1
|
Alharthi R, Sueiro-Olivares M, Storer I, Bin Shuraym H, Scott J, Al-Shidhani R, Fortune-Grant R, Bignell E, Tabernero L, Bromley M, Zhao C, Amich J. The sulfur-related metabolic status of Aspergillus fumigatus during infection reveals cytosolic serine hydroxymethyltransferase as a promising antifungal target. Virulence 2025; 16:2449075. [PMID: 39825596 PMCID: PMC11749473 DOI: 10.1080/21505594.2024.2449075] [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: 08/12/2024] [Revised: 11/07/2024] [Accepted: 12/28/2024] [Indexed: 01/20/2025] Open
Abstract
Sulfur metabolism is an essential aspect of fungal physiology and pathogenicity. Fungal sulfur metabolism comprises anabolic and catabolic routes that are not well conserved in mammals, therefore is considered a promising source of prospective novel antifungal targets. To gain insight into Aspergillus fumigatus sulfur-related metabolism during infection, we used a NanoString custom nCounter-TagSet and compared the expression of 68 key metabolic genes in different murine models of invasive pulmonary aspergillosis, at 3 time-points, and under a variety of in vitro conditions. We identified a set of 15 genes that were consistently expressed at higher levels in vivo than in vitro, suggesting that they may be particularly relevant for intrapulmonary growth and thus constitute promising drug targets. Indeed, the role of 5 of the 15 genes has previously been empirically validated, supporting the likelihood that the remaining candidates are relevant. In addition, the analysis of gene expression dynamics at early (16 h), mid (24 h), and late (72 h) time-points uncovered potential disease initiation and progression factors. We further characterized one of the identified genes, encoding the cytosolic serine hydroxymethyltransferase ShmB, and demonstrated that it is an essential gene of A. fumigatus, also required for virulence in a murine model of established pulmonary infection. We further showed that the structure of the ligand-binding pocket of the fungal enzyme differs significantly from its human counterpart, suggesting that specific inhibitors can be designed. Therefore, in vivo transcriptomics is a powerful tool for identifying genes crucial for fungal pathogenicity that may encode promising antifungal target candidates.
Collapse
Affiliation(s)
- Reem Alharthi
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Monica Sueiro-Olivares
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Isabelle Storer
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hajer Bin Shuraym
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jennifer Scott
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Reem Al-Shidhani
- Lydia Becker Institute for Immunology and Inflammation, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Rachael Fortune-Grant
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elaine Bignell
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Lydia Tabernero
- Lydia Becker Institute for Immunology and Inflammation, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Michael Bromley
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Can Zhao
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Jorge Amich
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Mycology Reference Laboratory (Laboratorio deReferencia e Investigación en Micología LRIM), National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- CiberInfec ISCIII, CIBER en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
da Silva Rangel R, Tamiosso RT, da Silva RS, da Silva de Oliveira LS, Biscarra Bortolotto Paz MF, De Paula Martins TT, Mazaro A, de Moraes Chitolina AB, Maurente MM, de Andrade CM, Ortiz JL, Vieira IM, da Veiga ML, Baldissera MD. Rutin attenuates oxidative damage-induced renal injury in rats experimentally infected with Cryptococcus neoformans by improving antioxidant capacity and reducing fungal burden. Microb Pathog 2025; 204:107540. [PMID: 40187580 DOI: 10.1016/j.micpath.2025.107540] [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: 12/24/2024] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Oxidative stress plays a pivotal role in the pathophysiology of infectious diseases, contributing to pathogenesis and the appearance of clinical symptoms. However, the involvement of oxidative stress in renal cryptococcosis remains unknown, as do the potential protective effects of rutin. Cryptococcus neoformans is considered the main agent of cryptococcosis, a systemic life-threatening opportunistic fungal disease that affects internal organs. In 2022, the World Health Organization classified it as a critical-priority group on its Fungal Priority Pathogens List. Rutin, a flavonoid with potent antioxidant and antifungal properties, has been proposed as a protective agent. Thus, this study aimed to evaluate whether 50 mg rutin/kg body weight prevents or reduces C. neoformans var. Grubii-induced renal oxidative stress. Renal fungal burden was significantly lower in rats that were treated with rutin and experimentally infected with C. neoformans, compared to those treated with saline solution and experimentally infected. Renal reactive oxygen species (ROS) and protein carbonylation levels were significantly higher in experimentally infected rats compared to uninfected controls, whereas catalase (CAT) and glutathione S-transferase (GST) activities were significantly lower. Treatment with rutin prevented the increase in renal ROS levels and the inhibition of CAT activity elicited by C. neoformans var. Grubii. However, no significant differences were observed in lipid damage or superoxide dismutase activity. This study is the first to demonstrate that C. neoformans var. Grubii infection induces renal oxidative damage in rats by promoting oxidative stress, increasing ROS levels, and impairing antioxidant defenses. Rutin treatment restored redox status in experimental rats through mechanisms involving oxidative stress. The protective effects of rutin against C. neoformans-induced kidney damage may result from its combined ability to scavenge ROS, inhibit protein damage, and enhance the antioxidant system.
Collapse
Affiliation(s)
| | - Raquel Tusi Tamiosso
- Laboratory of Mycological Diseases, Universidade Franciscana (UFN), Santa Maria, RS, Brazil
| | - Rúbia Schallenberger da Silva
- Veterinary Clinical Analysis Laboratory (LACVET), Centro de Ciências Rurais (CCR), Hospital Veterinário Universitário (HVU), Universidade Federal de Santa Maria (UFSM), Brazil
| | | | | | | | - Alice Mazaro
- Laboratory of Mycological Diseases, Universidade Franciscana (UFN), Santa Maria, RS, Brazil
| | | | | | - Cinthia Melazzo de Andrade
- Veterinary Clinical Analysis Laboratory (LACVET), Centro de Ciências Rurais (CCR), Hospital Veterinário Universitário (HVU), Universidade Federal de Santa Maria (UFSM), Brazil
| | - Jandora Lima Ortiz
- Department of Morphology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Brazil
| | | | - Marcelo Leite da Veiga
- Department of Morphology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Brazil
| | - Matheus Dellaméa Baldissera
- Laboratory of Mycological Diseases, Universidade Franciscana (UFN), Santa Maria, RS, Brazil; Laboratory of Bioprospecting and Experimental Biology, Universidade Franciscana (UFN), Santa Maria, RS, Brazil.
| |
Collapse
|
3
|
Yang J, Zhou K, Zhou C, Khamsi PS, Voloshchuk O, Hernandez L, Kovac J, Ebrahimi A, Liu Z. Label-free rapid antimicrobial susceptibility testing with machine-learning based dynamic holographic laser speckle imaging. Biosens Bioelectron 2025; 278:117312. [PMID: 40054155 PMCID: PMC11954659 DOI: 10.1016/j.bios.2025.117312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/18/2025] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
Antimicrobial resistance (AMR) presents a significant global challenge, creating an urgent need for rapid and sensitive antimicrobial susceptibility testing (AST) methods to guide timely treatment decisions. Traditional AST techniques, such as broth microdilution, disk diffusion, and gradient diffusion assays, require extended incubation times, delaying critical therapeutic interventions. In this study, we present a dynamic holographic laser speckle imaging (DhLSI) system, coupled with machine learning algorithms, for rapid assessment of bacterial susceptibility upon antibiotic treatment. Our method operates by utilizing a reference beam to enhance the detection of weak scattering signals, capable of performing AST at bacterial concentrations as low as 103 CFU/mL, while producing results consistent with those obtained using the standard concentration of 105 CFU/mL. By employing artificial neural networks (ANN) to analyze dynamic speckle patterns, the DhLSI system can determine bacterial susceptibility within 2-3 h. The system was validated using model Gram-positive and Gram-negative bacterial strains, as well as two antibiotic treatments with different mechanisms of action. Experiments conducted on bacteria incubated on different days demonstrated consistent performance. This approach offers a rapid, label-free platform for early-stage infection diagnosis and effective antimicrobial stewardship, with the potential to be implemented in clinical settings to address AMR challenges.
Collapse
Affiliation(s)
- Jinkai Yang
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Keren Zhou
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Chen Zhou
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Pouya Soltan Khamsi
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Olena Voloshchuk
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Landon Hernandez
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Jasna Kovac
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Aida Ebrahimi
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States; Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States.
| | - Zhiwen Liu
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, United States; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, United States.
| |
Collapse
|
4
|
Kim YG, Jeon H, Boya BR, Lee JH, Lee J. Targeting biofilm formation in Candida albicans with halogenated pyrrolopyrimidine derivatives. Eur J Med Chem 2025; 290:117528. [PMID: 40121868 DOI: 10.1016/j.ejmech.2025.117528] [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: 01/13/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Growing concern over environmental contaminants, including pharmaceuticals and antifungal agents, highlights their role in promoting resistance and biofilm formation by microorganisms. Antifungal resistance, especially in drug-resistant Candida spp., poses a global threat, worsened by the widespread use of antifungal agents in both clinical applications and environmental contamination. This study investigates the antibiofilm properties of various halogenated pyrrolo pyrimidine derivatives, specifically 4-chloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (10) and 2,4-dichloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (16), against fluconazole-resistant C. albicans. Both compounds demonstrated strong biofilm inhibition, with 16 showing greater efficacy even at lower concentrations. qRT-PCR analysis revealed downregulation of key biofilm- and hyphae/germ tube-relating genes, including ALS3, HWP1, and ECE1, alongside upregulation of stress response and biofilm regulator genes such as CDR11, GST3, IFD6, UCF1, YWP1, and ZAP1, indicating complex regulatory responses to the treatments. Molecular docking analysis revealed that these compounds bind effectively to the binding cavity of the ALS3 protein, with halogen atoms playing a key role in stabilizing interaction. Compound 16 exhibited minimal cytotoxicity in Brassica rapa and Caenorhabditis elegans models, suggesting a favorable ADMET safety profile. Confocal microscopy analysis confirmed the compounds effectiveness in preventing biofilm formation when applied as biodegradable PLGA coatings on biomaterial surfaces. These findings suggest that 16 holds promise as a potent antifungal agent with reduced environmental impact, offering both efficacy and sustainability.
Collapse
Affiliation(s)
- Yong-Guy Kim
- The Institute of Clean Technology, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Hyejin Jeon
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Bharath Reddy Boya
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Jin-Hyung Lee
- The Institute of Clean Technology, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea.
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea.
| |
Collapse
|
5
|
Poonsiri T, Stransky J, Demitri N, Haas H, Cianci M, Benini S. SidF, a dual substrate N5-acetyl-N5-hydroxy-L-ornithine transacetylase involved in Aspergillus fumigatus siderophore biosynthesis. J Struct Biol X 2025; 11:100119. [PMID: 39845173 PMCID: PMC11751504 DOI: 10.1016/j.yjsbx.2024.100119] [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: 11/08/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/24/2025] Open
Abstract
Siderophore-mediated iron acquisition is essential for the virulence of Aspergillus fumigatus, a fungus causing life-threatening aspergillosis. Drugs targeting the siderophore biosynthetic pathway could help improve disease management. The transacetylases SidF and SidL generate intermediates for different siderophores in A. fumigatus. A. fumigatus has a yet unidentified transacetylase that complements SidL during iron deficiency in SidL-lacking mutants. We present the first X-ray structure of SidF, revealing a two-domain architecture with tetrameric assembly. The N-terminal domain contributes to protein solubility and oligomerization, while the C-terminal domain containing the GCN5-related N-acetyltransferase (GNAT) motif is crucial for the enzymatic activity and mediates oligomer formation. Notably, AlphaFold modelling demonstrates structural similarity between SidF and SidL. Enzymatic assays showed that SidF can utilize acetyl-CoA as a donor, previously thought to be a substrate of SidL but not SidF, and selectively uses N5-hydroxy-L-ornithine as an acceptor. This study elucidates the structure of SidF and reveals its role in siderophore biosynthesis. We propose SidF as the unknown transacetylase complementing SidL activity, highlighting its central role in A. fumigatus siderophore biosynthesis. Investigation of this uncharacterized GNAT protein enhances our understanding of fungal virulence and holds promise for its potential application in developing antifungal therapies.
Collapse
Affiliation(s)
- Thanalai Poonsiri
- Bioorganic Chemistry and Bio-Crystallography Laboratory (B2Cl) Faculty of Agricultural, Environmental and Food Sciences, Libera Università di Bolzano, Piazza Università, 1, 39100 Bolzano, Italy
| | - Jan Stransky
- Institute of Biotechnology, AS CR, Centre of Molecular Structure, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Nicola Demitri
- Elettra –Sincrotrone Trieste, S.S. 14 Km 163.5 in Area Science Park, Basovizza, Trieste I-34149, Italy
| | - Hubertus Haas
- Institute of Molecular Biology/Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Michele Cianci
- Department of Agricultural, Food and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Stefano Benini
- Bioorganic Chemistry and Bio-Crystallography Laboratory (B2Cl) Faculty of Agricultural, Environmental and Food Sciences, Libera Università di Bolzano, Piazza Università, 1, 39100 Bolzano, Italy
| |
Collapse
|
6
|
Ishii H, Okane I, Yamaoka Y. Intrinsic Natural Resistance of Various Plant Pathogens to Ipflufenoquin, a New DHODH (Dihydroorotate Dehydrogenase)-Inhibiting Fungicide, in Relation to an Unaltered Amino Acid Sequence of the Target Site. PLANT DISEASE 2025:PDIS04240844RE. [PMID: 39475586 DOI: 10.1094/pdis-04-24-0844-re] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
In recent years, increasingly stringent pesticide regulations have made the development of new chemistries challenging. Under these regulations, the new fungicide ipflufenoquin (FRAC Code 52) was first released in Japan. Its mode of action is new; it inhibits dihydroorotate dehydrogenase (DHODH), a key enzyme in the biosynthesis of pyrimidine-based nucleotides. However, because it is a single-site inhibitor, the risk of resistance developing in pathogens must be carefully considered. The risk for dual use of DHODH inhibitors in agriculture and medicine has also become a great concern because a new antifungal olorofim is under development against human pathogens now, and cross-resistance has recently been reported between ipflufenoquin and olorofim in Aspergillus fumigatus. In this study, the sensitivity to ipflufenoquin was examined in culture and in plants using "baseline" isolates, which had never been exposed to DHODH inhibitors. Isolates of Alternaria alternata, Botrytis cinerea, B. elliptica, Colletotrichum fioriniae, C. fructicola, C. nymphaeae, C. orbiculare, C. siamense, C. tropicale, C. truncatum, and Sclerotinia sclerotiorum were highly sensitive to ipflufenoquin in culture, but isolates of Coniella vitis, Corynespora cassiicola, Pseudocercospora fuligena, and Rhizoctonia solani were inherently resistant. Ipflufenoquin had low efficacy against C. cassiicola and C. vitis after inoculation of cucumber and grapevine leaves, respectively. To understand the mechanism of natural resistance, we analyzed the partial sequence of pyrE genes, which encode the DHODH enzyme, but did not find any differences in the deduced amino acids that were thought to be associated with resistance. Thus, mechanisms other than target-site mutations might be involved in the intrinsic resistance.
Collapse
Affiliation(s)
- Hideo Ishii
- University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Izumi Okane
- University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | | |
Collapse
|
7
|
Si Z, Chan-Park MB. Chemical Innovations of Antimicrobial Polymers for Combating Antimicrobial Resistance. ACS Biomater Sci Eng 2025; 11:2470-2480. [PMID: 40241236 DOI: 10.1021/acsbiomaterials.4c02147] [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: 04/18/2025]
Abstract
The global rise of antimicrobial resistance (AMR) has rendered many traditional antibiotics ineffective, leading to an urgent need for alternative therapeutic strategies. Antimicrobial polymers, with their ability to rapidly kill bacteria by disrupting or crossing membranes and/or targeting multiple microbial functions without inducing resistance, offer a promising solution. This perspective explores recent innovations in the design and synthesis of antimicrobial polymers, focusing on their chemical motifs, structural derivatives, and their applications in combating systemic and topical infections. We also highlight key challenges in translating these materials from laboratory research to clinical practice, including issues related to the high dose required, bioavailability and stability in systemic infection treatment, and ability to disperse and kill biofilms in localized infection management. By addressing these challenges, antimicrobial polymers could play a crucial role in the development of next-generation therapeutics to combat multidrug-resistant pathogens. This perspective attempts to summarize significant insights for the design and development of advanced antimicrobial polymers to overcome AMR, offering potential pathways to improve clinical outcomes in treating systemic and local infections.
Collapse
Affiliation(s)
- Zhangyong Si
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China
| | - Mary B Chan-Park
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459 Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921 Singapore
| |
Collapse
|
8
|
Wiesner-Kiełczewska A, Zagrodzki P, Paśko P. The Impact of Dietary Interventions on the Pharmacokinetics of Antifungal Drugs: A Systematic Review with Meta-analyses. Clin Pharmacokinet 2025:10.1007/s40262-025-01511-6. [PMID: 40347349 DOI: 10.1007/s40262-025-01511-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND AND OBJECTIVE Managing food-drug interactions may help to optimize the efficacy and safety of antifungal therapy. This systematic review followed Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines to evaluate how food, beverages, antacids, and mineral supplements influence the pharmacokinetic (PK) parameters or pharmacokinetic/pharmacodynamic (PK/PD) indices of 14 orally administered antifungal drugs. METHODS We considered all studies evaluating the effects of food, beverages, antacids, and mineral supplements on PK parameters and PK/PD indices of oral antifungal drugs for inclusion. We excluded in vitro, in silico, animal studies, reviews, and alcohol-related investigations. Searches were conducted in Medline (via PubMed), Embase, and Cochrane Library from database inception to June 2024. We evaluated the risk of bias using the National Institutes of Health (NIH) tool for before-after studies and the Cochrane tool for parallel and cross-over trials. We performed meta-analyses when two or more studies with comparable designs were available; otherwise, results were summarized qualitatively. RESULTS The review included 73 studies from 68 reports. Only studies investigating the effect of dietary interactions on PK parameters were found. Meta-analyses were conducted for seven antifungal drugs, while qualitative synthesis covered the remaining drugs. Open-label, cross-over studies accounted for 58% of trials, aligning with Food and Drug Administration (FDA) recommendations. A high risk of bias appeared in 33% of studies, while only 7% showed low risk. Among 11 antifungals with food-effect data, seven (64%) exhibited clinically important interactions. High positive food effects (area under the concentration-time curve (AUC) or peak serum concentration (Cmax) increased by > 45%) were seen for griseofulvin, itraconazole capsules and tablets (except rice-based meals), and posaconazole immediate-release tablets and suspension. A moderate positive impact of high-fat meals (AUC or Cmax increased in the range of 35-45%) occurred for ibrexafungerp and oteseconazole. A high negative food effect was observed on the absorption of voriconazole and itraconazole oral suspension or super bioavailable (SUBA) capsules (AUC or Cmax decreased by > 40%). Antacids strongly reduced itraconazole and ketoconazole absorption, while nutritional supplements improved posaconazole bioavailability. Acidic beverages such as Coca Cola substantially enhanced the absorption of itraconazole, ketoconazole, and posaconazole, whereas orange juice significantly reduced itraconazole bioavailability. CONCLUSION Interactions were influenced by such factors as drug physicochemical properties, type of dietary intervention, drug formulation, and patient characteristics. Although the review largely filled the existing gaps in recommendations, we judged the overall quality of evidence as low owing to outdated studies, methodological inconsistencies, and uneven data availability. Further research involving PK/PD indices is needed to link the postprandial changes in the bioavailability of antifungal drugs with their clinical efficacy. OTHER The protocol of the systematic review was registered in March 2024 in the Open Science Framework (OSF) Registries ( https://doi.org/10.17605/OSF.IO/HAVK9 ).
Collapse
Affiliation(s)
- Agnieszka Wiesner-Kiełczewska
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, św. Łazarza 16, 31-530, Kraków, Poland
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Paweł Zagrodzki
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Paweł Paśko
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
9
|
Recio-Balsells A, Ristau ER, Pacciaroni A, Nicotra V, Casero C, García M. Rational search for natural antimicrobial compounds: relevance of sesquiterpene lactones. NATURAL PRODUCTS AND BIOPROSPECTING 2025; 15:28. [PMID: 40338458 PMCID: PMC12061821 DOI: 10.1007/s13659-025-00513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025]
Abstract
Antimicrobial resistance is one of the most pressing global health challenges, as many pathogens are rapidly evolving to evade existing treatments. Despite this urgent need for new solutions, natural plant-derived compounds remain relatively underexplored in the development of antimicrobial drugs. This report highlights an innovative approach to discovering potent antimicrobial agents through bioguided fractionation of numerous plant species from the rich Argentinean flora. By systematically screening 60 species (over 177 extracts) for antimicrobial activity against representative strains of gram-positive and gram-negative bacteria, we identified promising bioactive compounds within the Asteraceae family-particularly sesquiterpene lactones from the Xanthium genus. Building on this basis, we synthesized semi-synthetic derivatives by chemically modifying plant sub-extracts, focusing on structures incorporating heteroatoms and/or heterocycles containing oxygen and nitrogen (important for the bioavailability and bioactivity that they are capable of providing). These modifications were evaluated for their potential to enhance antimicrobial efficacy against bacteria and Candida species, including resistant strains. Our findings suggest that tailoring natural metabolites from Xanthium and related Asteraceae species can significantly improve their antimicrobial properties. This strategy offers a promising pathway for the development of novel therapeutic agents to combat bacterial and fungal infections in an era of rising drug resistance.
Collapse
Affiliation(s)
- Alejandro Recio-Balsells
- Instituto Multidisciplinario de Biología Vegetal (IMBIV), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Eugenia Rodriguez Ristau
- Instituto Multidisciplinario de Biología Vegetal (IMBIV), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Adriana Pacciaroni
- Instituto Multidisciplinario de Biología Vegetal (IMBIV), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Viviana Nicotra
- Instituto Multidisciplinario de Biología Vegetal (IMBIV), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Carina Casero
- Instituto Multidisciplinario de Biología Vegetal (IMBIV), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Manuela García
- Instituto Multidisciplinario de Biología Vegetal (IMBIV), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina.
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Ciudad Universitaria, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
10
|
Inman R, Warris A, Bignell E. A novel pan-fungal screening platform for antifungal drug discovery: proof of principle study. Antimicrob Agents Chemother 2025; 69:e0132824. [PMID: 40167378 PMCID: PMC12057344 DOI: 10.1128/aac.01328-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Broad-spectrum activity is a desirable property of novel antifungal drugs, but relevant in vitro testing is complicated by differential nutritional requirements and growth dynamics of fungal pathogens. Many screens for novel drugs are initiated against individual species or genera, with hit compounds later tested for "pan-fungal" activity. Hypothesizing that an optimized pan-fungal methodology would enhance the efficiency of early-stage drug discovery, a standardized assay was developed for a selection of World Health Organization-defined critical and high-priority fungal pathogens. Instead of using the standard susceptibility testing broth RPMI, an enriched media "fungal RPMI" (fRPMI), including multiple additional fungal growth-enhancing nutrients, was utilized. To assess utility for pan-fungal growth assessments, growth in fRPMI was compared to RPMI medium for 12 fungal pathogens. Growth was significantly improved in 7/12 species in fRPMI after 24 and/or 48 hours. For our proof-of-principle study, 500 chemical fragments from the Maybridge Ro3 Fragment library were screened at concentrations of 0.1 or 1 mM against five fungal pathogens: Aspergillus fumigatus, Candida albicans, Candida auris, Cryptococcus neoformans, and Nakaseomyces glabratus. Assay quality was assessed using z-factor analysis, and hits were normalized using a standard z-score to identify outliers. All assays achieved a high-quality z-factor (≥0.5) with readings at ≤24 hours, allowing the identification of 23 compounds with antifungal activity against at least one fungal species. From these, five compounds were identified as having pan-assay interference or broadly toxic properties. In conclusion, hits identified from pan-fungal phenotypic growth-based assays demonstrate reproducibility in all fungal species tested with carefully optimized conditions and precise timing.
Collapse
Affiliation(s)
- Rebecca Inman
- Department of Biosciences, Faculty of Health and Life Sciences, Medical Research Council Center for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Adilia Warris
- Department of Biosciences, Faculty of Health and Life Sciences, Medical Research Council Center for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Elaine Bignell
- Department of Biosciences, Faculty of Health and Life Sciences, Medical Research Council Center for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| |
Collapse
|
11
|
Yarava JR, Gautam I, Jacob A, Fu R, Wang T. Proton-Detected Solid-State NMR for Deciphering Structural Polymorphism and Dynamic Heterogeneity of Cellular Carbohydrates in Pathogenic Fungi. J Am Chem Soc 2025. [PMID: 40328234 DOI: 10.1021/jacs.5c04054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Carbohydrate polymers in their cellular context display highly polymorphic structures and dynamics essential to their diverse functions, yet they are challenging to analyze biochemically. Proton-detection solid-state NMR spectroscopy offers high isotopic abundance and sensitivity, enabling the rapid and high-resolution structural characterization of biomolecules. Here, an array of 2D/3D 1H-detection solid-state NMR techniques are tailored to investigate polysaccharides in fully protonated or partially deuterated cells of three prevalent pathogenic fungi: Rhizopus delemar, Aspergillus fumigatus, and Candida albicans, representing filamentous species and yeast forms. Selective detection of acetylated carbohydrates reveals 15 forms of N-acetylglucosamine units in R. delemar chitin, which coexists with chitosan, and associates with proteins only at limited sites. This is supported by distinct order parameters and effective correlation times of their motions, analyzed through relaxation measurements and model-free analysis. Five forms of α-1,3-glucan with distinct structural origins and dynamics were identified in A. fumigatus, important for this buffering polysaccharide to perform diverse roles of supporting wall mechanics and regenerating a soft matrix under antifungal stress. Eight α-1,2-mannan side chain variants in C. albicans were resolved, highlighting the crucial role of mannan side chains in maintaining interactions with other cell wall polymers to preserve structural integrity. These methodologies provide novel insights into the functional structures of key fungal polysaccharides and create new opportunities for exploring carbohydrate biosynthesis and modifications across diverse organisms.
Collapse
Affiliation(s)
- Jayasubba Reddy Yarava
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Isha Gautam
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Anand Jacob
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Riqiang Fu
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida 32310, United States
| | - Tuo Wang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
12
|
Kamble S, Chatterjee DR, Arjun KS, Kapoor S, Jadhav M, Gupta S, Chowdhury MG, Das R, Kombe PR, Borah S, Shard A. Synthesis, characterization, and microbiological evaluation of new triazolopyrimidine-based ferrocenes as potent antimicrobial prospects. J Inorg Biochem 2025; 270:112942. [PMID: 40339268 DOI: 10.1016/j.jinorgbio.2025.112942] [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: 11/25/2024] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Antimicrobial metallodrugs have gained considerable attention for their potent inhibitory activity and clinical success, driving the development of novel metallodrug candidates. These efforts have uncovered new bioactive scaffolds and mechanisms of action. However, the global challenge of antimicrobial resistance (AMR), fueled by the genetic adaptability of microbes and resistance to nearly all antibiotic classes, highlights the urgent need for innovative antibiotics. In this study, we expand the repertoire of metallodrugs by designing, synthesizing, and biologically evaluating triazolopyrimidine-based ferrocenes as antimicrobial agents. These compounds demonstrated broad-spectrum activity against both bacterial and fungal pathogens. Advanced characterization techniques, including NMR, HRMS, FE-SEM, and SCXRD, confirmed their structural integrity and properties. Notably, the ferrocenes exhibited potent antifungal activity against Candida species, comparable to fluconazole, and were effective against Escherichia coli and Staphylococcus aureus. Our findings reveal a new class of metallodrugs with significant antimicrobial potential, offering promising avenues to combat AMR in future.
Collapse
Affiliation(s)
- Sayali Kamble
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Kakad Shivani Arjun
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Madhav Jadhav
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Shivam Gupta
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Moumita Ghosh Chowdhury
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pranav Ravindra Kombe
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Sapan Borah
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Ahmedabad, Gandhinagar, Gujarat 382355, India.
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
13
|
van de Veerdonk FL, Carvalho A, Wauters J, Chamilos G, Verweij PE. Aspergillus fumigatus biology, immunopathogenicity and drug resistance. Nat Rev Microbiol 2025:10.1038/s41579-025-01180-z. [PMID: 40316713 DOI: 10.1038/s41579-025-01180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Aspergillus fumigatus is a saprophytic fungus prevalent in the environment and capable of causing severe invasive infection in humans. This organism can use strategies such as molecule masking, immune response manipulation and gene expression alteration to evade host defences. Understanding these mechanisms is essential for developing effective diagnostics and therapies to improve patient outcomes in Aspergillus-related diseases. In this Review, we explore the biology and pathogenesis of A. fumigatus in the context of host biology and disease, highlighting virus-associated pulmonary aspergillosis, a newly identified condition that arises in patients with severe pulmonary viral infections. In the post-pandemic landscape, in which immunotherapy is gaining attention for managing severe infections, we examine the host immune responses that are critical for controlling invasive aspergillosis and how A. fumigatus circumvents these defences. Additionally, we address the emerging issue of azole resistance in A. fumigatus, emphasizing the urgent need for greater understanding in an era marked by increasing antimicrobial resistance. This Review provides timely insights necessary for developing new immunotherapeutic strategies against invasive aspergillosis.
Collapse
Affiliation(s)
- Frank L van de Veerdonk
- Department of Internal Medicine, Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
- Radboudumc/CWZ Center of Expertise in Mycology (RCEM), Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Joost Wauters
- Medical Intensive Care, University Hospitals Leuven and Department for Clinical Infectious and Inflammatory Disorders, University Leuven, Leuven, Belgium
| | - George Chamilos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- School of Medicine, University of Crete, Heraklion, Greece
| | - Paul E Verweij
- Radboudumc/CWZ Center of Expertise in Mycology (RCEM), Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Chen P, Tian W, Zeng A, Gu H, Zeng J. Regulating Intratumoral Fungi With Hydrogels: A Novel Approach to Modulating the Tumor Microbiome for Cancer Therapy. Cancer Med 2025; 14:e70900. [PMID: 40304214 PMCID: PMC12041943 DOI: 10.1002/cam4.70900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Fungi in tumors act as a double-edged sword, potentially worsening or alleviating malignancy based on the ecological balance within the tumor microenvironment (TME). Hydrogels, as innovative drug delivery systems, are poised to redefine treatment paradigms. As advanced biomaterials, they offer a versatile platform for encapsulating and releasing antifungal agents and immunomodulators, responding to the TME's unique demands. METHODS We have conducted and collated numerous relevant reviews and studies in recent years from three aspects: Hydrogels, intra-tumoral fungi, and tumor microbe microenvironment, in the hope of identifying the connections between hydrogels and intra-tumoral microbes. RESULTS This review underscores the crucial role of intra-tumoral microbes, particularly fungi, in tumorigenesis, progression, and treatment efficacy. At the same time, we concentrated on the findings of hydrogels investigations, with their remarkable adaptability to the tumor microenvironment emerge as intelligent drug delivery systems. CONCLUSIONS Hydrogels unique ability to precisely target and modulate the tumor microflora, including fungi, endows them with a significant edge in enhancing treatment efficacy. This innovative approach not only holds great promise for improving cancer therapy outcomes but also paves the way for developing novel strategies to control metastasis and prevent cancer recurrence.
Collapse
Affiliation(s)
- Ping Chen
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| | - Weiwei Tian
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| | - Huan Gu
- College of Pharmacy and FoodSouthwest Minzu UniversityChengduChina
| | - Jin Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| |
Collapse
|
15
|
Cuypers L, Aerts R, Van de Gaer O, Vinken L, Merckx R, Gerils V, Vande Velde G, Reséndiz-Sharpe A, Maertens J, Lagrou K. Doubling of triazole resistance rates in invasive aspergillosis over a 10-year period, Belgium, 1 April 2022 to 31 March 2023. Euro Surveill 2025; 30. [PMID: 40341104 DOI: 10.2807/1560-7917.es.2025.30.18.2400559] [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: 05/10/2025] Open
Abstract
BackgroundDutch national treatment guidelines for fungal infections have been adapted based on surveillance findings of triazole resistance rates >10% in Aspergillus species isolates. In Belgium, nationwide resistance data have not been collected since 2011.AimOur objective was to evaluate changes in antifungal susceptibility among Aspergillus species isolates from patients with invasive aspergillosis.MethodsLaboratories across Belgium were invited to send all clinically relevant Aspergillus species isolates from patients diagnosed with invasive aspergillosis, collected between April 2022 and March 2023, to the National Reference Centre for Mycosis at UZ Leuven for identification and antifungal susceptibility testing.ResultsOverall, 29 clinical laboratories contributed 309 isolates from 297 patients. Median patient age was 66 years (range: 6 months-96 years). Among isolates, 61% (189/309) were from male patients. At species level, Aspergillus fumigatus isolates predominated (278/309, 90%), with a 9.7% (27/278) triazole resistance rate, compared to the 4.6% rate found in 2011. Of 27 resistant isolates, successful Cyp51A sequencing of 26 showed 20 with the TR34/L98H resistance mechanism. Across the country, local A. fumigatus triazole resistance rates varied. Among provinces in the Flanders region, Antwerp had the highest resistance rate (15.4%: 10/65; p = 0.082), Flemish Brabant (6/48) also had a rate >10%, while Limburg (2/46) had the lowest rate.ConclusionsGeographical differences in A. fumigatus triazole resistance rates stress the importance of implementing broad prospective surveillance initiatives, not limited to one region or one hospital. In Belgium, triazole resistance rates have doubled over 10 years, nearly attaining the 10% threshold, warranting re-evaluation of local empirical antifungal treatment regimen decisions.
Collapse
Affiliation(s)
- Lize Cuypers
- Department of Laboratory Medicine, National Reference Centre for Mycosis, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
- These authors contributed equally to this work and share first authorship
| | - Robina Aerts
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- These authors contributed equally to this work and share first authorship
| | - Otto Van de Gaer
- Department of Laboratory Medicine, National Reference Centre for Mycosis, University Hospitals Leuven, Leuven, Belgium
| | - Lore Vinken
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| | - Rita Merckx
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| | - Veerle Gerils
- Department of Laboratory Medicine, National Reference Centre for Mycosis, University Hospitals Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI unit, KU Leuven, Leuven, Belgium
| | | | - Johan Maertens
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
- Department of Haematology, University Hospitals Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Laboratory Medicine, National Reference Centre for Mycosis, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Liberatore A, Lombardi G, Lombardo D, Lazzarotto T, Foschi C, Ambretti S. Antifungal susceptibility of a collection of Aspergillus fumigatus strains isolated from patients with invasive pulmonary aspergillosis and COVID-19 associated aspergillosis. Diagn Microbiol Infect Dis 2025; 113:116884. [PMID: 40334335 DOI: 10.1016/j.diagmicrobio.2025.116884] [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: 03/29/2025] [Revised: 04/19/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025]
Abstract
We assessed the antifungal susceptibility of 86 Aspergillus fumigatus strains by a phenotypic test. Azole sensitivity was compared to a molecular test detecting cyp51A mutations. Azole resistance was quite limited, whereas strains from COVID-19 patients showed higher amphotericin B MICs. The molecular test showed a 100 %-agreement with the phenotypic assay.
Collapse
Affiliation(s)
- Andrea Liberatore
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Giulia Lombardi
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Donatella Lombardo
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy; Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Claudio Foschi
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy; Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Simone Ambretti
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy; Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
Ni Y, Gao X. Uncovering the role of mitochondrial genome in pathogenicity and drug resistance in pathogenic fungi. Front Cell Infect Microbiol 2025; 15:1576485. [PMID: 40308969 PMCID: PMC12040666 DOI: 10.3389/fcimb.2025.1576485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Fungal infections are becoming more prevalent globally, particularly affecting immunocompromised populations, such as people living with HIV, organ transplant recipients and those on immunomodulatory therapy. Globally, approximately 6.55 million people are affected by invasive fungal infections annually, leading to serious health consequences and death. Mitochondria are membrane-bound organelles found in almost all eukaryotic cells and play an important role in cellular metabolism and energy production, including pathogenic fungi. These organelles possess their own genome, the mitochondrial genome, which is usually circular and encodes proteins essential for energy production. Variation and evolutionary adaptation within and between species' mitochondrial genomes can affect mitochondrial function, and consequently cellular energy production and metabolic activity, which may contribute to pathogenicity and drug resistance in certain fungal species. This review explores the link between the mitochondrial genome and mechanisms of fungal pathogenicity and drug resistance, with a particular focus on Cryptococcus neoformans and Candida albicans. These insights deepen our understanding of fungal biology and may provide new avenues for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Yue Ni
- College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning, China
| | - Xindi Gao
- Department of Emergency, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
18
|
Zhou H, Zhang J, Wang R, Huang J, Xin C, Song Z. The unfolded protein response is a potential therapeutic target in pathogenic fungi. FEBS J 2025. [PMID: 40227882 DOI: 10.1111/febs.70100] [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: 11/03/2024] [Revised: 01/15/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Pathogenic fungal infections cause significant morbidity and mortality, particularly in immunocompromised patients. The frequent emergence of multidrug-resistant strains challenges existing antifungal therapies, driving the need to investigate novel antifungal agents that target new molecular moieties. Pathogenic fungi are subjected to various environmental stressors, including pH, temperature, and pharmacological agents, both in natural habitats and the host body. These stressors elevate the risk of misfolded or unfolded protein production within the endoplasmic reticulum (ER) which, if not promptly mitigated, can lead to the accumulation of these proteins in the ER lumen. This accumulation triggers an ER stress response, potentially jeopardizing fungal survival. The unfolded protein response (UPR) is a critical cellular defense mechanism activated by ER stress to restore the homeostasis of protein folding. In recent years, the regulatory role of the UPR in pathogenic fungi has garnered significant attention, particularly for its involvement in fungal adaptation, regulation of virulence, and drug resistance. In this review, we comparatively analyze the UPRs of fungi and mammals and examine the potential utility of the UPR as a molecular antifungal target in pathogenic fungi. By clarifying the specificity and regulatory functions of the UPR in pathogenic fungi, we highlight new avenues for identifying potential therapeutic targets for antifungal treatments.
Collapse
Affiliation(s)
- Hao Zhou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jinping Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Rong Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ju Huang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Caiyan Xin
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
- Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, China
| |
Collapse
|
19
|
Radosevich M, Head J, Couper L, Weaver A, Camponuri S, Montoya L, Taylor JW, Remais J. Characterizing the Soil Microbial Community Associated with the Fungal Pathogen Coccidioides immitis. J Fungi (Basel) 2025; 11:309. [PMID: 40278129 PMCID: PMC12028473 DOI: 10.3390/jof11040309] [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: 02/27/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Coccidioidomycosis is a fungal disease affecting humans and other mammals caused by environmental pathogens of the genus Coccidioides. Human exposure to the pathogen occurs via inhalation of spores aerosolized from soil. Thus, understanding the ecological factors that shape the distribution of Coccidioides in soils is important for minimizing the risk of human exposure, though this task remains challenging due to the pathogen's highly variable spatial distribution. Here, we examined the associations between the soil microbial community and Coccidioides immitis' presence within the Carrizo Plain National Monument, a minimally disturbed grassland ecosystem, and the site of a longitudinal study examining the effects of rodents and their burrows on C. immitis' presence in soils. Using internal transcribed spacer 2 (ITS2) and 16S amplicon sequencing to characterize the soil fungal and bacterial communities, we found over 30 fungal species, including several other members of the Onygenales order, that co-occurred with C. immitis more frequently than would be expected by chance. Coccidioides-positive samples were significantly higher in fungal and bacterial diversity than negative samples, an association partly driven by higher Coccidioides presence within rodent burrows compared to surface soils. Soil source (i.e., rodent burrow versus surface soil) explained the largest amount of variation in bacterial and fungal community diversity and composition, with soils collected from rodent burrows having higher fungal and bacterial diversity than those collected from adjacent surface soils. While prior evidence is mixed regarding the relationship between the presence of Coccidioides and microbial diversity, we find that favorable microhabitats, such as rodent burrows, lead to a positive association between soil microbial diversity and Coccidioides presence, particularly in otherwise resource-limited natural environments.
Collapse
Affiliation(s)
- Molly Radosevich
- Environmental Health Sciences, University of California Berkeley, Berkeley, CA 94720, USA; (M.R.); (S.C.)
| | - Jennifer Head
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA;
- Institute of Global Change Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lisa Couper
- Environmental Health Sciences, University of California Berkeley, Berkeley, CA 94720, USA; (M.R.); (S.C.)
| | - Amanda Weaver
- Environmental Health Sciences, University of California Berkeley, Berkeley, CA 94720, USA; (M.R.); (S.C.)
| | - Simon Camponuri
- Environmental Health Sciences, University of California Berkeley, Berkeley, CA 94720, USA; (M.R.); (S.C.)
| | - Liliam Montoya
- Plant and Microbial Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - John W. Taylor
- Plant and Microbial Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Justin Remais
- Environmental Health Sciences, University of California Berkeley, Berkeley, CA 94720, USA; (M.R.); (S.C.)
| |
Collapse
|
20
|
He T, Li X, Flores-Vallejo RDC, Radu AM, van Dijl JM, Haslinger K. The endophytic fungus Cosmosporella sp. VM-42 from Vinca minor is a source of bioactive compounds with potent activity against drug-resistant bacteria. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100390. [PMID: 40297407 PMCID: PMC12036058 DOI: 10.1016/j.crmicr.2025.100390] [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] [Indexed: 04/30/2025] Open
Abstract
Medicinal plants serve as valuable resources for the isolation of endophytic fungi. Vinca minor is a well-known producer of important vinca alkaloids and emerges as a promising source of endophytic fungi with antibacterial potential and biosynthetic capacity. In this study, we isolated an endophytic fungus from V. minor and identified it as Cosmosporella sp. VM-42. To date, relatively little is known about this fungal genus. The ethyl acetate extract of this isolate selectively inhibited Gram-positive bacteria, such as methicillin-sensitive and methicillin-resistant Staphylococcus aureus (MSSA and MRSA). Therefore, we isolated the most abundant compound from the crude extract and identified it as nectriapyrone with MIC and MBC values ranging from 125 to 62.5 µg/mL against MSSA and MRSA strains. We further sequenced and annotated the 39.07 Mb genome of the isolate, revealing that it encodes 9842 protein-coding genes, including 415 genes for carbohydrate-active enzymes and various biosynthetic gene clusters. Our untargeted metabolomic analysis shows that the fungus produces various secondary metabolites, including cyclodepsipeptides, dimeric naphtho-γ-pyrones, and macrolactones, which are known to have antifungal and antibacterial activities. In addition, we used small-molecule epigenetic modulators to activate the expression of silent biosynthetic gene clusters to broaden the chemical profile of Cosmosporella sp. VM-42. Taken together, we provide a first systematic analysis of Cosmosporella sp. VM-42, and our results show that it is a promising source of compounds with pharmacological potential against drug resistant bacteria.
Collapse
Affiliation(s)
- Ting He
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, the Netherlands
| | - Xiao Li
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, the Netherlands
| | - Rosario del Carmen Flores-Vallejo
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9700RB, the Netherlands
| | - Ana-Maria Radu
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, the Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9700RB, the Netherlands
| | - Kristina Haslinger
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, the Netherlands
| |
Collapse
|
21
|
Desmarini D, Truong D, Sethiya P, Liu G, Bowring B, Jessen H, Dinh H, Cain AK, Thompson PE, Djordjevic JT. Synthesis of a New Purine Analogue Class with Antifungal Activity and Improved Potency against Fungal IP 3-4K. ACS Infect Dis 2025; 11:940-953. [PMID: 40164150 PMCID: PMC11997995 DOI: 10.1021/acsinfecdis.4c00975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
New antifungals are urgently needed to treat deadly fungal infections. Targeting the fungal inositol polyphosphate kinases IP3-4K (Arg1) and IP6K (Kcs1) is a promising strategy as it has been validated genetically to be crucial for fungal virulence but never pharmacologically. We now report the synthesis of DT-23, an analogue of N2-(m-trifluorobenzylamino)-N6-(p-nitrobenzylamino)purine (TNP), and demonstrate that it more potently inhibits recombinant Arg1 from the priority pathogen Cryptococcus neoformans (Cn) (IC50 = 0.6 μM) than previous analogues (IC50 = 10-30 μM). DT-23 also inhibits recombinant Kcs1 with similar potency (IC50 = 0.68 μM) and Arg1 and Kcs1 activity in vivo. Unlike previous analogues, DT-23 inhibits fungal growth (MIC50 = 15 μg/mL) and only 1.5 μg/mL synergizes with Amphotericin B to kill Cn in vitro. DT-23/Amphotericin B is also more protective against Cn infection in an insect model compared to each drug alone. Transcription profiling shows that DT-23 impacts early stages in IP synthesis and cellular functions impacted by IPK gene deletion, consistent with its targeted effect. This study establishes the first pharmacological link between inhibiting IPK activity and antifungal activity, providing tools for studying IPK function and a foundation to potentially develop a new class of antifungal drug.
Collapse
Affiliation(s)
- Desmarini Desmarini
- Centre
for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Sydney
Institute for Infectious Diseases, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Daniel Truong
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Pooja Sethiya
- Centre
for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Sydney
Institute for Infectious Diseases, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Guizhen Liu
- Institute
of Organic Chemistry, University of Freiburg, Albertstrasse 21, 79104 Freiburg i.B, Germany
- CIBSS-Centre
for Integrative Biological Signaling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg im Breisgau, Germany
| | - Bethany Bowring
- Centre
for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Sydney
Institute for Infectious Diseases, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Henning Jessen
- Institute
of Organic Chemistry, University of Freiburg, Albertstrasse 21, 79104 Freiburg i.B, Germany
- CIBSS-Centre
for Integrative Biological Signaling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg im Breisgau, Germany
| | - Hue Dinh
- ARC
Centre
of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, North Ryde, NSW 2019, Australia
| | - Amy K. Cain
- ARC
Centre
of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, North Ryde, NSW 2019, Australia
| | - Philip E. Thompson
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Julianne T. Djordjevic
- Centre
for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Sydney
Institute for Infectious Diseases, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Westmead
Hospital, Westmead, NSW 2145, Australia
| |
Collapse
|
22
|
Bastos RW, de Aguiar Peres NT, da Silva KJG, Eufrasio LG, de Carvalho DS, Cruz GS, Lucini F, Silva LS, Silva SE, Vechi HT, do Monte Alves M, Ribeiro LRF, de Souza KL, Moreira JA, de Souza JG, de Miranda IL, de Freitas GJC, Leocádio VAT, da Silva DL, Dos Santos Corrêa L, Rocha BL, Magalhães MHC, Magalhães VCR, Cenachi ARC, Moura AS, Azevedo MI, Colombo SA, de Melo Guedes GM, Parra ALC, Brasil JA, Aguilar JBA, Gonçalves KEA, Silva AGN, da Cruz GHR, Santos GV, Morio F, Castelo-Branco DSCM, Rossato L, Santos DA. Antifungal resistance in yeasts from One Health perspective: A Brazilian study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179139. [PMID: 40117748 DOI: 10.1016/j.scitotenv.2025.179139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
Environmental exposure to fungicides can induce cross-resistance in filamentous fungi, but little is known about their effects on medically important yeasts. This multicenter study investigated the resistance in Candida isolated from different sources and evaluated the influence of clinical and environmental antifungals. Different yeasts were collected from patients, hospital environments, swine, poultry and their surroundings in four different states of Brazil. A total of 571 isolates were identified, of which 82.5 % belonged to the genus Candida. Antifungal susceptibility testing revealed that C. tropicalis had the highest resistance rate (12 %) to fluconazole, followed by C. glabrata and C. albicans with resistance rates of 8 % and 6 %, respectively. Resistance to micafungin was most common in C. glabrata (15 %). For amphotericin B, we identified two non-wild-type isolates, one belonging to the C. tropicalis species and the other to C. glabrata. Most resistant isolates were recovered from patients; however, resistance was also observed in animal and environmental isolates. In addition, susceptibility to tebuconazole, an environmental azole, was evaluated and a cut-off value of 8 μg/mL was proposed to assess potential resistance. Among the isolates, 10.5 % had a high minimum inhibitory concentrations (MIC) for tebuconazole. Moreover, cross-resistance is possible to occur, as 3.6 % of Candida isolates were resistant to fluconazole and had high MIC for tebuconazole, particularly C. albicans, C. tropicalis, and C. krusei. This study demonstrates the importance of monitoring yeast resistance to antifungals and environmental fungicides in different niches, and calls for a multidisciplinary approach to understand multiple factors involved in resistance development.
Collapse
Affiliation(s)
- Rafael Wesley Bastos
- Departamento de Microbiologia e Parasitologia, Centro de Biociências, Federal University of Rio Grande do Norte, 3000 Sen. Salgado Filho Avenue, Natal, RN 59064-741, Brazil; Brazilian National Institute of Science and Technology in Human Pathogenic Fungi, Brazil
| | - Nalu Teixeira de Aguiar Peres
- Brazilian National Institute of Science and Technology in Human Pathogenic Fungi, Brazil; Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Kássia Jéssica Galdino da Silva
- Departamento de Microbiologia e Parasitologia, Centro de Biociências, Federal University of Rio Grande do Norte, 3000 Sen. Salgado Filho Avenue, Natal, RN 59064-741, Brazil
| | - Ludmila Gouveia Eufrasio
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Daniel Santana de Carvalho
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Gabriela Silva Cruz
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Fabíola Lucini
- Health Sciences Research Laboratory, Federal University of Grande Dourados, Dourados/Itahum Road, Dourados, MS 79804-970, Brazil
| | - Lorena Souza Silva
- Departamento de Microbiologia e Parasitologia, Centro de Biociências, Federal University of Rio Grande do Norte, 3000 Sen. Salgado Filho Avenue, Natal, RN 59064-741, Brazil
| | - Sthefany Emanuelle Silva
- Departamento de Microbiologia e Parasitologia, Centro de Biociências, Federal University of Rio Grande do Norte, 3000 Sen. Salgado Filho Avenue, Natal, RN 59064-741, Brazil
| | - Hareton Teixeira Vechi
- Instituto de Medicina Tropical do Rio Grande do Norte, Federal University of Rio Grande do Norte, 300, Cônego Monte, Quintas, Natal, RN 59037-170, Brazil; Escola Multicampi de Ciências Médicas, Federal University of Rio Grande do Norte, 610, Manoel Elpídio, Caicó, RN 59300-000, Brazil; Hospital of Heart, Rio Grande do Norte, 235 Coronel Auris Coelho, Natal, RN 59075-050, Brazil
| | - Manoella do Monte Alves
- Hospital Giselda Trigueiro, 110 Cônego Monte St, Natal, RN 59037-170, Brazil; Departamento de Infectologia, Universidade Federal do Rio Grande do Norte, Brazil
| | | | - Karine Lilian de Souza
- Hospital of Heart, Rio Grande do Norte, 235 Coronel Auris Coelho, Natal, RN 59075-050, Brazil
| | - José Aparecido Moreira
- Jundiaí Agricultural School, Federal University of Rio Grande do Norte, RN 160, Km 03, Macaíba, RN 59280-000, Brazil
| | - Janete Gouveia de Souza
- Jundiaí Agricultural School, Federal University of Rio Grande do Norte, RN 160, Km 03, Macaíba, RN 59280-000, Brazil
| | - Isabela Lima de Miranda
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Gustavo Jose Cota de Freitas
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Victor Augusto Teixeira Leocádio
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Danielle Leticia da Silva
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Lislaina Dos Santos Corrêa
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Bianca Letícia Rocha
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Martha Helena Chaves Magalhães
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil
| | - Vanessa Caroline Randi Magalhães
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil; Hospital Eduardo de Menezes, Minas Gerais, 2213 Dr. Cristiano Rezende St, Belo Horizonte, MG 30622-020, Brazil
| | - Adriana Regina Campolina Cenachi
- Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil; Hospital Eduardo de Menezes, Minas Gerais, 2213 Dr. Cristiano Rezende St, Belo Horizonte, MG 30622-020, Brazil
| | - Alexandre Sampaio Moura
- Hospital Eduardo de Menezes, Minas Gerais, 2213 Dr. Cristiano Rezende St, Belo Horizonte, MG 30622-020, Brazil
| | - Maria Isabel Azevedo
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, 6627 Antônio Carlos Avenue, Belo-Horizonte, MG 31270-901, Brazil
| | - Salene Angelini Colombo
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, 6627 Antônio Carlos Avenue, Belo-Horizonte, MG 31270-901, Brazil
| | - Glaucia Morgana de Melo Guedes
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Aura Lucia Chacón Parra
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Jaiane Alves Brasil
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Jorge Bryan Andino Aguilar
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Kézia Emily Araújo Gonçalves
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Antônio Gabriel Nascimento Silva
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | | | - Gabriel Vichoski Santos
- Health Sciences Research Laboratory, Federal University of Grande Dourados, Dourados/Itahum Road, Dourados, MS 79804-970, Brazil
| | - Florent Morio
- University of Nantes, France, Department of Parasitology and Medical Mycology, 22 Boulevard Bénoni-Goullin, 44200 Nantes, France
| | - Débora S C M Castelo-Branco
- Department of Pathology and Forensic Medicine, Center of Biomedicine, Federal University of Ceará, 1315 Coronel Nunes de Melo St, Fortaleza, CE 60430-275, Brazil
| | - Luana Rossato
- Health Sciences Research Laboratory, Federal University of Grande Dourados, Dourados/Itahum Road, Dourados, MS 79804-970, Brazil
| | - Daniel Assis Santos
- Brazilian National Institute of Science and Technology in Human Pathogenic Fungi, Brazil; Department of Microbiology, Federal University of Minas Gerais, 6627 Pres. Antônio Carlos Avenue, Belo Horizonte, MG 31275-013, Brazil.
| |
Collapse
|
23
|
da Silva KJG, Lucini F, Dos Santos RAC, Santos DA, Meis JF, Melhem MDSC, Peres NTDA, Bastos RW, Rossato L. How does antifungal resistance vary in Candida (Candidozyma) auris and its clades? Quantitative and qualitative analyses and their clinical implications. Clin Microbiol Infect 2025:S1198-743X(25)00163-6. [PMID: 40216246 DOI: 10.1016/j.cmi.2025.04.003] [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: 12/18/2024] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Candida (Candidozyma) auris is a multidrug-resistant yeast that emerged as a significant health care-associated pathogen. It is classified as an urgent threat to public health due to the high resistance to available antifungal agents. Globally six distinct clades of C. auris have been identified with varying antifungal susceptibility patterns and geographical distributions. OBJECTIVES The aim of this review is to investigate the (published) antifungal susceptibility profiles of different C. auris clades to identify those with a higher prevalence of resistance. SOURCES A comprehensive literature review was conducted using PubMed, SciELO, Google Scholar, and MEDLINE databases to collect data on MIC distributions and clade designations of C. auris strains. CONTENT A total of 1031 C. auris strains were included. Clades I and III, which are closely related phylogenetically, displayed the highest resistance rates, particularly to fluconazole, with 94% and 96% of isolates, respectively. Clade IV also exhibited resistance to both azoles and echinocandins. In contrast, clades II, V, and VI had lower resistance rates, with clade VI being entirely susceptible to fluconazole. Anidulafungin demonstrated the greatest efficacy across all clades, with resistance rates ranging from 0% to 3.67%. Furthermore, clades V and VI showed complete susceptibility to all antifungal agents evaluated. IMPLICATIONS This study highlights significant variations in antifungal resistance profiles across the six C. auris clades. Clades I, III, and IV stand out because of their multidrug resistance, particularly to fluconazole and amphotericin B, posing serious challenges for treatment. Continuous global surveillance and tailored management strategies are essential for controlling C. auris infections, especially in highly resistant clades. Enhanced diagnostic capabilities and further genomic studies are critical to understanding the evolving nature of resistance in this emerging pathogen and improving therapeutic outcomes. Clade-specific antifungal resistance in C. auris requires monitoring to optimize therapy selection during outbreaks.
Collapse
Affiliation(s)
| | - Fabíola Lucini
- Health Sciences Research Laboratory, Universidade Federal da Grande Dourados (UFGD), Dourados, Mato Grosso do Sul, Brazil
| | - Renato Augusto Corrêa Dos Santos
- Computational, Evolutionary and Systems Biology Laboratory, Center for Nuclear Energy in Agriculture, University of São Paulo (USP), Piracicaba, São Paulo, Brazil
| | - Daniel Assis Santos
- Microbiology Department, Institut of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil; Brazilian National Institute of Science and Technology in Human Pathogenic Fungi, Brazil
| | - Jacques F Meis
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Excellence Center for Medical Mycology (ECMM), University of Cologne, 50923 Cologne, Germany; Centre of Expertise in Mycology Radboudumc/CWZ Nijmegen, The Netherlands
| | | | - Nalu Teixeira de Aguiar Peres
- Microbiology Department, Institut of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Rafael Wesley Bastos
- Centro de Biociências, Universidade Federal do Rio grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil; Brazilian National Institute of Science and Technology in Human Pathogenic Fungi, Brazil.
| | - Luana Rossato
- Health Sciences Research Laboratory, Universidade Federal da Grande Dourados (UFGD), Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
24
|
Wu Q, Cen F, Xie Y, Ning X, Wang J, Lin Z, Huang J. Nanoparticle-based antifungal therapies innovations mechanisms and future prospects. PeerJ 2025; 13:e19199. [PMID: 40226540 PMCID: PMC11988106 DOI: 10.7717/peerj.19199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Fungal infections present an increasing global health challenge, with a substantial annual mortality rate of 1.6 million deaths each year in certain situations. The emergence of antifungal resistance has further complicated treatment strategies, underscoring the urgent need for novel therapeutic approaches. This review explores recent advances in nanoparticle-based therapies targeting fungal infections, emphasizing their unique potential to enhance drug solubility, bioavailability, and targeted delivery. Nanoparticles offer the ability to penetrate biological barriers, improve drug stability, and act as direct antifungal agents by disrupting fungal cell walls and generating reactive oxygen species. Despite their promising applications, challenges such as potential toxicity, scalability of production, and the need for controlled drug release remain. Future research should focus on optimizing nanoparticle properties, evaluating long-term safety profiles, developing environmentally sustainable synthesis methods, and exploring synergistic approaches with existing antifungal drugs. Nanotechnology offers a transformative opportunity in the management of fungal diseases, paving the way for more effective and targeted treatments.
Collapse
Affiliation(s)
- Qinglin Wu
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Fulan Cen
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Ying Xie
- Graduate School of Public Administration, Seoul National University, Seoul, Republic of South Korea
| | - Xianjia Ning
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jinghua Wang
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Zhenghao Lin
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jia Huang
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
25
|
Friberg LIM, Kavanagh A, Amado M, Lowe G, Zuegg J, Novais de Paula M, Onawole A, Floyd HEE, Kravchenko AN, Guan D, Elliott AG, Kuchar J, Mohr F, Luzina O, Salakhutdinov N, Volcho K, Farrukh HSUB, Kalsoom U, Saleem RSZ, Blanchfield J, Blaskovich MAT, Hansford KA. Organoselenium compounds as an enriched source for the discovery of new antimicrobial agents. RSC Med Chem 2025:d4md00677a. [PMID: 40352676 PMCID: PMC12063247 DOI: 10.1039/d4md00677a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/28/2025] [Indexed: 05/14/2025] Open
Abstract
The urgent need for novel antifungal drugs is underscored by the limited number of antifungal agents in clinical development and the global spread of fungal resistance. This study highlights the potential of organoselenium compounds as a new source of scaffolds with promising antifungal activity against a variety of fungal strains. Analysis of over 300 000 compounds screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD) for antibacterial and antifungal activity identified 233 organoselenium derivatives. A remarkable 33% (77) of these exhibited antifungal activity against two representative strains of Candida and Cryptococcus, in contrast to only 2% of the non-selenium-containing organic compounds tested by CO-ADD. Of these compounds that displayed antifungal activity (MIC < 16 μg mL-1), 87% did not exhibit cytotoxicity against mammalian cell lines or haemolytic properties at similar concentrations, compared to only 35% of the organic 'hits'. A subset of these 69 compounds was further evaluated against three clinically relevant fungal strains that often exhibit resistance to current antifungals (Candida auris, MDR Candida auris, and Cryptococcus deuterogattii), and generally retained good activity. Lastly, we compared a handful of matched sulfur and selenium compounds which further highlighted the beneficial impact of this bioisosteric conversion on antifungal activity.
Collapse
Affiliation(s)
- Louise I M Friberg
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Angela Kavanagh
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Maite Amado
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Gabrielle Lowe
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Johannes Zuegg
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Michelle Novais de Paula
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Abdulmujeeb Onawole
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Holly E E Floyd
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Angelina N Kravchenko
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Moscow 119991 Russia
| | - Davy Guan
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Alysha G Elliott
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Julia Kuchar
- Inorganic Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal 42119 Wuppertal Germany
| | - Fabian Mohr
- Inorganic Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal 42119 Wuppertal Germany
| | - Olga Luzina
- N.N Vorozhtsov Novosibirsk Institute of Organic Chemistry of the Siberian Branch of Russian Academy of Sciences Novosibirsk Russian Federation
| | - Nariman Salakhutdinov
- N.N Vorozhtsov Novosibirsk Institute of Organic Chemistry of the Siberian Branch of Russian Academy of Sciences Novosibirsk Russian Federation
| | - Konstantin Volcho
- N.N Vorozhtsov Novosibirsk Institute of Organic Chemistry of the Siberian Branch of Russian Academy of Sciences Novosibirsk Russian Federation
| | - Hafiz S U B Farrukh
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Umme Kalsoom
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Rahman S Z Saleem
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Joanne Blanchfield
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia QLD Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| | - Karl A Hansford
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St Lucia QLD Australia
| |
Collapse
|
26
|
Gao D, Shi L, Huang Y, Lv Y, Yang X, Du Z. Synthesis of 2-Amino-4, 5-Diarylthiazole Derivatives and Evaluation of Their Anti- Candida Albicans Activity. Molecules 2025; 30:1643. [PMID: 40286249 PMCID: PMC11990618 DOI: 10.3390/molecules30071643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
The thiazole heterocycle is one of the most common moieties found in various drugs. Using 2-aminothiazole as the core structure, the amino group was functionalized with an amide. As a result, 30 trisubstituted 2-amino-4, 5-diarylthiazole derivatives were synthesized, with different substitutions introduced at the C2, C4, and C5 positions. The anti-Candida albicans biological activities of these synthetic compounds on five kinds of Candida albicans at different concentrations were detected by the microdilution method. In the first round, four derivatives of 2-amino-4, 5-diarylthiazole exhibited moderate anti-Candida albicans activity. Among them, 4a8 was chosen to be subjected to a demethylation process. Thus, 5a8 was synthesized successfully, giving anti-Candida albicans activity (MIC80 = 9 μM) similar to that of a typical antifungal drug, fluconazole. To understand the mechanism of anti-Candida albicans, molecular docking of the most active 5a8 against four target proteins of anti-Candida albicans, such as glutamine-fructose-6-phosphoamidamitransferase (GFAT), protein kinase (Yck2), heat-shock protein 90 (Hsp90), and lanosterol 14a-demethylase (CYP51) was carried out. Our research will provide an experimental basis and theoretical guidance for the further design of a new aminothiazole-leading pharmaceutical molecule.
Collapse
Affiliation(s)
- Dongmei Gao
- Yangling Vocational & Technical College, Yangling 712100, China
| | - Lele Shi
- School of Chemistry and Pharmacy, Northwest A & F University, Yangling 712100, China; (L.S.); (Y.H.); (Y.L.); (X.Y.)
| | - Yuhang Huang
- School of Chemistry and Pharmacy, Northwest A & F University, Yangling 712100, China; (L.S.); (Y.H.); (Y.L.); (X.Y.)
| | - Yingmei Lv
- School of Chemistry and Pharmacy, Northwest A & F University, Yangling 712100, China; (L.S.); (Y.H.); (Y.L.); (X.Y.)
| | - Xuan Yang
- School of Chemistry and Pharmacy, Northwest A & F University, Yangling 712100, China; (L.S.); (Y.H.); (Y.L.); (X.Y.)
| | - Zhenting Du
- School of Chemistry and Pharmacy, Northwest A & F University, Yangling 712100, China; (L.S.); (Y.H.); (Y.L.); (X.Y.)
| |
Collapse
|
27
|
Smith DJ, Melhem MSC, Dirven J, de Azevedo CMPES, Marques SG, Jacomel Favoreto de Souza Lima B, Vicente VA, Teixeira Sousa MDG, Venturini J, Wiederhold NP, Seyedmousavi A, Dufresne PJ, de Hoog S, Lockhart SR, Hagen F, Santos DWDCL. Establishment of epidemiological cutoff values for Fonsecaea pedrosoi, the primary etiologic agent of chromoblastomycosis, and eight antifungal medications. J Clin Microbiol 2025:e0190324. [PMID: 40183549 DOI: 10.1128/jcm.01903-24] [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: 12/02/2024] [Accepted: 01/15/2025] [Indexed: 04/05/2025] Open
Abstract
Chromoblastomycosis, a fungal neglected tropical disease, is acquired through traumatic inoculation and is clinically characterized by a chronic granulomatous infection of the skin and subcutaneous tissue. Fonsecaea pedrosoi is the most commonly reported etiologic agent globally. Itraconazole is considered first-line therapy, but successful treatment with terbinafine, voriconazole, and posaconazole has been reported. F. pedrosoi minimum inhibitory concentration (MIC) data are limited, and epidemiological cutoffs (ECVs) are lacking; such data are important to help monitor antifungal resistance trends and guide initial antifungal selection. Thus, we performed antifungal susceptibility testing (AFST) on F. pedrosoi isolates and determined the MIC distributions and ECVs. AFST on Fonsecaea pedrosoi isolates was conducted at six laboratories from October 2023 to June 2024. Species identification was previously confirmed by DNA sequence analysis. AFST was performed by CLSI M38 standard broth microdilution method for itraconazole, voriconazole, posaconazole, isavuconazole, ketoconazole, terbinafine, flucytosine, and amphotericin B. The ECVs were established using the iterative statistical method with ECOFFinder (version 2.1) following CLSI M57 guidelines. We analyzed MIC results from 148 Fonsecaea pedrosoi isolates. The calculated ECVs were itraconazole, 0.5 µg/mL; voriconazole, 0.5 µg/mL; posaconazole, 0.5 µg/mL; isavuconazole, 1 µg/mL; ketoconazole, bimodal, no ECV determined; terbinafine, 0.25 µg/mL; flucytosine, rejected; and amphotericin, 8 µg/mL. These Fonsecaea pedrosoi ECVs, obtained through a multicenter international effort, provide a baseline to better understand the in vitro antifungal susceptibility profile of this species and monitor resistance. Clinicians and researchers can use these values to detect non-wild-type isolates with reduced susceptibility, reevaluate therapeutic options, and investigate potential clinical resistance if treatment failure occurs.IMPORTANCEChromoblastomycosis is a neglected tropical disease caused by an environmental, dematiaceous fungus. This fungal disease is acquired after a break in the skin that allows the fungus to enter, leading to a chronic infection in the skin and subcutaneous tissue. It is difficult to treat and often requires years of antifungal treatment. Fonsecaea pedrosoi is the most reported causative agent globally. Limited antifungal susceptibility data exist for F. pedrosoi making interpreting minimum inhibitory concentration (MIC) results difficult. We performed antifungal susceptibility testing on 148 F. pedrosoi isolates to establish MIC distributions and epidemiologic cutoff values (ECVs) for eight antifungals, including those commonly used to treat chromoblastomycosis. The calculated ECVs for the commonly used antifungals itraconazole and terbinafine were 0.5 and 0.25 µg/mL, respectively. ECVs can be helpful in choosing potential treatment options for F. pedrosoi and monitoring antifungal resistance epidemiology.
Collapse
Affiliation(s)
- Dallas J Smith
- Mycotic Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Marcia S C Melhem
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, State of Mato Grosso do Sul, Brazil
- Tropical Medicine Institute, Medical School, University of São Paulo, São Paulo, Brazil
- Mycology Unit, Parasitology and Mycology Department, São Paulo State University, São Paulo, Brazil
| | - Jessy Dirven
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | | | - Sirlei Garcia Marques
- Laboratório Cedro, São Luís, Maranhão, Brazil
- Department of Infectious Diseases and Infection Control, Universidade Federal do Maranhão, EBSERH, São Luís, Maranhão, Brazil
| | - Bruna Jacomel Favoreto de Souza Lima
- Post-graduation Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, Brazil
- Radboudumc/CWZ Center of Expertise for Mycology, Nijmegen, The Netherlands
| | - Vania Aparecida Vicente
- Post-graduation Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, Brazil
- Post-graduation Program of Engineering Bioprocess and Biotechnology, Federal University of Parana, Curitiba, Brazil
| | - Maria da Glória Teixeira Sousa
- Faculdade de Medicina FMUSP, Institute of Tropical Medicine, Universidade de Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - James Venturini
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, State of Mato Grosso do Sul, Brazil
| | - Nathan P Wiederhold
- Fungus Testing Laboratory, Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Amir Seyedmousavi
- Microbiology Service, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Philippe J Dufresne
- Laboratoire de santé publique du Québec, Institut National de Santé Publique du Québec (INSPQ), Sainte-Anne-de-Bellevue, Québec, Canada
| | - Sybren de Hoog
- Radboudumc/CWZ Center of Expertise for Mycology, Nijmegen, The Netherlands
| | - Shawn R Lockhart
- Mycotic Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ferry Hagen
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, Amsterdam, The Netherlands
| | - Daniel Wagner de C L Santos
- Department of Infectious Diseases and Infection Control, Universidade Federal do Maranhão, EBSERH, São Luís, Maranhão, Brazil
- Instituto D´Or de Pesquisa e Ensino, IDOR, São Paulo, Brazil
| |
Collapse
|
28
|
Chedraoui C, Fattouh N, El Hachem S, Younes M, Khalaf RA. Induction of Antifungal Tolerance Reveals Genetic and Phenotypic Changes in Candida glabrata. J Fungi (Basel) 2025; 11:284. [PMID: 40278105 PMCID: PMC12028409 DOI: 10.3390/jof11040284] [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: 10/10/2024] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 04/26/2025] Open
Abstract
Candida glabrata is an opportunistic, pathogenic fungus that is increasingly isolated from hospitalized patients. The incidence of drug tolerance, heteroresistance, and resistance is on the rise due to an overuse of antifungal drugs. The aim of this study was to expose a sensitive C. glabrata strain to sequentially increasing concentrations of two antifungal drugs, fluconazole, an azole that targets ergosterol biosynthesis, or caspofungin, an echinocandin that targets cell wall glucan synthesis. Analysis of the drug-exposed isolates showed development of antifungal tolerance, chromosomal abnormalities, decreased adhesion, attenuated virulence, and an increase in efflux pump activity. Furthermore, whole genome sequencing of all isolates exposed to different concentrations of fluconazole or caspofungin was performed to determine mutations in key genes that could correlate with the observed phenotypes. Mutations were found in genes implicated in adhesion, such as in the AWP, PWP, and EPA family of genes. Isolates exposed to higher drug concentrations displayed more mutations than those at lower concentrations.
Collapse
Affiliation(s)
- Christy Chedraoui
- Department of Biological Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon; (C.C.); (N.F.); (S.E.H.); (M.Y.)
| | - Nour Fattouh
- Department of Biological Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon; (C.C.); (N.F.); (S.E.H.); (M.Y.)
- Department of Biology, Saint George University of Beirut, Beirut 1100-2807, Lebanon
| | - Setrida El Hachem
- Department of Biological Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon; (C.C.); (N.F.); (S.E.H.); (M.Y.)
| | - Maria Younes
- Department of Biological Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon; (C.C.); (N.F.); (S.E.H.); (M.Y.)
| | - Roy A. Khalaf
- Department of Biological Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon; (C.C.); (N.F.); (S.E.H.); (M.Y.)
| |
Collapse
|
29
|
Daliu P, Bogdan I, Rosca O, Licker M, Stanga LC, Hogea E, Vaduva DB, Muntean D. Fungal Pulmonary Coinfections in COVID-19: Microbiological Assessment, Inflammatory Profiles, and Clinical Outcomes. Biomedicines 2025; 13:864. [PMID: 40299432 PMCID: PMC12024551 DOI: 10.3390/biomedicines13040864] [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: 01/25/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background and Objectives: Secondary pulmonary fungal infections in coronavirus disease 2019 (COVID-19) remain underexplored despite emerging reports linking them to heightened morbidity. Comorbidities, steroid use, and prolonged hospital stays can predispose patients to opportunistic fungi. This study aimed to evaluate the impact of fungal coinfection on inflammatory markers, disease severity, antifungal resistance profiles, and outcomes in hospitalized COVID-19 patients. Methods: This retrospective observational study enrolled 280 adults (≥18 years) with real-time polymerase chain reaction (RT-PCR)-confirmed COVID-19 admitted to a tertiary care center (January 2023-December 2024). Patients were divided into a COVID-19-only group (n = 216) and a COVID-fungal group (n = 64) based on bronchoalveolar lavage, sputum, and/or blood culture positivity for fungal pathogens. Inflammatory markers (C-reactive protein (CRP), procalcitonin, the neutrophil-to-lymphocyte ratio, and the systemic immune inflammation index) and severity scores (Acute Physiology and Chronic Health Evaluation II, CURB-65 score, and the National Early Warning Score) were measured. We assessed antifungal susceptibilities and recorded ICU admissions, ventilation, hospital length of stay, and mortality. Results:Aspergillus fumigatus (31.3%), Candida albicans (28.1%), Cryptococcus neoformans (7.8%), Pneumocystis jirovecii (6.3%), and Mucorales (6.3%) dominated; Candida glabrata, Candida tropicalis, and mixed infections were also noted. Multidrug-resistant (MDR) isolates or resistance to triazoles occurred in 25.0% of cultures. The COVID-19-fungal group showed significantly higher CRP (85.7 vs. 71.6 mg/L, p < 0.001), procalcitonin (2.4 vs. 1.3 ng/mL, p < 0.001), and APACHE II scores (18.6 vs. 14.8, p < 0.001). intensive-care unit admissions (39.1% vs. 19.9%, p = 0.004) and mechanical ventilation (26.6% vs. 10.2%, p = 0.01) were more frequent with fungal coinfection. Mortality trended at a higher rate (15.6% vs. 7.4%, p = 0.06). Conclusions: Pulmonary fungal coinfections intensify the inflammatory milieu, elevate severity scores, and lead to more frequent ICU-level interventions in COVID-19 patients. Early identification, guided by culture-based and molecular diagnostics, alongside prompt antifungal therapy, could mitigate adverse outcomes. These findings underscore the critical need for proactive fungal surveillance and rigorous stewardship in managing severe COVID-19 pneumonia.
Collapse
Affiliation(s)
- Petrinela Daliu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Iulia Bogdan
- Department of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (I.B.); (O.R.)
- Methodological and Infectious Diseases Research Center, Department of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ovidiu Rosca
- Department of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (I.B.); (O.R.)
- Methodological and Infectious Diseases Research Center, Department of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Monica Licker
- Discipline of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (M.L.); (D.B.V.); (D.M.)
- Microbiology Laboratory, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania
| | - Livia Claudia Stanga
- Discipline of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (M.L.); (D.B.V.); (D.M.)
| | - Elena Hogea
- Discipline of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (M.L.); (D.B.V.); (D.M.)
| | - Delia Berceanu Vaduva
- Discipline of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (M.L.); (D.B.V.); (D.M.)
| | - Delia Muntean
- Discipline of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (M.L.); (D.B.V.); (D.M.)
| |
Collapse
|
30
|
Roy A, Ganguly A, Pal S, Bhowmick S, Das N. Study on antifungal usage in patients with vaginal itching and discharge at a private hospital in Kolkata using the National Accreditation Board for Hospitals and Healthcare Providers and the World Health Organization prescribing criteria. Perspect Clin Res 2025; 16:81-86. [PMID: 40322472 PMCID: PMC12048093 DOI: 10.4103/picr.picr_78_24] [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: 05/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 05/08/2025] Open
Abstract
Introduction One in every 10 women presents with abnormal vaginal discharge in Gynaecology and Obstetrics Department, with the most common cause being candidiasis. Irrational use of antifungal for treatment of this condition leads to antifungal resistance and increase morbidity. Materials and Methods This retrospective study was conducted at the Department of Pharmacology in collaboration of Department of Obstetrics and Gynaecology of the private medical college hospital in Kolkata. One hundred and forty outpatient department prescriptions were screened and included in this study from March 2024 to April 2024, and the prescriptions of the previous 6 months were collected. Results The average number of drugs per prescription was 1.1. Antifungals were prescribed in 89.3%, out of which only 8.6% were in accordance with the standard treatment guidelines of management of vaginal candidiasis. The dose of the drug was written in 11.5% of the prescriptions while the duration of treatment was written in 84.9% of the prescriptions. The fungal culture and sensitivity testing was documented in only 10.7% of the prescriptions. 29.5% of the prescriptions had the generic name of the medicines whereas the complete diagnosis was written in only 13.7% of the prescriptions. Conclusion This study highlights the commonly encountered errors in prescribing of antifungal drugs in a tertiary care teaching hospital. These errors may lead to irrational prescribing of antifungal and development of antifungal resistance in the long run. Active surveillance in the form of regular prescription audit and organizing regular training workshop for the prescribers will improve the prescribing practice.
Collapse
Affiliation(s)
- Ankita Roy
- Department of Pharmacology, KPC Medical College and Hospital, Kolkata, West Bengal, India
| | - Arpan Ganguly
- Department of Pharmacology, KPC Medical College and Hospital, Kolkata, West Bengal, India
| | - Soumyajit Pal
- Department of Gynaecology & Obstetrics, KPC Medical College and Hospital, Kolkata, West Bengal, India
| | - Subhrojyoti Bhowmick
- Department of Pharmacology, KPC Medical College and Hospital, Kolkata, West Bengal, India
| | - Nina Das
- Department of Pharmacology, KPC Medical College and Hospital, Kolkata, West Bengal, India
| |
Collapse
|
31
|
Dishan A, Ozkaya Y, Temizkan MC, Barel M, Gonulalan Z. Candida species covered from traditional cheeses: Characterization of C. albicans regarding virulence factors, biofilm formation, caseinase activity, antifungal resistance and phylogeny. Food Microbiol 2025; 127:104679. [PMID: 39667852 DOI: 10.1016/j.fm.2024.104679] [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: 01/18/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024]
Abstract
This study has provided characterization data (carriage of virulence, antifungal resistance, caseinase activity, biofilm-forming ability and genotyping) of Candida albicans isolates and the occurrence of Candida species in traditional cheeses collected from Kayseri, Türkiye. Phenotypic (E-test, Congo red agar and microtiter plate tests) and molecular tests (identification, virulence factors, biofilm-formation, antifungal susceptibility) were carried out. The phylogenetic relatedness of C. albicans isolates was obtained by constructing the PCA dendrogram from the mass spectra data. Of 102 samples, 13 (12.7%) were found to be contaminated with C. albicans, 15 (14.7%), 10 (9.8%) and five (4.9%) were found to be contaminated with C. krusei, C. lusitane and C. paraplosis, respectively. While seven (16.2%) of 43 Candida spp. isolates were obtained from cheese collected from villages, 36 (83.7%) belonged to cheeses collected from traditional retail stores. The carriage rate of C. albicans isolates belonging to virulence factors HSP90 and PLB1 genes was 30.7%. ALST1, ALST3, BCR, ECE, andHWP (virulence and biofilm-associated) genes were harbored by 30.7%, 23%, 38.4%, 53.8%, and 38.4% of the 13 isolates. According to the microplate test, eight (61.5%) of 13 isolates had strong biofilm production. ERG11 and FKS1 (antifungal resistance genes) were found in 46.1% and 23% of 13 isolates, respectively. Due to missense mutations, K128T, E266D and V488I amino acid changes were detected for some isolates regarding azole resistance. As a result of the E-test, of the 13 isolates, one (7.6%) was resistant to flucytosine, four (30.7%) were resistant to caspofungin, and nine (69.2%) were resistant to fluconazole. The PCA analysis clustered the studied isolates into two major clades. C. albicans isolates of traditional cheese collected from villages were grouped in the same cluster. Among the C. albicans isolates from village cheese, there were those obtained from the same dairy milk at different times. Samples from the same sales points produced at different dairy farms were also contaminated with C. albicans. Concerning food safety standards applied from farm to fork, in order to prevent these pathogenic agents from contaminating cheeses, attention to the hygiene conditions of the sale points, conscious personnel, prevention of cross contamination will greatly reduce public health threats in addition to the application of animal health control, milking hygiene, pasteurization parameters in traditional cheese production.
Collapse
Affiliation(s)
- Adalet Dishan
- Yozgat Bozok University, Faculty of Veterinary Medicine, Dept. of Food Hygiene and Technology, Yozgat, Turkiye.
| | - Yasin Ozkaya
- Erciyes University, Faculty of Veterinary Medicine, Dept. of Veterinary Public Health, Kayseri, Turkiye
| | - Mehmet Cevat Temizkan
- Yozgat Bozok University, Faculty of Veterinary Medicine, Dept. of Veterinary Genetics, Yozgat, Turkiye
| | - Mukaddes Barel
- Erciyes University, Faculty of Veterinary Medicine, Dept. of Veterinary Public Health, Kayseri, Turkiye
| | - Zafer Gonulalan
- Erciyes University, Faculty of Veterinary Medicine, Dept. of Veterinary Public Health, Kayseri, Turkiye
| |
Collapse
|
32
|
Hu T, Zheng Q, Cao C, Li S, Huang Y, Guan Z, Ji L, Bing J, Du H, Perry AM, Nobile CJ, Li B, Chu H, Huang G. An agricultural triazole induces genomic instability and haploid cell formation in the human fungal pathogen Candida tropicalis. PLoS Biol 2025; 23:e3003062. [PMID: 40168394 PMCID: PMC11960876 DOI: 10.1371/journal.pbio.3003062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/07/2025] [Indexed: 04/03/2025] Open
Abstract
The human fungal pathogen Candida tropicalis is widely distributed in clinical and natural environments. It is known to be an obligate diploid organism with an incomplete and atypical sexual cycle. Azole-resistant C. tropicalis isolates have been observed with increasing prevalence in many countries in recent years. Here, we report that tebuconazole (TBZ), a triazole fungicide widely used in agriculture, can induce ploidy plasticity and the formation of haploid cells in C. tropicalis. The evolved C. tropicalis strains with ploidy variations exhibit a cross-resistance between TBZ and standard azoles used in clinical settings (such as fluconazole and voriconazole). Similar to its diploid cells, these newly discovered C. tropicalis haploid cells are capable of undergoing filamentation, white-opaque switching, and mating. However, compared to its diploid cells, these haploid C. tropicalis cells grow more slowly under in vitro culture conditions and are less virulent in a mouse model of systemic infection. Interestingly, flow cytometry analysis of a clinical strain with extremely low genome heterozygosity indicates the existence of natural C. tropicalis haploids. Discovery of this C. tropicalis haploid state sheds new light into the biology and genetic plasticity of C. tropicalis and could provide the framework for the development of new genetic tools in the field.
Collapse
Affiliation(s)
- Tianren Hu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Qiushi Zheng
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Chengjun Cao
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Shuaihu Li
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanfeng Huang
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhangyue Guan
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Lingyu Ji
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jian Bing
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Han Du
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Austin M. Perry
- Department of Molecular and Cell Biology, University of California, Merced, California, United States of America
- Quantitative and Systems Biology Graduate Program, University of California, Merced, California, United States of America
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, University of California, Merced, California, United States of America
- Health Sciences Research Institute, University of California, Merced, California, United States of America
| | - Bing Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haiqing Chu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guanghua Huang
- Department of infectious diseases, Huashan Hospital, Shanghai Institute of Infectious Disease and Biosecurity and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| |
Collapse
|
33
|
Deng Q, Li Y, He W, Chen T, Liu N, Ma L, Qiu Z, Shang Z, Wang Z. A polyene macrolide targeting phospholipids in the fungal cell membrane. Nature 2025; 640:743-751. [PMID: 40108452 PMCID: PMC12003179 DOI: 10.1038/s41586-025-08678-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Abstract
The global spread of multidrug-resistant pathogenic fungi presents a serious threat to human health, necessitating the discovery of antifungals with unique modes of action1. However, conventional activity-based screening for previously undescribed antibiotics has been hampered by the high-frequency rediscovery of known compounds and the lack of new antifungal targets2. Here we report the discovery of a polyene antifungal antibiotic, mandimycin, using a phylogeny-guided natural-product discovery platform. Mandimycin is biosynthesized by the mand gene cluster, has evolved in a distinct manner from known polyene macrolide antibiotics and is modified with three deoxy sugars. It has demonstrated potent and broad-spectrum fungicidal activity against a wide range of multidrug-resistant fungal pathogens in both in vitro and in vivo settings. In contrast to known polyene macrolide antibiotics that target ergosterol, mandimycin has a unique mode of action that involves targeting various phospholipids in fungal cell membranes, resulting in the release of essential ions from fungal cells. This unique ability to bind multiple targets gives it robust fungicidal activity as well as the capability to evade resistance. The identification of mandimycin using the phylogeny-guided natural-product discovery strategy represents an important advancement in uncovering antimicrobial compounds with distinct modes of action, which could be developed to combat multidrug-resistant fungal pathogens.
Collapse
Affiliation(s)
- Qisen Deng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yinchuan Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenyan He
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tao Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Nan Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhixia Qiu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhuo Shang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| | - Zongqiang Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
34
|
Ceballos-Garzon A, Holzapfel M, Welsch J, Mercer D. Identification and antifungal susceptibility patterns of reference yeast strains to novel and conventional agents: a comparative study using CLSI, EUCAST and Sensititre YeastOne methods. JAC Antimicrob Resist 2025; 7:dlaf040. [PMID: 40110552 PMCID: PMC11920621 DOI: 10.1093/jacamr/dlaf040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/02/2025] [Indexed: 03/22/2025] Open
Abstract
Objectives The aim of this study was to identify and determine the MICs of 13 antifungal drugs, including the novel agents ibrexafungerp, manogepix and rezafungin, against 22 laboratory reference strains from 14 different Candida spp. and allied yeast genera using the EUCAST, CLSI and Sensititre™ YeastOne™ (SYO) methods. Results Complete agreement between molecular and proteomics methods was observed for identification. The compounds with the greatest in vitro activity, as indicated by the lowest geometric mean MIC (GM), were manogepix (GM: 0.01), isavuconazole (GM: 0.05) and rezafungin (GM: 0.03-0.07). The overall essential agreement (EA) (within ±0 to ±2 2-fold dilutions) between the reference methods, EUCAST and CLSI, was 95%, with results ranging from 82% (ibrexafungerp) to 100% (amphotericin B, anidulafungin, fluconazole, 5-flucytosine and micafungin). Regarding EA for EUCAST and CLSI compared with SYO, values were 91% and 89%, respectively. Nevertheless, when the MIC values were transformed into log2, significant differences were observed (e.g. fluconazole, ibrexafungerp and 5-flucytosine). At the species level, Candidozyma auris and Candida duobushaemulonii exhibited the highest number of cases with significant differences when comparing the three techniques for each antifungal. Conclusions The high EA observed reinforces the reliability of EUCAST, CLSI and SYO in guiding antifungal therapy. However, the differences in EA, particularly for ibrexafungerp and 5-flucytosine, highlight the importance of continued evaluation of these methodologies to ensure consistency. Given that antifungal susceptibility testing plays a critical role in treatment decisions, understanding these variations is essential to prevent potential misclassification of susceptibility profiles, which could impact clinical outcomes.
Collapse
Affiliation(s)
- Andres Ceballos-Garzon
- BIOASTER Research Institute, 40 avenue Tony Garnier, Lyon F-69007, France
- Translational Microbiology and Emerging Diseases Research Group (MICROS), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Marion Holzapfel
- BIOASTER Research Institute, 40 avenue Tony Garnier, Lyon F-69007, France
| | - Jeremy Welsch
- BIOASTER Research Institute, 40 avenue Tony Garnier, Lyon F-69007, France
| | - Derry Mercer
- BIOASTER Research Institute, 40 avenue Tony Garnier, Lyon F-69007, France
| |
Collapse
|
35
|
Keck JM, Dare RK, Jenkins MB, Rico JC, Grisham L, McDonald J, Viteri A, Bradsher RW. It's Here, It's There, There's Fungi Everywhere: A Case Series Utilizing Rezafungin for Invasive Candidiasis. Infect Dis Ther 2025; 14:889-895. [PMID: 40069441 PMCID: PMC11993525 DOI: 10.1007/s40121-025-01120-7] [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: 12/03/2024] [Accepted: 02/25/2025] [Indexed: 04/13/2025] Open
Abstract
Rezafungin is a long-acting echinocandin with broad coverage against Candida. Rezafungin has primarily been indicated for candidemia, with limited literature available on its use for infections outside of the bloodstream. Herein, three patient cases are presented from an academic medical center. Infectious processes presented include drug-resistant mucosal candidiasis, prosthetic joint infection, and candidemia involving Candida auris. In all three cases, patients received rezafungin. Clinical response was demonstrated in all patients as was tolerability of rezafungin. Together these cases provide further evidence for the use of rezafungin, including its use for treatment of invasive infections other than candidemia.
Collapse
Affiliation(s)
- Jacob M Keck
- Department of Pharmacy, University of Arkansas for Medical Sciences, 4301 W. Markham St. #529, Little Rock, AR, 72205, USA.
| | - Ryan K Dare
- Department of Medicine, Division of Infectious Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Mitchell B Jenkins
- Department of Medicine, Division of Infectious Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Juan C Rico
- Department of Medicine, Division of Infectious Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Luke Grisham
- Department of Medicine, Division of Infectious Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Jennifer McDonald
- Department of Pharmacy, University of Arkansas for Medical Sciences, 4301 W. Markham St. #529, Little Rock, AR, 72205, USA
| | - Alina Viteri
- Department of Pharmacy, University of Arkansas for Medical Sciences, 4301 W. Markham St. #529, Little Rock, AR, 72205, USA
| | - Robert W Bradsher
- Department of Medicine, Division of Infectious Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| |
Collapse
|
36
|
Jospe-Kaufman M, Fridman M. Illuminating antifungal mode of action and resistance with fluorescent probes. Curr Opin Chem Biol 2025; 85:102570. [PMID: 39965367 DOI: 10.1016/j.cbpa.2025.102570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/27/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025]
Abstract
The rise in fungal infections, driven by pathogens resistant to the limited scope of antifungal agents available, poses an increasing threat to global health and the economy. Addressing this challenge requires a thorough understanding of the mechanisms of antifungal agents and the development of advanced resistance diagnostic methods. This opinion manuscript highlights recent advancements in antifungal research, with a focus on chemical biology approaches, particularly the development of fluorescent probes derived from various antifungal agents. These probes reveal new aspects of antifungal activity and provide deeper insights into modes of action and resistance mechanisms. Live cell imaging of fungal pathogens labeled with these probes has uncovered novel strategies to enhance antifungal efficacy, understand virulence factors, and detect resistance. These unique small-molecule tools offer powerful new avenues for addressing the fungal infections crisis, harnessing chemical biology approaches to develop innovative solutions to the global challenges posed by fungi.
Collapse
Affiliation(s)
- Moriah Jospe-Kaufman
- 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
|
37
|
De Paiva Macedo J, Watanabe AAS, Machado ABF, Diniz CG, Da Silva VL, Dias VC. Unveiling antifungal resistance and biocide tolerance in clinical isolates of Candida spp. Future Microbiol 2025; 20:457-468. [PMID: 40119655 PMCID: PMC11980505 DOI: 10.1080/17460913.2025.2480924] [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: 01/06/2025] [Accepted: 03/14/2025] [Indexed: 03/24/2025] Open
Abstract
AIMS Candidiasis, caused by Candida spp. is an opportunistic infection with significant healthcare risks, worsened by trends in antifungal resistance. This study aimed to evaluate the antifungal susceptibility profile, investigate resistance mechanisms, assess efflux pump activity, and examine biocide tolerance in clinical Candida isolates. METHODS A total of 100 Candida isolates from hospitalized and outpatient individuals were analyzed for their antifungal susceptibility profile, molecular resistance mechanisms through PCR, efflux pump activity with the Cartwheel method, and biocide tolerance (sodium hypochlorite, hydrogen peroxide, and benzalkonium chloride), which was assessed by disk diffusion. RESULTS A high prevalence of resistance (87%) to at least one antifungal was observed, with 47.12% of isolates showing simultaneous multiple resistance to three azole derivatives. The highest antifungal agent resistance was observed for fluconazole (n = 70) and the highest susceptibility for amphotericin B (n = 1). The most common mutation was in the ERG11 gene (n = 38/43.7%). Efflux pump activity was detected in both C. albicans and non-albicans Candida species. Biocide testing revealed a higher tolerance for sodium hypochlorite, with an inhibition zone ranging from 18.25 (4.40) to 34.0 (4.00). CONCLUSIONS This study highlights significant antifungal resistance in Candida spp. particularly to azoles, stressing the need for improved infection control and novel therapeutic strategies.
Collapse
Affiliation(s)
- Jamile De Paiva Macedo
- Department of Biological Sciences, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Aripuana Aranha Sakurada Watanabe
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Alessandra Barbosa Ferreira Machado
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Cláudio Galuppo Diniz
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Vania Lucia Da Silva
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Vanessa Cordeiro Dias
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| |
Collapse
|
38
|
Eletel L, Thomas T, Berry EA, Kearns GL. Emerging Treatments in Neonatal Fungal Infections: Progress and Prospects. Paediatr Drugs 2025:10.1007/s40272-025-00688-4. [PMID: 40117020 DOI: 10.1007/s40272-025-00688-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 03/23/2025]
Abstract
Fungal infections in neonates are potentially life threatening. The differential diagnosis for neonatal rashes is extensive, with common culprits including both bacteria and fungi. Candida albicans is the predominant fungal pathogen, causing infections that range from superficial disease to severe systemic conditions, including sepsis and meningitis. Neonates, especially those who are preterm, are particularly susceptible because of developmentally immature immune systems and the use of invasive procedures and devices in neonatal intensive care units. Congenital cutaneous candidiasis, acquired in utero or during delivery, can lead to disseminated infection with high mortality rates. Early diagnosis and prompt antifungal treatment are crucial but challenging because of subtle clinical presentations, making accurate identification of the offending organism essential for selecting the appropriate treatment. Candida species account for the majority of neonatal fungal infections, with different species necessitating distinct treatments because of varying susceptibility profiles. Aspergillus, another significant pathogen, poses high mortality risks and can present either cutaneously or systemically. Malassezia, though less common, primarily affects preterm infants with catheter-related fungemia. Other fungal species, including Zygomycetes, Trichosporon, and Cryptococcus, rarely produce neonatal infections but are noteworthy for consideration. Treatment of fungal infection is critical despite the relative paucity of information regarding the clinical pharmacology of many antifungal drugs in neonates. We review the major antifungal agents (e.g., amphotericin B, the echinocandins, the azoles) and provide pharmacologic and dosing information. Finally, preventive strategies, including the use of stringent aseptic techniques and careful clinical monitoring, are essential to mitigate both the incidence and severity of these infections in neonates and infants in the first months of life.
Collapse
Affiliation(s)
- Lucy Eletel
- Department of Medical Education, Anne Marion Burnett School of Medicine at Texas Christian University, Fort Worth, TX, USA
| | - Talia Thomas
- Department of Medical Education, Anne Marion Burnett School of Medicine at Texas Christian University, Fort Worth, TX, USA
| | - Emily A Berry
- Department of Medical Education, Anne Marion Burnett School of Medicine at Texas Christian University, Fort Worth, TX, USA
| | - Gregory L Kearns
- Department of Pediatrics, Anne Marion Burnett School of Medicine at Texas Christian University, 1100 W. Rosedale St., Fort Worth, TX, 76104, USA.
| |
Collapse
|
39
|
Sun Y, Bock R, Li Z. A hidden intrinsic ability of bicistronic expression based on a novel translation reinitiation mechanism in yeast. Nucleic Acids Res 2025; 53:gkaf220. [PMID: 40156854 PMCID: PMC11952965 DOI: 10.1093/nar/gkaf220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Gene organization in operons and co-expression as polycistronic transcripts is characteristic of prokaryotes. With the evolution of the eukaryotic translation machinery, operon structure and expression of polycistrons were largely abandoned. Whether eukaryotes still possess the ability to express polycistrons, and how they functionally activate bacterial operons acquired by horizontal DNA transfer is unknown. Here, we demonstrate that a polycistron can be rapidly activated in yeast by induction of bicistronic expression under selection. We show that induced translation of the downstream cistron in a bicistronic transcript is based on a novel type of reinitiation mediated by the 80S ribosome and triggered by inefficient stop codon recognition, and that induced bicistronic expression is stable and independent of cis-elements. These results provide key insights into the epigenetic mechanism of the pathway of activation. We also developed a yeast strain that efficiently expresses bicistronic constructs, but does not carry any genomic DNA sequence change, and utilized this strain to synthesize a high-value metabolite from a bicistronic expression construct. Together, our results reveal the capacity of yeast to express bicistrons in a previously unrecognized pathway. While this capacity is normally hidden, it can be rapidly induced by selection to improve fitness.
Collapse
Affiliation(s)
- Yiwen Sun
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Ralph Bock
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam-Golm, Germany
| | - Zhichao Li
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| |
Collapse
|
40
|
Zhao WB, An JX, Jin YR, Jing CX, Zhang SY, Liang HJ, Dai TL, Luo XF, Zhang BQ, Zhang ZJ, Liu YQ. Indoloquinoline alkaloid neocryptolepine derivative inhibits Botrytis cinerea by targeting thiamine thiazole synthase. SCIENCE ADVANCES 2025; 11:eadq5329. [PMID: 40073123 PMCID: PMC11900860 DOI: 10.1126/sciadv.adq5329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025]
Abstract
The emergence and rapid spread of multidrug-resistant Botrytis cinerea strains pose a great challenge to the quality and safety of agricultural products and the efficient use of pesticides. Previously unidentified fungicides and targets are urgently needed to combat B. cinerea-associated infections as alternative therapeutic options. In this study, the promising compound Z24 demonstrated efficacy against all tested plant pathogenic fungi. Thiamine thiazole synthase (Bcthi4) was identified as a target protein of Z24 by drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), and surface plasmon resonance (SPR) assays. Molecular docking and enzyme activity experiments have demonstrated that Z24 can affect the function of Bcthi4. Last, mechanistic studies show that Z24 inhibits thiamine biosynthesis by binding to Bcthi4 and induces up-regulation of alternative splicing [alternative 5' splice site (A5SS)] of the Bcthi4 gene. In conclusion, by targeting Bcthi4, Z24 has the potential to be developed as a previously unidentified anti-B. cinerea candidate.
Collapse
Affiliation(s)
- Wen-Bin Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jun-Xia An
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ya-Rui Jin
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Chen-Xin Jing
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Shao-Yong Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Hong-Jie Liang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Tian-Li Dai
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xiong-Fei Luo
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Bao-Qi Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zhi-Jun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, Zhejiang 313000, China
| |
Collapse
|
41
|
Pagano L, Fernández OM. Clinical aspects and recent advances in fungal diseases impacting human health. J Antimicrob Chemother 2025; 80:i2-i8. [PMID: 40085539 PMCID: PMC11908536 DOI: 10.1093/jac/dkaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/04/2024] [Indexed: 03/16/2025] Open
Abstract
Fungal diseases are of growing clinical concern in human medicine as the result of changes in the epidemiology, diversity in clinical presentation, emergence of new pathogens, difficulties in diagnosis and increasing resistance to antifungals of current available classes. There is a need for high disease awareness among the public and healthcare physicians, improvement in diagnostic methods and the development of drugs from new therapeutic classes with an improved resistance profile. In this article, we will explore some key aspects of fungal diseases in humans and provide a general overview of this important topic.
Collapse
Affiliation(s)
- Livio Pagano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli – IRCCS, Rome, Italy
| | | |
Collapse
|
42
|
Yarava JR, Gautam I, Jacob A, Fu R, Wang T. Proton-Detected Solid-State NMR for Deciphering Structural Polymorphism and Dynamic Heterogeneity of Cellular Carbohydrates in Pathogenic Fungi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642223. [PMID: 40161786 PMCID: PMC11952318 DOI: 10.1101/2025.03.09.642223] [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: 04/02/2025]
Abstract
Carbohydrate polymers in their cellular context display highly polymorphic structures and dynamics essential to their diverse functions, yet they are challenging to analyze biochemically. Proton-detection solid-state NMR spectroscopy offers high isotopic abundance and sensitivity, enabling rapid and high-resolution structural characterization of biomolecules. Here, an array of 2D/3D 1H-detection solid-state NMR techniques are tailored to investigate polysaccharides in fully protonated or partially deuterated cells of three prevalent pathogenic fungi: Rhizopus delemar, Aspergillus fumigatus, and Candida albicans, representing filamentous species and yeast forms. Selective detection of acetylated carbohydrates reveals fifteen forms of N-acetylglucosamine units in R. delemar chitin, which coexists with chitosan as separate domains or polymers and associates with proteins only at limited sites. This is supported by distinct order parameters and effective correlation times of their motions, analyzed through relaxation measurements and model-free analysis. Five forms of α-1,3-glucan with distinct structural origins and dynamics were identified in A. fumigatus, important for this buffering polysaccharide to perform diverse roles of supporting wall mechanics and regenerating soft matrix under antifungal stress. Eight α-1,2-mannan sidechain variants in C. albicans were resolved, highlighting the crucial role of mannan sidechains in maintaining interactions with other cell wall polymers to preserve structural integrity. These methodologies provide novel insights into the functional structures of key fungal polysaccharides and create new opportunities for exploring carbohydrate biosynthesis and modifications across diverse organisms.
Collapse
Affiliation(s)
| | - Isha Gautam
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Anand Jacob
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Riqiang Fu
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Tuo Wang
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
43
|
Safi C, Camaioni L, Othman M, Lambert D, Buisine M, Lawson AM, Ghinet A, Daïch A, Jawhara S. Cyclic N, O-acetals and corresponding opened N, N-aminals as new scaffolds with promising anti-inflammatory and antifungal activities against Candida albicans. Sci Rep 2025; 15:8364. [PMID: 40069300 PMCID: PMC11897400 DOI: 10.1038/s41598-025-92635-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
P2 × 7R is crucial in the pathogenesis of chronic inflammatory diseases, and its activation leads to the release of pro-inflammatory cytokines, exacerbating the inflammatory response. Two new series of scarce cyclic N, O-acetals (ATF 61-74) and corresponding opened N, N-aminals (CS 1-14) have been designed as novel potential P2RX7 antagonists, then synthesized and evaluated for their anti-inflammatory properties through investigating the pro-inflammatory markers and also for their antifungal activity against Candida albicans. Three compounds (ATF 64, CS 8, and CS 9) exhibited dual antifungal and anti-inflammatory properties. ATF 64, CS 8, and CS 9 reduced ROS production and IL-1β expression in macrophages and intestinal cells in a manner correlated with NF-KB expression. These compounds showed excellent antifungal activity against clinical isolates of C. albicans resistant to fluconazole and caspofungin, and reduced C. albicans biofilm formation. Treatment with CS 8 or CS 9 protected the nematode Caenorhabditis elegans against infection with C. albicans and enhanced antimicrobial gene expression. This duality of action offers a promising new pharmacological strategy to counteract inflammatory diseases and propels N, N-aminals as promising candidates for future optimization and investigation.
Collapse
Affiliation(s)
- Christine Safi
- URCOM UR 3221, Université Le Havre Normandie, Normandie Université, Le Havre, 76600, France
- INC3M, FR CNRS 3038, 25 Rue Philippe Lebon, BP 1123, 76063, Le Havre Cedex, France
- JUNIA, Health and Environment, Laboratory of Sustainable Chemistry and Health, 59000, Lille, France
| | - Louis Camaioni
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Institut National de la Santé et de la Recherche Médicale U1285, Centre National de la Recherche Scientifique, University of Lille, 59000, Lille, France
- Medicine Faculty, University of Lille, 59000, Lille, France
- Service de Parasitologie Mycologie, Pôle de Biologie Pathologie Génétique, CHU Lille, 59000, Lille, France
| | - Mohamed Othman
- URCOM UR 3221, Université Le Havre Normandie, Normandie Université, Le Havre, 76600, France
- INC3M, FR CNRS 3038, 25 Rue Philippe Lebon, BP 1123, 76063, Le Havre Cedex, France
| | - Dylan Lambert
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Institut National de la Santé et de la Recherche Médicale U1285, Centre National de la Recherche Scientifique, University of Lille, 59000, Lille, France
- Medicine Faculty, University of Lille, 59000, Lille, France
- Service de Parasitologie Mycologie, Pôle de Biologie Pathologie Génétique, CHU Lille, 59000, Lille, France
| | - Mathys Buisine
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Institut National de la Santé et de la Recherche Médicale U1285, Centre National de la Recherche Scientifique, University of Lille, 59000, Lille, France
- Medicine Faculty, University of Lille, 59000, Lille, France
- Service de Parasitologie Mycologie, Pôle de Biologie Pathologie Génétique, CHU Lille, 59000, Lille, France
| | - Ata Martin Lawson
- URCOM UR 3221, Université Le Havre Normandie, Normandie Université, Le Havre, 76600, France
- INC3M, FR CNRS 3038, 25 Rue Philippe Lebon, BP 1123, 76063, Le Havre Cedex, France
| | - Alina Ghinet
- JUNIA, Health and Environment, Laboratory of Sustainable Chemistry and Health, 59000, Lille, France
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut National de la Santé et de la Recherche Médicale, CHU Lille, Institut Pasteur de Lille, University of Lille, Lille, 59000, France
- Faculty of Chemistry, 'Alexandru Ioan Cuza' University of Iasi, Bd. Carol I, Nr. 11, 700506, Iasi, Romania
| | - Adam Daïch
- URCOM UR 3221, Université Le Havre Normandie, Normandie Université, Le Havre, 76600, France
- INC3M, FR CNRS 3038, 25 Rue Philippe Lebon, BP 1123, 76063, Le Havre Cedex, France
| | - Samir Jawhara
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Institut National de la Santé et de la Recherche Médicale U1285, Centre National de la Recherche Scientifique, University of Lille, 59000, Lille, France.
- Medicine Faculty, University of Lille, 59000, Lille, France.
- Service de Parasitologie Mycologie, Pôle de Biologie Pathologie Génétique, CHU Lille, 59000, Lille, France.
| |
Collapse
|
44
|
Gawor W, Góralska K, Galant S, Majewski S, Piotrowski W, Miłkowska-Dymanowska J, Kiszałkiewicz J, Brzeziańska-Lasota E. Fungal microbiota in COPD patients during exacerbations. J Mycol Med 2025; 35:101543. [PMID: 40101573 DOI: 10.1016/j.mycmed.2025.101543] [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: 04/21/2024] [Revised: 01/30/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
Acute exacerbations of chronic obstructive pulmonary disease (COPD) results in increased mortality and can be triggered by a range of factors, including microorganisms. Very little studies have examined the role of fungi and fungal diversity in COPD patients. The aim of the study was to determine the role of Candida in COPD during an exacerbation. Oral swabs, sputum, feces and whole blood samples were collected from the AECOPD patients and control group. Mycological and serological analysis were performed. Yeast were statistically significantly more often isolated from the AECOPD group (97.06%) than from the control group (26.32%). 7 species were isolated from the AECOPD, and 3 from the control group. Dominated Candida albicans followed by C. tropicalis. α-diversity was much greater in AECOPD patients than in controls. β-diversity was also assessed. A much higher level of antimycotic resistance was observed in isolates from the AECOPD group, which affects the effectiveness of therapy. Serological tests showed twice the frequency of positive results in the AECOPD group. The mycobiota of AECOPD patients is numerically and taxonomically richer than controls, including species less frequently recorded in humans. Our research confirms that fungal mycobiota may be a potential factor influencing the development of exacerbations and progression of COPD.
Collapse
Affiliation(s)
- Weronika Gawor
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, Mazowiecka 5 Street, Lodz 92-215, Poland.
| | - Katarzyna Góralska
- Department of Biology and Parasitology, Chair of Biology and Medical Microbiology, Medical University of Lodz, Lucjan Żeligowski 7/9 Street, Lodz 90-752, Poland
| | - Sandra Galant
- Department of Biology and Parasitology, Chair of Biology and Medical Microbiology, Medical University of Lodz, Lucjan Żeligowski 7/9 Street, Lodz 90-752, Poland
| | - Sebastian Majewski
- Department of Pneumology, Medical University of Lodz, Stefan Kopciński 22 Street, Lodz 90-153, Poland
| | - Wojciech Piotrowski
- Department of Pneumology, Medical University of Lodz, Stefan Kopciński 22 Street, Lodz 90-153, Poland
| | | | - Justyna Kiszałkiewicz
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, Mazowiecka 5 Street, Lodz 92-215, Poland
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, Mazowiecka 5 Street, Lodz 92-215, Poland
| |
Collapse
|
45
|
Ma Y, Zhou Y, Jia T, Zhuang Z, Xue P, Yang L. Deciphering the role of mitochondria in human fungal drug resistance. Mycology 2025:1-14. [DOI: 10.1080/21501203.2025.2473507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 05/04/2025] Open
Affiliation(s)
| | | | | | | | | | - Liang Yang
- Southern University of Science and Technology
| |
Collapse
|
46
|
Chaudhary R, Thakur Z. From detection to action: new diagnostic insights into antifungal resistance. Expert Rev Mol Diagn 2025:1-6. [PMID: 40052616 DOI: 10.1080/14737159.2025.2477027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/05/2025] [Indexed: 03/12/2025]
Affiliation(s)
- Renu Chaudhary
- Structural Biology Lab, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Zoozeal Thakur
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be), University, Ambala, India
| |
Collapse
|
47
|
Carneiro JNP, Dos Santos ATL, Fonseca VJA, de Freitas MA, Dos Santos Silva F, de Souza LAL, Araújo NMS, de Oliveira Bezerra de Sousa D, Silva RGG, da Silva Neto JX, de Menezes IRA, Coutinho HDM, Morais-Braga MFB. Antifungal Action of Valencene and Nootkatone Compounds in Association with Fluconazole and Their Mechanism of Action Against Candida spp. and Pichia kudriavzevii Strains. Curr Microbiol 2025; 82:168. [PMID: 40038128 DOI: 10.1007/s00284-025-04133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025]
Abstract
Candidemia is a public health challenge as it causes thousands of annual deaths and combating it has become difficult due to the development of resistance in Candida spp. Compounds derived from natural products may counter this resistance. Therefore, we evaluate the intrinsic and combined antifungal activity of valencene and nootkatone compounds and their possible mechanism of action against Candida spp. Using the microdilution method, the antifungal effect of sesquiterpenes and their combination with fluconazole was determined. The results comprised the yeast growth curve and its 50% Inhibitory Concentration (IC50). They showed that the compounds alone inhibited microbial growth at a concentration of 1024 µg/mL for valencene being able to kill the fungus Pichia kudriavzevii (Candida krusei), for nootkatone the inhibition occurred at 512 µg/mL and was able to kill the species P. kudriavzevii and Candida tropicalis. Combined with the antifungal, the inhibition occurred at low concentrations (2 and 4 µg/mL) against all strains except P. kudriavzevii, which the combination with nootkatone inhibited at 512 µg/mL. The IC50 revealed inhibition of the strains at higher concentrations in the compounds and fluconazole alone compared to the combination concentrations. In addition, both compounds acted through the production of reactive oxygen species, helping the antifungal against C. albicans and P. kudriavzevii, contributing minimally to compromising membrane viability. Thus, the compounds show promise for combined activity with fluconazole.
Collapse
|
48
|
Puumala E, Nandakumar M, Yiu B, Stogios PJ, Strickland BG, Zarnowski R, Wang X, Williams NS, Savchenko A, Andes DR, Robbins N, Whitesell L, Willson TM, Cowen LE. Structure-guided optimization of small molecules targeting Yck2 as a strategy to combat Candida albicans. Nat Commun 2025; 16:2156. [PMID: 40038303 PMCID: PMC11880385 DOI: 10.1038/s41467-025-57346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
Candida albicans is the most common cause of life-threatening fungal infection in the developed world but remains a therapeutic challenge. Protein kinases have been rewarding drug targets across diverse indications but remain untapped for antifungal development. Previously, screening kinase inhibitors against C. albicans revealed a 2,3-aryl-pyrazolopyridine, GW461484A (GW), which targets casein kinase 1 (CK1) family member Yck2. Here, we report optimization of GW via two complementary approaches, synthesis of bioisosteres possessing an imidazo[1,2-a]pyridine core, and R-group substitution of GW's pyrazolo[1,5-a]pyridine core. Characterization of compounds reveals two 6-cyano derivatives with improved pharmacological properties that retain whole-cell bioactivity and selectivity for fungal Yck2 compared to human CK1α. Efficacy studies in mice indicate both analogs possess single-agent activity against C. albicans resistant to first-line echinocandin antifungals and potentiate non-curative echinocandin treatment. Results validate Yck2 as an antifungal target and encourage further development of inhibitors acting by this previously unexploited mode of action.
Collapse
Affiliation(s)
- Emily Puumala
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Meganathan Nandakumar
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bonnie Yiu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Peter J Stogios
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Benjamin G Strickland
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert Zarnowski
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Xiaoyu Wang
- Department of Biochemistry, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Alexei Savchenko
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, Illinois, USA
| | - David R Andes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Timothy M Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
49
|
Ghosh S, Zheng M, He J, Wu Y, Zhang Y, Wang W, Shen J, Yeung KWK, Neelakantan P, Xu C, Qiao W. Electrically-driven drug delivery into deep cutaneous tissue by conductive microneedles for fungal infection eradication and protective immunity. Biomaterials 2025; 314:122908. [PMID: 39454504 DOI: 10.1016/j.biomaterials.2024.122908] [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: 07/13/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Fungal infections affect over 13 million people worldwide and are responsible for 1.5 million deaths annually. Some deep cutaneous fungal infections may extend the dermal barriers to cause systemic infection, resulting in substantial morbidity and mortality. However, the management of deep cutaneous fungal infection is challenging and yet overlooked by traditional treatments, which only offer limited drug availability within deep tissue. In this study, we have developed an electrically stimulated microneedle patch to deliver miconazole into the subcutaneous layer. We tested its antifungal efficacy using in vitro and ex vivo models that mimic fungal infection. Moreover, we confirmed its anti-fungal and wound-healing effects in a murine subcutaneous fungal infection model. Furthermore, our findings also showed that the combination of miconazole and applied current synergistically stimulated the nociceptive sensory nerves, thereby activating protective cutaneous immunity mediated by dermal dendritic and γδ-T cells. Collectively, this study provides a new strategy for minimally invasive delivery of therapeutic agents and the modulation of the neuro-immune axis in deep tissue.
Collapse
Affiliation(s)
- Sumanta Ghosh
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China; Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Mengjia Zheng
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Jiahui He
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Yefeng Wu
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yaming Zhang
- Department of Pharmacy and Pharmacology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Weiping Wang
- Department of Pharmacy and Pharmacology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jie Shen
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Kelvin W K Yeung
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Prasanna Neelakantan
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China; Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China.
| | - Wei Qiao
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
50
|
Gavandi TC, Basrani ST, Chougule SA, Patil SB, Nille OS, Kolekar GB, Yankanchi SR, Karuppayil SM, Jadhav AK. Vidarabine as a novel antifungal agent against Candida albicans: insights on mechanism of action. Int Microbiol 2025; 28:589-602. [PMID: 39126447 DOI: 10.1007/s10123-024-00565-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/11/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Around 1.5 million mortality cases due to fungal infection are reported annually, posing a massive threat to global health. However, the effectiveness of current antifungal therapies in the treatment of invasive fungal infections is limited. Repurposing existing antifungal drugs is an advisable alternative approach for enhancing their effectiveness. This study evaluated the antifungal efficacy of the antiviral drug vidarabine against Candida albicans ATCC 90028. Antifungal susceptibility testing was performed by microbroth dilution assay and further processed to find the minimum fungicidal concentration. Investigation on probable mode of vidarabine action against C. albicans was assessed by using the ergosterol reduction assay, reactive oxygen species (ROS) accumulation, nuclear condensation, and apoptosis assay. Results revealed that C. albicans was susceptible to vidarabine action and exhibited minimum inhibitory concentration at 150 µg/ml. At a concentration of 300 µg/ml, vidarabine had fungicidal activity against C. albicans. 300 µg/ml vidarabine-treated C. albicans cells demonstrated 91% reduced ergosterol content. Annexin/FITC/PI assay showed that vidarabine (150 µg/ml) had increased late apoptotic cells up to 31%. As per the fractional inhibitory concentration index, vidarabine had synergistic activity with fluconazole and caspofungin against this fungus. The mechanism underlying fungicidal action of vidarabine was evaluated at the intracellular level, and probably because of increased nuclear condensation, enhanced ROS generation, and cell cycle arrest. In conclusion, this data is the first to report that vidarabine has potential to be used as a repurposed antifungal agent alone or in combination with standard antifungal drugs, and could be a quick and safe addition to existing therapies for treating fungal infections.
Collapse
Affiliation(s)
- Tanjila C Gavandi
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416-003, Maharashtra, India
| | - Sargun T Basrani
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416-003, Maharashtra, India
| | - Sayali A Chougule
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416-003, Maharashtra, India
| | - Shivani B Patil
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416-003, Maharashtra, India
| | - Omkar S Nille
- Department of Chemistry, Shivaji University, Kolhapur, 416-004, Maharashtra, India
| | - Govind B Kolekar
- Department of Chemistry, Shivaji University, Kolhapur, 416-004, Maharashtra, India
| | | | - S Mohan Karuppayil
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416-003, Maharashtra, India.
| | - Ashwini K Jadhav
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416-003, Maharashtra, India.
| |
Collapse
|