and Max F. Perutz Laboratories, Vienna 1090, Austria
| | - Keunsook K. Lee
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
- NGeneBio Company, 288 Digital-ro, Guro-gu, Seoul 08390, South Korea
| | - Louise A. Walker
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Matt Edmondson
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter EX4 4QD, UK
| | - Alexandra C. Brand
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter EX4 4QD, UK
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Megan D. Lenardon
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Neil A. R. Gow
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter EX4 4QD, UK
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
50
|
Mbuayama KR, Taute H, Strӧmstedt AA, Bester MJ, Gaspar ARM. Antifungal activity and mode of action of synthetic peptides derived from the tick OsDef2 defensin. J Pept Sci 2021; 28:e3383. [PMID: 34866278 DOI: 10.1002/psc.3383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/29/2023]
Abstract
Candida albicans is the principal opportunistic fungal pathogen in nosocomial settings and resistance to antifungal drugs is on the rise. Antimicrobial peptides from natural sources are promising novel therapeutics against C. albicans. OsDef2 defensin was previously found to be active against only Gram-positive bacteria, whereas derived fragments Os and its cysteine-free analogue, Os-C, are active against Gram-positive and Gram-negative bacteria at low micromolar concentrations. In this study, OsDef2-derived analogues and fragments were screened for anticandidal activity with the aim to identify peptides with antifungal activity and in so doing obtain a better understanding of the structural requirements for activity and modes of action. Os, Os-C and Os(11-22)NH2 , a Os-truncated carboxy-terminal-amidated fragment, had the most significant antifungal activities, with minimum fungicidal concentrations (MFCs) in the micromolar range (6-28 μM). C. albicans killing was rapid and occurred within 30-60 min. Further investigations showed all three peptides interacted with cell wall derived polysaccharides while both Os and Os(11-22)NH2 permeabilized fungal liposomes. Confocal laser scanning microscopy confirmed that Os-C and Os(11-22)NH2 could enter the cytosol of live cells and subsequent findings suggest that the uptake of Os and Os-C, in contrast to Os(11-22)NH2 , is energy dependent. Although Os, Os-C and Os(11-22)NH2 induced the production of reactive oxygen species (ROS), co-incubation with ascorbic acid revealed that only ROS generated by Os-C and to a lesser extent Os(11-22)NH2 resulted in cell death. Overall, Os, Os-C and Os(11-22)NH2 are promising candidacidal agents.
Collapse
Affiliation(s)
- Kabuzi R Mbuayama
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Helena Taute
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Adam A Strӧmstedt
- Pharmacognosy, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Megan J Bester
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Anabella R M Gaspar
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|