1
|
Yadav V, Floyd Averette A, Upadhya R, Heitman J. Calcineurin controls the cytokinesis machinery during thermal stress in Cryptococcus deneoformans. Proc Natl Acad Sci U S A 2025; 122:e2503751122. [PMID: 40397671 DOI: 10.1073/pnas.2503751122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/18/2025] [Indexed: 05/23/2025] Open
Abstract
Calcineurin is a highly conserved phosphatase that plays a central role in sensing calcium and governing transcriptional, posttranscriptional, and posttranslational signaling networks. Calcineurin is a heterodimer consisting of a catalytic A subunit and a regulatory B subunit. Through downstream effectors, calcineurin signaling drives myriad responses in different organisms. In the fungal pathogenic Cryptococcus species complex that infects humans, calcineurin governs thermotolerance and is essential for growth at high temperature and pathogenesis. In Cryptococcus deneoformans, the underlying molecular functions of this critical signaling cascade are not well understood. In this study, we conducted a genetic screen and identified genetic changes that suppress the requirement for calcineurin during high-temperature growth. Our results identified two mechanisms that bypass the requirement for calcineurin function. The first mechanism involves segmental aneuploidy via both amplification as well as loss of chromosome fragments. The second mechanism involves dominant amino acid substitution mutations in the genes encoding three proteins, Chs6, Imp2, and Cts1, orthologs of components of the Ingression Progression Complex required for septation and budding in Saccharomyces cerevisiae. Loss of calcineurin activity causes chitin and chitosan accumulation and severe budding defects, whereas suppressor mutations largely restore growth and cytokinesis in the absence of calcineurin. These findings reveal that the calcineurin signaling cascade controls a conserved cytokinesis machinery at the mitotic exit network during thermal stress.
Collapse
Affiliation(s)
- Vikas Yadav
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| | - Anna Floyd Averette
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| | - Rajendra Upadhya
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
2
|
Kowalewski ME, Zagler S, Redinbo MR. Structural Insights into Selectively Targeting Candida albicans Hsp90. Biochemistry 2025. [PMID: 40397669 DOI: 10.1021/acs.biochem.5c00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
The threat of drug-resistant pathogens continues to rise and underscores the need for new antimicrobial and antifungal strategies. Diverse chemical scaffolds have been shown with high affinity to bind the human heat-shock protein Hsp90. Orthologous proteins are present in microbial pathogens and have been shown to be particularly abundant in these organisms, suggesting they may serve as therapeutic targets. Here, we examine the potency and selectivity of human Hsp90 ligands for their capacity to bind to the nucleotide binding domain of Hsp90 from the pathogenic fungi, Candida albicans. Using a series of biochemical, structural, and fragment and in silico screening investigations, we define key chemical features that lead to effective C. albicans Hsp90 (CaHsp90) binding. We support these studies with crystal structures of five diverse human Hsp90 ligands in complex with CaHsp90, as well as the structure of this protein with a nonhydrolyzable ATP analog. We demonstrate the structural basis for the selectivity of the human Hsp90 inhibitor TAS116 for CaHsp90, features that may be exploited in the future development of improved CaHsp90 inhibitors.
Collapse
Affiliation(s)
- Mark E Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Sebastian Zagler
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Matthew R Redinbo
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Microbiology and Immunology, and Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
3
|
Gomez-Artiguez L, de la Cámara-Fuentes S, Sun Z, Hernáez ML, Borrajo A, Pitarch A, Molero G, Monteoliva L, Moritz RL, Deutsch EW, Gil C. Candida albicans: A Comprehensive View of the Proteome. J Proteome Res 2025; 24:1636-1648. [PMID: 40084908 PMCID: PMC12123666 DOI: 10.1021/acs.jproteome.4c01020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/30/2025] [Accepted: 02/21/2025] [Indexed: 03/16/2025]
Abstract
We describe a new release of the Candida albicans PeptideAtlas proteomics spectral resource (build 2024-03), providing a sequence coverage of 79.5% at the canonical protein level, matched mass spectrometry spectra, and experimental evidence identifying 3382 and 536 phosphorylated serine and threonine sites with false localization rates of 1% and 5.3%, respectively. We provide a tutorial on how to use the PeptideAtlas and associated tools to access this information. The C. albicans PeptideAtlas summary web page provides "Build overview", "PTM coverage", "Experiment contribution", and "Data set contribution" information. The protein and peptide information can also be accessed via the Candida Genome Database via hyperlinks on each protein page. This allows users to peruse identified peptides, protein coverage, post-translational modifications (PTMs), and experiments that identify each protein. Given the value of understanding the PTM landscape in the sequence of each protein, a more detailed explanation of how to interpret and analyze PTM results is provided in the PeptideAtlas of this important pathogen. Candida albicans PeptideAtlas web page: https://db.systemsbiology.net/sbeams/cgi/PeptideAtlas/buildDetails?atlas_build_id=578.
Collapse
Affiliation(s)
- Leticia Gomez-Artiguez
- Microbiology
and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040Madrid, Spain
| | | | - Zhi Sun
- Institute
for Systems Biology, 401 Terry Ave North, Seattle, Washington98109, United States
| | - María Luisa Hernáez
- Proteomics
Unit, Faculty of Pharmacy, Complutense University
of Madrid, 28040Madrid, Spain
| | - Ana Borrajo
- Microbiology
and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040Madrid, Spain
| | - Aída Pitarch
- Microbiology
and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040Madrid, Spain
| | - Gloria Molero
- Microbiology
and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040Madrid, Spain
| | - Lucía Monteoliva
- Microbiology
and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040Madrid, Spain
| | - Robert L. Moritz
- Institute
for Systems Biology, 401 Terry Ave North, Seattle, Washington98109, United States
| | - Eric W. Deutsch
- Institute
for Systems Biology, 401 Terry Ave North, Seattle, Washington98109, United States
| | - Concha Gil
- Microbiology
and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040Madrid, Spain
- Proteomics
Unit, Faculty of Pharmacy, Complutense University
of Madrid, 28040Madrid, Spain
| |
Collapse
|
4
|
Amann V, Kissmann AK, Firacative C, Rosenau F. Biofilm-Associated Candidiasis: Pathogenesis, Prevalence, Challenges and Therapeutic Options. Pharmaceuticals (Basel) 2025; 18:460. [PMID: 40283897 PMCID: PMC12030374 DOI: 10.3390/ph18040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The rising prevalence of fungal infections, especially those caused by Candida species, presents a major risk to global health. With approximately 1.5 million deaths annually, the urgency for effective treatment options has never been greater. Candida spp. are the leading cause of invasive infections, significantly impacting immunocompromised patients and those in healthcare settings. C. albicans, C. parapsilosis and the emerging species C. auris are categorized as highly dangerous species because of their pathogenic potential and increasing drug resistance. This review comparatively describes the formation of microbial biofilms of both bacterial and fungal origin, including major pathogens, thereby creating a novel focus. Biofilms can further complicate treatment, as these structures provide enhanced resistance to antifungal therapies. Traditional antifungal agents, including polyenes, azoles and echinocandins, have shown effectiveness, yet resistance development continues to rise, necessitating the exploration of novel therapeutic approaches. Antimicrobial peptides (AMPs) such as the anti-biofilm peptides Pom-1 and Cm-p5 originally isolated from snails represent promising candidates due to their unique mechanisms of action and neglectable cytotoxicity. This review article discusses the challenges posed by Candida infections, the characteristics of important species, the role of biofilms in virulence and the potential of new therapeutic options like AMPs.
Collapse
Affiliation(s)
- Valerie Amann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Ann-Kathrin Kissmann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Carolina Firacative
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogota 111221, Colombia;
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| |
Collapse
|
5
|
Yue D, Zheng D, Yang L, Bai Y, Song Z, Li D, Yu X, Li Y. Berberine disrupts the high-affinity iron transport system to reverse the fluconazole-resistance in Candida albicans. Microb Pathog 2025; 200:107370. [PMID: 39929396 DOI: 10.1016/j.micpath.2025.107370] [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: 10/10/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Invasive fungal infection is usually caused by Candida albicans infection, which has a high incidence rate and mortality in critically ill patients. New drugs are needed to combat this pathogen since the limited treatment options currently available and increasing resistance to existing drugs. Berberine (BBR) is an active compound in Coptis chinensis, Phellodendron chinense and Radix berberidis, which is clinically used to treat inflammatory bowel disease, but its inhibitory effect on drug-resistant fungi has not been clarified. In this study, based on the evidence of BBR inhibiting the expression of azole-resistance genes, reducing cell adhesion and disrupting biofilm formation, transcriptome analysis revealed that the disruption of iron acquisition pathway may be the core link in BBR inhibiting drug-resistant fungi. Combined with the subsequent experimental results, including the reduction of intracellular ferrous ion content, the weakening of iron reductase activity and the overall downregulation of the coding gene of the high-affinity iron reduction system, it is speculated that the fungal growth defect under BBR treatment is the result of the interruption of the high-affinity iron acquisition pathway. Ftr1 plays a central role in the drug targeting of this transport system. Meanwhile, due to the iron deficiency within the cell, the biological function of mitochondria is impaired, ultimately leading to fungal death. This study not only reflects the application value of BBR in the clinical treatment of fungal infections, but also provides a potential strategy to address the current drug-resistance dilemma.
Collapse
Affiliation(s)
- Daifan Yue
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dongming Zheng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Nuclear Medicine, Ya'an People's Hospital, Ya'an, 625000, China
| | - Linlan Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuxin Bai
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhen Song
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dongmei Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaoqin Yu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yan Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
6
|
Xiong J, Lu H, Jiang Y. Mechanisms of Azole Potentiation: Insights from Drug Repurposing Approaches. ACS Infect Dis 2025; 11:305-322. [PMID: 39749640 DOI: 10.1021/acsinfecdis.4c00657] [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: 01/04/2025]
Abstract
The emergence of azole resistance and tolerance in pathogenic fungi has emerged as a significant public health concern, emphasizing the urgency for innovative strategies to bolster the efficacy of azole-based treatments. Drug repurposing stands as a promising and practical avenue for advancing antifungal therapy, with the potential for swift clinical translation. This review offers a comprehensive overview of azole synergistic agents uncovered through drug repurposing strategies, alongside an in-depth exploration of the mechanisms by which these agents augment azole potency. Drawing from these mechanisms, we delineate strategies aimed at enhancing azole effectiveness, such as inhibiting efflux pumps to elevate azole concentrations within fungal cells, intensifying ergosterol synthesis inhibition, mitigating fungal cell resistance to azoles, and disrupting biological processes extending beyond ergosterol synthesis. This review is beneficial for the development of these potentiators, as it meticulously examines instances and provides nuanced discussions on the mechanisms underlying the progression of azole potentiators through drug repurposing strategies.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
7
|
Su Y, Li Y, Yi Q, Xu Y, Sun T, Li Y. Insight into the Mechanisms and Clinical Relevance of Antifungal Heteroresistance. J Fungi (Basel) 2025; 11:143. [PMID: 39997437 PMCID: PMC11856953 DOI: 10.3390/jof11020143] [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: 01/04/2025] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Antifungal resistance poses a critical global health threat, particularly in immuno-compromised patients. Beyond the traditional resistance mechanisms rooted in heritable and stable mutations, a distinct phenomenon known as heteroresistance has been identified, wherein a minority of resistant fungal cells coexist within a predominantly susceptible population. Heteroresistance may be induced by pharmacological factors or non-pharmacological agents. The reversible nature of it presents significant clinical challenges, as it can lead to undetected resistance during standard susceptibility testing. As heteroresistance allows fungal pathogens to survive antifungal treatment, this adaptive strategy often leads to treatment failure and recurring infection. Though extensively studied in bacteria, limited research has explored its occurrence in fungi. This review summarizes the current findings on antifungal heteroresistance mechanisms, highlighting the clinical implications of fungal heteroresistance and the pressing need for deeper mechanism insights. We aim to bring together the latest research advances in the field of antifungal heteroresistance, summarizing in detail its known characteristics, inducing factors, molecular mechanisms, and clinical significance, and describing the similarities and differences between heteroresistance, tolerance and persistence. Further research is needed to understand this phenomenon and develop more effective antifungal therapies to combat fungal infections.
Collapse
Affiliation(s)
- Yanyu Su
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- Graduate School, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yi Li
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Qiaolian Yi
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Tianshu Sun
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- Clinical Biobank, Center for Biomedical Technology, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yingxing Li
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
8
|
Zhang M, Zhao Y, Cui H, Huang W, Xiong K, Yang S, Duan Y, He Y, Yang L, Su C, Lu Y. CO 2 potentiates echinocandin efficacy during invasive candidiasis therapy via dephosphorylation of Hsp90 by Ptc2 in condensates. Proc Natl Acad Sci U S A 2025; 122:e2417721122. [PMID: 39908105 PMCID: PMC11831212 DOI: 10.1073/pnas.2417721122] [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: 08/30/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025] Open
Abstract
Carbon dioxide is a signaling cue critical for fungal pathogenesis. Ptc2, a type 2C protein phosphatase (PP2C), serves as a conserved CO2 sensor in fungi. By combining phosphoproteomic and biochemical assays, we identified Hsp90 as a direct target of Ptc2 at host CO2 concentrations and Ssb1 as a Ptc2 target protein regardless of CO2 levels in Candida albicans, the most prevalent human fungal pathogen. Ptc2 forms reversible condensates at elevated CO2, which enables the recruitment of Hsp90, but not Ssb1, to condensates, allowing efficient dephosphorylation. This process confers an enhanced susceptibility to caspofungin in vitro and during in vivo infection therapy. Importantly, we demonstrate this phenomenon in non-albicans Candida species. Sequential passages of C. albicans in mice with caspofungin treatment readily induce in vivo drug tolerance, causing therapeutic failure. These evolved strains display increased resistance to caspofungin under host concentrations of CO2 but remain susceptible in air. Collectively, our study reveals a profound impact of host concentrations of CO2 on antifungal drug susceptibility and connects this phenotype to therapeutic outcomes and highlights condensate formation as an efficient means that enables selective recruitment of substrates for certain signaling events.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei230032, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei230032, China
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan430072, China
| | - Youzhi Zhao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan430072, China
| | - Hao Cui
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan430072, China
| | - Wenqiang Huang
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei230032, China
| | - Kang Xiong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan430072, China
| | - Shan Yang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Yuanyuan Duan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan430072, China
| | - Yong He
- Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei230032, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei230032, China
| | - Lianjuan Yang
- Shanghai Dermatology Hospital, School of Medicine, Tongji University, Shanghai200443, China
| | - Chang Su
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Yang Lu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan430072, China
| |
Collapse
|
9
|
Gomez-Artiguez L, de la Cámara-Fuentes S, Sun Z, Hernáez ML, Borrajo A, Pitarch A, Molero G, Monteoliva L, Moritz RL, Deutsch EW, Gil C. Candida albicans: a comprehensive view of the proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.20.629377. [PMID: 39763837 PMCID: PMC11702768 DOI: 10.1101/2024.12.20.629377] [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] [Indexed: 01/15/2025]
Abstract
We describe a new release of the Candida albicans PeptideAtlas proteomics spectral resource (build 2024-03), providing a sequence coverage of 79.5% at the canonical protein level, matched mass spectrometry spectra, and experimental evidence identifying 3382 and 536 phosphorylated serine and threonine sites with false localization rates of 1% and 5.3%, respectively. We provide a tutorial on how to use the PeptideAtlas and associated tools to access this information. The C. albicans PeptideAtlas summary web page provides "Build overview", "PTM coverage", "Experiment contribution", and "Dataset contribution" information. The protein and peptide information can also be accessed via the Candida Genome Database via hyperlinks on each protein page. This allows users to peruse identified peptides, protein coverage, post-translational modifications (PTMs), and experiments identifying each protein. Given the value of understanding the PTM landscape in the sequence of each protein, a more detailed explanation of how to interpret and analyse PTM results is provided in the PeptideAtlas of this important pathogen. Candida albicans PeptideAtlas web page: https://db.systemsbiology.net/sbeams/cgi/PeptideAtlas/buildDetails?atlas_build_id=578.
Collapse
Affiliation(s)
- Leticia Gomez-Artiguez
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| | | | - Zhi Sun
- Institute for Systems Biology, 401 Terry Ave North, Seattle, WA, USA. 98109
| | - María Luisa Hernáez
- Proteomics Unit, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| | - Ana Borrajo
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| | - Aída Pitarch
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| | - Gloria Molero
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| | - Lucía Monteoliva
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| | - Robert L. Moritz
- Institute for Systems Biology, 401 Terry Ave North, Seattle, WA, USA. 98109
| | - Eric W. Deutsch
- Institute for Systems Biology, 401 Terry Ave North, Seattle, WA, USA. 98109
| | - Concha Gil
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
- Proteomics Unit, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid
| |
Collapse
|
10
|
Bende G, Zsindely N, Laczi K, Kristóffy Z, Papp C, Farkas A, Tóth L, Sáringer S, Bodai L, Rákhely G, Marx F, Galgóczy L. The Neosartorya (Aspergillus) fischeri antifungal protein NFAP2 has low potential to trigger resistance development in Candida albicans in vitro. Microbiol Spectr 2025; 13:e0127324. [PMID: 39560388 PMCID: PMC11705825 DOI: 10.1128/spectrum.01273-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Due to the increase in the number of drug-resistant Candida albicans strains, new antifungal compounds with limited potential for the development of resistance are urgently needed. NFAP2, an antifungal protein (AFP) secreted by Neosartorya (Aspergillus) fischeri, is a promising candidate. We investigated the ability of C. albicans to develop resistance to NFAP2 in a microevolution experiment compared with generic fluconazole (FLC). C. albicans adapted to only 1× minimum inhibitory concentration (MIC) of NFAP2, which can be considered tolerance rather than resistance, compared with 32× MIC of FLC. Genome analysis revealed non-silent mutations in only two genes in NFAP2-tolerant strains and in several genes in FLC-resistant strains. Tolerance development to NFAP2 did not influence cell morphology. The susceptibility of NFAP2-tolerant strains did not change to FLC, amphotericin B, micafungin, and terbinafine. These strains did not show altered susceptibility to AFPs from Penicillium chrysogenum, except one which had less susceptibility to Penicillium chrysogenum antifungal protein B. FLC-resistant strains had decreased susceptibility to terbinafine and NFAP2, but not to other drugs and AFPs from P. chrysogenum. NFAP2-tolerant and FLC-resistant strains showed decreased and increased NFAP2 binding and uptake, respectively. The development of tolerance to NFAP2 decreased tolerance to cell wall, heat, and UV stresses. The development of FLC resistance increased tolerance to cell wall stress and decreased tolerance to heat and UV stresses. Tolerance to NFAP2 did not have significant metabolic fitness cost and could not increase virulence, compared with resistance to FLC.IMPORTANCEDue to the increasing number of (multi)drug-resistant strains, only a few effective antifungal drugs are available to treat infections caused by opportunistic Candida species. Therefore, the incidence of hard-to-treat candidiasis has increased dramatically in the past decade, and the demand to identify antifungal compounds with minimal potential to trigger resistance is substantial. The features of NFAP2 make it a promising candidate for the topical treatment of Candida infection. Data on the development of resistance to antifungal proteins in Candida albicans are lacking. In this study, we provide evidence that NFAP2 has a low potential to trigger resistance in C. albicans in vitro, and the developed tolerance to NFAP2 is not associated with severe phenotypic changes compared with development of resistance to generic fluconazole. These results suggest the slow emergence of NFAP2-resistant Candida strains, and NFAP2 can reliably be used long-term in the clinic.
Collapse
Affiliation(s)
- Gábor Bende
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Nóra Zsindely
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Krisztián Laczi
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Plant Biology, HUN-REN Biological Research Center, Szeged, Hungary
| | - Zsolt Kristóffy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Csaba Papp
- Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Attila Farkas
- Institute of Plant Biology, HUN-REN Biological Research Center, Szeged, Hungary
| | - Liliána Tóth
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Szabolcs Sáringer
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gábor Rákhely
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biophysics, HUN-REN Biological Research Center, Szeged, Hungary
| | - Florentine Marx
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, HUN-REN Biological Research Center, Szeged, Hungary
| |
Collapse
|
11
|
Jay A, Jordan DF, Gerstein A, Landry CR. The role of gene copy number variation in antimicrobial resistance in human fungal pathogens. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:1. [PMID: 39781035 PMCID: PMC11703754 DOI: 10.1038/s44259-024-00072-1] [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: 09/25/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
Faced with the burden of increasing resistance to antifungals in many fungal pathogens and the constant emergence of new drug-resistant strains, it is essential to assess the importance of various resistance mechanisms. Fungi have relatively plastic genomes and can tolerate genomic copy number variation (CNV) caused by aneuploidy and gene amplification or deletion. In many cases, these genomic changes lead to adaptation to stressful conditions, including those caused by antifungal drugs. Here, we specifically examine the contribution of CNVs to antifungal resistance. We undertook a thorough literature search, collecting reports of antifungal resistance caused by a CNV, and classifying the examples of CNV-conferred resistance into four main mechanisms. We find that in human fungal pathogens, there is little evidence that gene copy number plays a major role in the emergence of antifungal resistance compared to other types of mutations. We discuss why we might be underestimating their importance and new approaches being used to study them.
Collapse
Affiliation(s)
- Adarsh Jay
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec City, G1V 0A6 Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec City, G1V 0A6 Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Université Laval, Québec City, G1V 0A6 Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec City, G1V 0A6 Canada
| | - David F. Jordan
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec City, G1V 0A6 Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec City, G1V 0A6 Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Université Laval, Québec City, G1V 0A6 Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec City, G1V 0A6 Canada
| | - Aleeza Gerstein
- Department of Microbiology, The University of Manitoba, Winnipeg, R3T 2N2 Canada
- Department of Statistics, The University of Manitoba, Winnipeg, R3T 2N2 Canada
| | - Christian R. Landry
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec City, G1V 0A6 Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec City, G1V 0A6 Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Université Laval, Québec City, G1V 0A6 Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec City, G1V 0A6 Canada
| |
Collapse
|
12
|
Zobi C, Algul O. The Significance of Mono- and Dual-Effective Agents in the Development of New Antifungal Strategies. Chem Biol Drug Des 2025; 105:e70045. [PMID: 39841631 PMCID: PMC11753615 DOI: 10.1111/cbdd.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/25/2024] [Accepted: 01/04/2025] [Indexed: 01/24/2025]
Abstract
Invasive fungal infections (IFIs) pose significant challenges in clinical settings, particularly due to their high morbidity and mortality rates. The rising incidence of these infections, coupled with increasing antifungal resistance, underscores the urgent need for novel therapeutic strategies. Current antifungal drugs target the fungal cell membrane, cell wall, or intracellular components, but resistance mechanisms such as altered drug-target interactions, enhanced efflux, and adaptive cellular responses have diminished their efficacy. Recent research has highlighted the potential of dual inhibitors that simultaneously target multiple pathways or enzymes involved in fungal growth and survival. Combining pharmacophores, such as lanosterol 14α-demethylase (CYP51), heat shock protein 90 (HSP90), histone deacetylase (HDAC), and squalene epoxidase (SE) inhibitors, has led to the development of compounds with enhanced antifungal activity and reduced resistance. This dual-target approach, along with novel chemical scaffolds, not only represents a promising strategy for combating antifungal resistance but is also being utilized in the development of anticancer agents. This review explores the development of new antifungal agents that employ mono-, dual-, or multi-target strategies to combat IFIs. We discuss emerging antifungal targets, resistance mechanisms, and innovative therapeutic approaches that offer hope in managing these challenging infections.
Collapse
Affiliation(s)
- Cengiz Zobi
- Department of Pharmaceutical Chemistry, Faculty of PharmacyErzincan Binali Yildirim UniversityErzincanTurkiye
- Department of İliç Dursun Yildirim MYOErzincan Binali Yildirim UniversityErzincanTurkiye
| | - Oztekin Algul
- Department of Pharmaceutical Chemistry, Faculty of PharmacyErzincan Binali Yildirim UniversityErzincanTurkiye
- Department of Pharmaceutical Chemistry, Faculty of PharmacyMersin UniversityMersinTurkiye
| |
Collapse
|
13
|
Fang T, Xiong J, Huang X, Fang X, Shen X, Jiang Y, Lu H. Extracellular Hsp90 of Candida albicans contributes to the virulence of the pathogen by activating the NF-κB signaling pathway and inducing macrophage pyroptosis. Microbiol Res 2025; 290:127964. [PMID: 39522202 DOI: 10.1016/j.micres.2024.127964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Strategies aimed at targeting fungal extracellular heat shock protein 90 (eHsp90) using vaccines and antibodies have demonstrated encouraging potential in the prevention and management of invasive fungal diseases (IFDs). However, the precise underlying mechanism by which eHsp90 contributes to the heightened virulence of Candida albicans (C. albicans) remains an enigma, awaiting further elucidation. In our current research, we have found that the 47-kDa fragment of C. albicans Hsp90 (CaHsp90), which serves as the primary antigenic determinant, is not degraded within C. albicans cells. Moreover, we have discovered that extracellular CaHsp90 (eCaHsp90) is derived from the components of lysed C. albicans cells. We also generated recombinant CaHsp90 in Escherichia coli, and found that eCaHsp90 spreads beyond the initial C. albicans colonization site, thereby enhancing the overall virulence of the organism. Our results further clarify that eCaHsp90 activates the nuclear factor kappa-B (NF-κB) signaling pathway and upregulates the expression of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3). This upregulation results in the activation of Gasdermin D (GSDMD) and subsequent macrophage pyroptosis, ultimately increasing the virulence of C. albicans. This study provides valuable insights into the mechanism by which eCaHsp90 contributes to the virulence of C. albicans, offering a pharmacological basis for antifungal strategies targeting fungal eHsp90.
Collapse
Affiliation(s)
- Ting Fang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Juan Xiong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xin Huang
- Department of Dermatology, Hair Medical Center of Shanghai Tongji Hospital, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xinyu Fang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xuqing Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
14
|
Balla N, Kovács F, Tóth Z, Harmath A, Bozó A, Majoros L, Kovács R, Jakab Á. Isolate Specific Transcriptome Changes Exerted by Isavuconazole Treatment in Candida auris. Mycopathologia 2024; 190:5. [PMID: 39729249 DOI: 10.1007/s11046-024-00919-1] [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: 08/25/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
The sudden emergence of multidrug- and pan-resistant Candida auris isolates, combined with limited treatment options, poses significant global challenges in healthcare settings. Combination based therapies are promising alternative options to overcome C. auris related infections, where echinocandin and isavuconazole (ISA) combinations may be an interesting and promising approach. Understanding the molecular mechanisms underlying ISA treatment is crucial for developing novel therapeutic recommendations. Therefore, we investigated the gene transcription profiles of non-wild type (non-WT) and wild type (WT) C. auris isolates from the South Asian clade following ISA exposure using total RNA sequencing. The non-WT isolate was classified according to the previously reported tentative epidemiological cut-off value of ≤ 1 mg/L. ISA treatment resulted in the upregulation of 158 and 134 genes and the downregulation of 119 and 96 genes in the non-WT and WT isolates, respectively, compared with untreated samples. In general, ISA-treated isolates exhibited increased transcription of the transcriptional factor UPC2, the drug transporter MDR1, vacuolar calcium-ATPase PMC1, and several ergosterol biosynthesis genes. The WT isolate showed pronounced enrichment of genes involved in sphingolipid biosynthesis, adhesion, and drug transport. These findings suggest that alterations in membrane lipid composition and modulation of drug efflux transporters are critical processes contributing to ISA susceptibility in case of WT isolates.
Collapse
Affiliation(s)
- Noémi Balla
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Zoltán Tóth
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary
| | - Andrea Harmath
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Aliz Bozó
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary
| | - László Majoros
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary
| | - Renátó Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary.
| | - Ágnes Jakab
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98., Debrecen, 4032, Hungary.
| |
Collapse
|
15
|
Stevens I, Silao FG, Huch S, Liu H, Ryman K, Carvajal-Jimenez A, Ljungdahl PO, Pelechano V. The early transcriptional and post-transcriptional responses to fluconazole in sensitive and resistant Candida albicans. Sci Rep 2024; 14:29012. [PMID: 39578617 PMCID: PMC11586853 DOI: 10.1038/s41598-024-80435-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024] Open
Abstract
Candida albicans is a leading cause of fungal infections in immunocompromised patients. Management of candidemia relies on a few antifungal agents, with fluconazole being first line therapy. The emergence of fluconazole-resistant strains highlights the pressing need to improve our molecular understanding of the drug response mechanisms. By sequencing the 5'P mRNA degradation intermediates, we establish that co-translational mRNA decay occurs in C. albicans and characterize how in vivo 5´-3´ exonuclease degradation trails the last translating ribosome. Thus, the study of the 5' Phosphorylated mRNA degradome (5PSeq) offers a simple and affordable way to measure ribosome dynamics and identify codon specific ribosome stalls in response to drugs and amino acid deprivation. Building upon this, we combine RNA-Seq and 5PSeq to study the early response of sensitive and resistant C. albicans isolates to fluconazole. Our results highlight that transcriptional responses, rather than changes in ribosome dynamics, are the main driver of Candida resistance to fluconazole.
Collapse
Affiliation(s)
- Irene Stevens
- Science for Life Laboratory (SciLifeLab), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Fitz Gerald Silao
- SciLifeLab, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Susanne Huch
- Science for Life Laboratory (SciLifeLab), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Honglian Liu
- Science for Life Laboratory (SciLifeLab), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Kicki Ryman
- SciLifeLab, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Adriana Carvajal-Jimenez
- Science for Life Laboratory (SciLifeLab), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Per O Ljungdahl
- SciLifeLab, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Vicent Pelechano
- Science for Life Laboratory (SciLifeLab), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
16
|
Yiu B, Robbins N, Cowen LE. Interdisciplinary approaches for the discovery of novel antifungals. Trends Mol Med 2024; 30:723-735. [PMID: 38777733 PMCID: PMC11987087 DOI: 10.1016/j.molmed.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/10/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Pathogenic fungi are an increasing public health concern. The emergence of antifungal resistance coupled with the scarce antifungal arsenal highlights the need for novel therapeutics. Fortunately, the past few years have witnessed breakthroughs in antifungal development. Here, we discuss pivotal interdisciplinary approaches for the discovery of novel compounds with efficacy against diverse fungal pathogens. We highlight breakthroughs in improving current antifungal scaffolds, as well as the utility of compound combinations to extend the lifespan of antifungals. Finally, we describe efforts to refine candidate chemical scaffolds by leveraging structure-guided approaches, and the use of functional genomics to expand our knowledge of druggable antifungal targets. Overall, we emphasize the importance of interdisciplinary collaborations in the endeavor to develop innovative antifungal strategies.
Collapse
Affiliation(s)
- Bonnie Yiu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada.
| |
Collapse
|
17
|
Zheng L, Xu Y, Wang C, Guo L. Ketoconazole induces reversible antifungal drug tolerance mediated by trisomy of chromosome R in Candida albicans. Front Microbiol 2024; 15:1450557. [PMID: 39139375 PMCID: PMC11319258 DOI: 10.3389/fmicb.2024.1450557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Background The emergence of tolerance to antifungal agents in Candida albicans complicates the treatment of fungal infections. Understanding the mechanisms underlying this tolerance is crucial for developing effective therapeutic strategies. Objective This study aims to elucidate the genetic and molecular basis of ketoconazole tolerance in C. albicans, focusing on the roles of chromosomal aneuploidy, Hsp90, and calcineurin. Methods The wild-type C. albicans strain SC5314 was exposed to increasing concentrations of ketoconazole (0.015-32 μg/mL) to select for tolerant adaptors. Disk diffusion and spot assays were used to assess tolerance. Whole-genome sequencing identified chromosomal changes in the adaptors. The roles of Hsp90 and calcineurin in maintaining and developing ketoconazole tolerance were investigated using specific inhibitors and knockout strains. Results Adaptors exhibited tolerance to ketoconazole concentrations up to 16 μg/mL, a significant increase from the parent strain's inhibition at 0.015 μg/mL. All tolerant adaptors showed amplification of chromosome R, with 29 adaptors having trisomy and one having tetrasomy. This aneuploidy was unstable, reverting to euploidy and losing tolerance in drug-free conditions. Both Hsp90 and calcineurin were essential for maintaining and developing ketoconazole tolerance. Inhibition of these proteins resulted in loss of tolerance. The efflux gene CDR1 was not required for the development of tolerance. Chromosome R trisomy and tetrasomy induce cross-tolerance to other azole antifungal agents, including clotrimazole and miconazole, but not to other antifungal classes, such as echinocandins and pyrimidines, exemplified by caspofungin and 5-flucytosine. Conclusion Ketoconazole tolerance in C. albicans is mediated by chromosomal aneuploidy, specifically chromosome R amplification, and requires Hsp90 and calcineurin. These findings highlight potential targets for therapeutic intervention to combat antifungal tolerance and improve treatment outcomes.
Collapse
Affiliation(s)
- Lijun Zheng
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Xu
- Department of Pharmacy, The 960th Hospital of PLA, Jinan, China
| | - Chen Wang
- Department of Pharmacy, The 960th Hospital of PLA, Jinan, China
| | - Liangsheng Guo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
18
|
Xiong J, Wang L, Feng Y, Zhen C, Hang S, Yu J, Lu H, Jiang Y. Geldanamycin confers fungicidal properties to azole by triggering the activation of succinate dehydrogenase. Life Sci 2024; 348:122699. [PMID: 38718854 DOI: 10.1016/j.lfs.2024.122699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/17/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
AIMS Azoles have been widely employed for the treatment of invasive fungal diseases; however, their efficacy is diminished as pathogenic fungi tolerate them due to their fungistatic properties. Geldanamycin (GdA) can render azoles fungicidal by inhibiting the ATPase and molecular chaperone activities of heat shock protein 90 (Hsp90). Nonetheless, the clinical applicability of GdA is restricted due to its cytotoxic ansamycin scaffold structure, its induction of cytoprotective heat shock responses, and the conservative nature of Hsp90. Hence, it is imperative to elucidate the mechanism of action of GdA to confer fungicidal properties to azoles and mitigate the toxic adverse effects associated with GdA. MATERIALS AND METHODS Through various experimental methods, including the construction of gene-deleted Candida albicans mutants, in vitro drug sensitivity experiments, Western blot analysis, reactive oxygen species (ROS) assays, and succinate dehydrogenase activity assays, we identified Hsp90 client proteins associated with the tolerance of C. albicans to azoles. KEY FINDINGS It was observed that GdA effectively hindered the entry of Hsp90 into mitochondria, resulting in the alleviation of inhibitory effect of Hsp90 on succinate dehydrogenase. Consequently, the activation of succinate dehydrogenase led to an increased production of ROS. within the mitochondria, thereby facilitating the antifungal effects of azoles against C. albicans. SIGNIFICANCE This research presents a novel approach for conferring fungicidal properties to azoles, which involves specifically disrupting the interaction of between Hsp90 and succinate dehydrogenase rather than employing a non-specific inhibition of ATPase activity of Hsp90.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Li Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Zhen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Sijin Hang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jinhua Yu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
19
|
Dladla M, Gyzenhout M, Marias G, Ghosh S. Azole resistance in Aspergillus fumigatus- comprehensive review. Arch Microbiol 2024; 206:305. [PMID: 38878211 DOI: 10.1007/s00203-024-04026-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Aspergillus fumigatus is a ubiquitous filamentous fungus commonly found in the environment. It is also an opportunistic human pathogen known to cause a range of respiratory infections, such as invasive aspergillosis, particularly in immunocompromised individuals. Azole antifungal agents are widely used for the treatment and prophylaxis of Aspergillus infections due to their efficacy and tolerability. However, the emergence of azole resistance in A. fumigatus has become a major concern in recent years due to their association with increased treatment failures and mortality rates. The development of azole resistance in A. fumigatus can occur through both acquired and intrinsic mechanisms. Acquired resistance typically arises from mutations in the target enzyme, lanosterol 14-α-demethylase (Cyp51A), reduces the affinity of azole antifungal agents for the enzyme, rendering them less effective, while intrinsic resistance refers to a natural resistance of certain A. fumigatus isolates to azole antifungals due to inherent genetic characteristics. The current review aims to provide a comprehensive overview of azole antifungal resistance in A. fumigatus, discusses underlying resistance mechanisms, including alterations in the target enzyme, Cyp51A, and the involvement of efflux pumps in drug efflux. Impact of azole fungicide uses in the environment and the spread of resistant strains is also explored.
Collapse
Affiliation(s)
- Mthokozisi Dladla
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa
| | - Marieka Gyzenhout
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa.
| | - Gert Marias
- Department of Plant Sciences, Division of Plant Pathology, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, South Africa
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa.
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Birkat Al Mawz, Oman.
| |
Collapse
|
20
|
da Silva CM, de Lima Neto RG, de Carvalho AMR, Macêdo DPC, de Azevedo Melo AS, Neves RP. Taxonomy of Candida parapsilosis complex isolated from neonates and the role of Hsp90 inhibitors to enhanced the antifungal activity of micafungin. Lett Appl Microbiol 2024; 77:ovae044. [PMID: 38658187 DOI: 10.1093/lambio/ovae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/25/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
Species from Candida parapsilosis complex are frequently found in neonatal candidemia. The antifungal agents to treat this infection are limited and the occurrence of low in vitro susceptibility to echinocandins such as micafungin has been observed. In this context, the chaperone Hsp90 could be a target to reduce resistance. Thus, the objective of this research was to identify isolates from the C. parapsilosis complex and verify the action of Hsp90 inhibitors associated with micafungin. The fungal identification was based on genetic sequencing and mass spectrometry. Minimal inhibitory concentrations were determined by broth microdilution method according to Clinical Laboratory and Standards Institute. The evaluation of the interaction between micafungin with Hsp90 inhibitors was realized using the checkerboard methodology. According to the polyphasic taxonomy, C. parapsilosis sensu stricto was the most frequently identified, followed by C. orthopsilosis and C. metapsilosis, and one isolate of Lodderomyces elongisporus was identified by genetic sequencing. The Hsp90 inhibitor geladanamycin associated with micafungin showed a synergic effect in 31.25% of the isolates, a better result was observed with radicicol, which shows synergic effect in 56.25% tested yeasts. The results obtained demonstrate that blocking Hsp90 could be effective to reduce antifungal resistance to echinocandins.
Collapse
Affiliation(s)
| | | | | | | | | | - Rejane Pereira Neves
- Federal University of Pernambuco, Mycology Department, Recife-PE, 50670-90, Brazil
| |
Collapse
|
21
|
Cosio T, Pica F, Fontana C, Pistoia ES, Favaro M, Valsecchi I, Zarabian N, Campione E, Botterel F, Gaziano R. Stephanoascus ciferrii Complex: The Current State of Infections and Drug Resistance in Humans. J Fungi (Basel) 2024; 10:294. [PMID: 38667965 PMCID: PMC11050938 DOI: 10.3390/jof10040294] [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: 01/07/2024] [Revised: 03/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
In recent years, the incidence of fungal infections in humans has increased dramatically, accompanied by an expansion in the number of species implicated as etiological agents, especially environmental fungi never involved before in human infection. Among fungal pathogens, Candida species are the most common opportunistic fungi that can cause local and systemic infections, especially in immunocompromised individuals. Candida albicans (C. albicans) is the most common causative agent of mucosal and healthcare-associated systemic infections. However, during recent decades, there has been a worrying increase in the number of emerging multi-drug-resistant non-albicans Candida (NAC) species, i.e., C. glabrata, C. parapsilosis, C. tropicalis, C. krusei, C. auris, and C. ciferrii. In particular, Candida ciferrii, also known as Stephanoascus ciferrii or Trichomonascus ciferrii, is a heterothallic ascomycete yeast-like fungus that has received attention in recent decades as a cause of local and systemic fungal diseases. Today, the new definition of the S. ciferrii complex, which consists of S. ciferrii, Candida allociferrii, and Candida mucifera, was proposed after sequencing the 18S rRNA gene. Currently, the S. ciferrii complex is mostly associated with non-severe ear and eye infections, although a few cases of severe candidemia have been reported in immunocompromised individuals. Low susceptibility to currently available antifungal drugs is a rising concern, especially in NAC species. In this regard, a high rate of resistance to azoles and more recently also to echinocandins has emerged in the S. ciferrii complex. This review focuses on epidemiological, biological, and clinical aspects of the S. ciferrii complex, including its pathogenicity and drug resistance.
Collapse
Affiliation(s)
- Terenzio Cosio
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.P.); (E.S.P.); (M.F.); (R.G.)
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy;
| | - Francesca Pica
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.P.); (E.S.P.); (M.F.); (R.G.)
| | - Carla Fontana
- Laboratory of Microbiology and BioBank, National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.C.S., 00149 Rome, Italy;
| | - Enrico Salvatore Pistoia
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.P.); (E.S.P.); (M.F.); (R.G.)
| | - Marco Favaro
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.P.); (E.S.P.); (M.F.); (R.G.)
| | - Isabel Valsecchi
- DYNAMYC 7380, Faculté de Santé, Université Paris-Est Créteil (UPEC), 94010 Créteil, France; (I.V.); (F.B.)
| | - Nikkia Zarabian
- School of Medicine and Health Sciences, George Washington University, 2300 I St NW, Washington, DC 20052, USA
| | - Elena Campione
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy;
| | - Françoise Botterel
- DYNAMYC 7380, Faculté de Santé, Université Paris-Est Créteil (UPEC), 94010 Créteil, France; (I.V.); (F.B.)
| | - Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.P.); (E.S.P.); (M.F.); (R.G.)
| |
Collapse
|
22
|
Fallah S, Duncan D, Reichl KD, Smith MJ, Wang W, Porco JA, Brown LE, Whitesell L, Robbins N, Cowen LE. A chemical screen identifies structurally diverse metal chelators with activity against the fungal pathogen Candida albicans. Microbiol Spectr 2024; 12:e0409523. [PMID: 38376363 PMCID: PMC10986608 DOI: 10.1128/spectrum.04095-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/25/2024] [Indexed: 02/21/2024] Open
Abstract
Candida albicans, one of the most prevalent human fungal pathogens, causes diverse diseases extending from superficial infections to deadly systemic mycoses. Currently, only three major classes of antifungal drugs are available to treat systemic infections: azoles, polyenes, and echinocandins. Alarmingly, the efficacy of these antifungals against C. albicans is hindered both by basal tolerance toward the drugs and the development of resistance mechanisms such as alterations of the drug's target, modulation of stress responses, and overexpression of efflux pumps. Thus, the need to identify novel antifungal strategies is dire. To address this challenge, we screened 3,049 structurally-diverse compounds from the Boston University Center for Molecular Discovery (BU-CMD) chemical library against a C. albicans clinical isolate and identified 17 molecules that inhibited C. albicans growth by >80% relative to controls. Among the most potent compounds were CMLD013360, CMLD012661, and CMLD012693, molecules representing two distinct chemical scaffolds, including 3-hydroxyquinolinones and a xanthone natural product. Based on structural insights, CMLD013360, CMLD012661, and CMLD012693 were hypothesized to exert antifungal activity through metal chelation. Follow-up investigations revealed all three compounds exerted antifungal activity against non-albicans Candida, including Candida auris and Candida glabrata, with the xanthone natural product CMLD013360 also displaying activity against the pathogenic mould Aspergillus fumigatus. Media supplementation with metallonutrients, namely ferric or ferrous iron, rescued C. albicans growth, confirming these compounds act as metal chelators. Thus, this work identifies and characterizes two chemical scaffolds that chelate iron to inhibit the growth of the clinically relevant fungal pathogen C. albicansIMPORTANCEThe worldwide incidence of invasive fungal infections is increasing at an alarming rate. Systemic candidiasis caused by the opportunistic pathogen Candida albicans is the most common cause of life-threatening fungal infection. However, due to the limited number of antifungal drug classes available and the rise of antifungal resistance, an urgent need exists for the identification of novel treatments. By screening a compound collection from the Boston University Center for Molecular Discovery (BU-CMD), we identified three compounds representing two distinct chemical scaffolds that displayed activity against C. albicans. Follow-up analyses confirmed these molecules were also active against other pathogenic fungal species including Candida auris and Aspergillus fumigatus. Finally, we determined that these compounds inhibit the growth of C. albicans in culture through iron chelation. Overall, this observation describes two novel chemical scaffolds with antifungal activity against diverse fungal pathogens.
Collapse
Affiliation(s)
- Sara Fallah
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Dustin Duncan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, Brock University, St. Catharines, Ontario, Canada
| | - Kyle D. Reichl
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts, USA
| | - Michael J. Smith
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts, USA
| | - Wenyu Wang
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts, USA
| | - John A. Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts, USA
| | - Lauren E. Brown
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts, USA
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Day AW, Kumamoto CA. Selection of ethanol tolerant strains of Candida albicans by repeated ethanol exposure results in strains with reduced susceptibility to fluconazole. PLoS One 2024; 19:e0298724. [PMID: 38377103 PMCID: PMC10878505 DOI: 10.1371/journal.pone.0298724] [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: 11/09/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Candida albicans is a commensal yeast that has important impacts on host metabolism and immune function, and can establish life-threatening infections in immunocompromised individuals. Previously, C. albicans colonization has been shown to contribute to the progression and severity of alcoholic liver disease. However, relatively little is known about how C. albicans responds to changing environmental conditions in the GI tract of individuals with alcohol use disorder, namely repeated exposure to ethanol. In this study, we repeatedly exposed C. albicans to high concentrations (10% vol/vol) of ethanol-a concentration that can be observed in the upper GI tract of humans following consumption of alcohol. Following this repeated exposure protocol, ethanol small colony (Esc) variants of C. albicans isolated from these populations exhibited increased ethanol tolerance, altered transcriptional responses to ethanol, and cross-resistance/tolerance to the frontline antifungal fluconazole. These Esc strains exhibited chromosomal copy number variations and carried polymorphisms in genes previously associated with the acquisition of fluconazole resistance during human infection. This study identifies a selective pressure that can result in evolution of fluconazole tolerance and resistance without previous exposure to the drug.
Collapse
Affiliation(s)
- Andrew W. Day
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, United States of America
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, United States of America
| |
Collapse
|
24
|
Vanzolini T, Magnani M. Old and new strategies in therapy and diagnosis against fungal infections. Appl Microbiol Biotechnol 2024; 108:147. [PMID: 38240822 PMCID: PMC10799149 DOI: 10.1007/s00253-023-12884-8] [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: 06/26/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024]
Abstract
Fungal infections represent a serious global health threat. The new emerging pathogens and the spread of different forms of resistance are now hardly challenging the tools available in therapy and diagnostics. With the commonly used diagnoses, fungal identification is often slow and inaccurate, and, on the other hand, some drugs currently used as treatments are significantly affected by the decrease in susceptibility. Herein, the antifungal arsenal is critically summarized. Besides describing the old approaches and their mechanisms, advantages, and limitations, the focus is dedicated to innovative strategies which are designed, identified, and developed to take advantage of the discrepancies between fungal and host cells. Relevant pathways and their role in survival and virulence are discussed as their suitability as sources of antifungal targets. In a similar way, molecules with antifungal activity are reported as potential agents/precursors of the next generation of antimycotics. Particular attention was devoted to biotechnological entities, to their novelty and reliability, to drug repurposing and restoration, and to combinatorial applications yielding significant improvements in efficacy. KEY POINTS: • New antifungal agents and targets are needed to limit fungal morbidity and mortality. • Therapeutics and diagnostics suffer of delays in innovation and lack of targets. • Biologics, drug repurposing and combinations are the future of antifungal treatments.
Collapse
Affiliation(s)
- Tania Vanzolini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy.
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| |
Collapse
|
25
|
Lefranc M, Accoceberry I, Fitton-Ouhabi V, Biteau N, Noël T. Rapamycin and caspofungin show synergistic antifungal effects in caspofungin-susceptible and caspofungin-resistant Candida strains in vitro. J Antimicrob Chemother 2024; 79:151-156. [PMID: 37991226 DOI: 10.1093/jac/dkad359] [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: 06/30/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVES Caspofungin is an echinocandin antifungal agent that inhibits synthesis of glucan required for the fungal cell wall. Resistance is mediated by mutation of Fks1 glucan synthase, among which S645P is the most common resistance-associated polymorphism. Rapamycin is a macrolide that inhibits the mechanistic target of rapamycin (mTOR) protein kinase activity. This study investigated the interaction between rapamycin and caspofungin in inhibiting the growth of WT Candida albicans and Fks1 S645P mutant clinical isolate, and WT Candida lusitaniae and genetically engineered isogenic strain with Fks1 S645P mutation at equivalent position. METHODS Interactions between caspofungin and rapamycin were evaluated using the microdilution chequerboard method in liquid medium. The results were analysed using the Loewe additivity model (FIC index, FICI) and the Bliss independence model (response surface, RS, analysis). RESULTS Synergy between rapamycin and caspofungin was shown for C. albicans and C. lusitaniae strains by RS analysis of the chequerboard tests. Synergy was observed in strains susceptible and resistant to caspofungin. Weak subinhibitory concentrations of rapamycin were sufficient to restore caspofungin susceptibility. CONCLUSIONS We report here, for the first time, synergy between caspofungin and rapamycin in Candida species. Synergy was shown for strains susceptible and resistant to caspofungin. This study highlights the possible implication of the TOR pathway in sensing antifungal-mediated cell wall stress and in modulating the cellular response to echinocandins in Candida yeasts.
Collapse
Affiliation(s)
- Maxime Lefranc
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, CHU Bordeaux, 33000 Bordeaux, France
| | - Isabelle Accoceberry
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, CHU Bordeaux, 33000 Bordeaux, France
| | - Valérie Fitton-Ouhabi
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
| | - Nicolas Biteau
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
| | - Thierry Noël
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
| |
Collapse
|
26
|
Lin Y, Jung H, Bulman CA, Ng J, Vinck R, O’Beirne C, Zhong S, Moser MS, Tricoche N, Peguero R, Li RW, Urban JF, Le Pape P, Pagniez F, Moretto M, Weil T, Lustigman S, Cariou K, Mitreva M, Sakanari JA, Gasser G. Discovery of New Broad-Spectrum Anti-Infectives for Eukaryotic Pathogens Using Bioorganometallic Chemistry. J Med Chem 2023; 66:15867-15882. [PMID: 38009931 PMCID: PMC11840807 DOI: 10.1021/acs.jmedchem.3c01333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Drug resistance observed with many anti-infectives clearly highlights the need for new broad-spectrum agents to treat especially neglected tropical diseases (NTDs) caused by eukaryotic parasitic pathogens, including fungal infections. Herein, we show that the simple modification of one of the most well-known antifungal drugs, fluconazole, with organometallic moieties not only improves the activity of the parent drug but also broadens the scope of application of the new derivatives. These compounds were highly effective in vivo against pathogenic fungal infections and potent against parasitic worms such as Brugia, which causes lymphatic filariasis and Trichuris, one of the soil-transmitted helminths that infects millions of people globally. Notably, the identified molecular targets indicate a mechanism of action that differs greatly from that of the parental antifungal drug, including targets involved in biosynthetic pathways that are absent in humans, offering great potential to expand our armamentarium against drug-resistant fungal infections and neglected tropical diseases (NTDs) targeted for elimination by 2030.
Collapse
Affiliation(s)
- Yan Lin
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Hyeim Jung
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christina A. Bulman
- University of California, San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA 94158, USA
| | - James Ng
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Cillian O’Beirne
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Shuai Zhong
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Matthew S. Moser
- University of California, San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA 94158, USA
| | - Nancy Tricoche
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Ricardo Peguero
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Robert W. Li
- Animal Parasitic Diseases Laboratory, United States Department of Agricultural Research Service (USDA-ARS), Beltsville, MD 20705, USA
| | - Joseph F. Urban
- Diet, Genomics and Immunology Laboratory, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Patrice Le Pape
- Nantes Université, CHU de Nantes, Cibles et Médicaments des Infections et de l’Immunité, IICiMed, UR 1155, F-44000 Nantes, France
| | - Fabrice Pagniez
- Nantes Université, CHU de Nantes, Cibles et Médicaments des Infections et de l’Immunité, IICiMed, UR 1155, F-44000 Nantes, France
| | - Marco Moretto
- Fondazione Edmund Mach Via E. Mach 1, Research and Innovation Centre, Via E. Mach 1, 38010 San Michele all’Adige, Italy
| | - Tobias Weil
- Fondazione Edmund Mach Via E. Mach 1, Research and Innovation Centre, Via E. Mach 1, 38010 San Michele all’Adige, Italy
| | - Sara Lustigman
- Molecular Parasitology, New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Judy A. Sakanari
- University of California, San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA 94158, USA
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| |
Collapse
|
27
|
Zheng L, Xu Y, Wang C, Yang F, Dong Y, Guo L. Susceptibility to caspofungin is regulated by temperature and is dependent on calcineurin in Candida albicans. Microbiol Spectr 2023; 11:e0179023. [PMID: 37966204 PMCID: PMC10715083 DOI: 10.1128/spectrum.01790-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 10/06/2023] [Indexed: 11/16/2023] Open
Abstract
IMPORTANCE Echinocandins are the newest antifungal drugs and are first-line treatment option for life-threatening systemic infections. Due to lack of consensus regarding what temperature should be used when evaluating susceptibility of yeasts to echinocandins, typically either 30°C, 35°C, or 37°C is used. However, the impact of temperature on antifungal efficacy of echinocandins is unexplored. In the current study, we demonstrated that Candida albicans laboratory strain SC5314 was more susceptible to caspofungin at 37°C than at 30°C. We also found that calcineurin was required for temperature-modulated caspofungin susceptibility. Surprisingly, the altered caspofungin susceptibility was not due to differential expression of some canonical genes such as FKS, CHS, or CHT genes. The molecular mechanism of temperature-modulated caspofungin susceptibility is undetermined and deserves further investigations.
Collapse
Affiliation(s)
- Lijun Zheng
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Xu
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Chen Wang
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Feng Yang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yubo Dong
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Liangsheng Guo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
28
|
Sah SK, Yadav A, Kruppa MD, Rustchenko E. Identification of 10 genes on Candida albicans chromosome 5 that control surface exposure of the immunogenic cell wall epitope β-glucan and cell wall remodeling in caspofungin-adapted mutants. Microbiol Spectr 2023; 11:e0329523. [PMID: 37966256 PMCID: PMC10714753 DOI: 10.1128/spectrum.03295-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
IMPORTANCE Candida infections are often fatal in immuno-compromised individuals, resulting in many thousands of deaths per year. Caspofungin has proven to be an excellent anti-Candida drug and is now the frontline treatment for infections. However, as expected, the number of resistant cases is increasing; therefore, new treatment modalities are needed. We are determining metabolic pathways leading to decreased drug susceptibility in order to identify mechanisms facilitating evolution of clinical resistance. This study expands the understanding of genes that modulate drug susceptibility and reveals new targets for the development of novel antifungal drugs.
Collapse
Affiliation(s)
- Sudisht K. Sah
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| | - Anshuman Yadav
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael D. Kruppa
- Department of Biomedical Sciences, Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Elena Rustchenko
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
29
|
Feng Y, Lu H, Whiteway M, Jiang Y. Understanding fluconazole tolerance in Candida albicans: implications for effective treatment of candidiasis and combating invasive fungal infections. J Glob Antimicrob Resist 2023; 35:314-321. [PMID: 37918789 DOI: 10.1016/j.jgar.2023.10.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/07/2023] [Accepted: 10/22/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVES Fluconazole (FLC) tolerant phenotypes in Candida species contribute to persistent candidemia and the emergence of FLC resistance. Therefore, making FLC fungicidal and eliminating FLC tolerance are important for treating invasive fungal diseases (IFDs) caused by Candida species. However, the mechanisms of FLC tolerance in Candida species remain to be fully explored. METHODS This review discusses the high incidence of FLC tolerance in Candida species and the importance of successfully clearing FLC tolerance in treating candidiasis. We further define and characterize FLC tolerance in C. albicans. RESULTS This review identifies global factors affecting FLC tolerance and suggest that FLC tolerance is a strategy of C. albicans response to FLC damage whose mechanism differs from FLC resistance. CONCLUSIONS This review highlights the significance of the cell membrane and cell wall integrity in FLC tolerance, guiding approaches to combat IFDs caused by Candida species..
Collapse
Affiliation(s)
- Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | | | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
30
|
Rouges C, Asad M, Laurent AD, Marchand P, Le Pape P. Is the C-Terminal Domain an Effective and Selective Target for the Design of Hsp90 Inhibitors against Candida Yeast? Microorganisms 2023; 11:2837. [PMID: 38137982 PMCID: PMC10745388 DOI: 10.3390/microorganisms11122837] [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: 09/08/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Improving the armamentarium to treat invasive candidiasis has become necessary to overcome drug resistance and the lack of alternative therapy. In the pathogenic fungus Candida albicans, the 90-kDa Heat-Shock Protein (Hsp90) has been described as a major regulator of virulence and resistance, offering a promising target. Some human Hsp90 inhibitors have shown activity against Candida spp. in vitro, but host toxicity has limited their use as antifungal drugs. The conservation of Hsp90 across all species leads to selectivity issues. To assess the potential of Hsp90 as a druggable antifungal target, the activity of nine structurally unrelated Hsp90 inhibitors with different binding domains was evaluated against a panel of Candida clinical isolates. The Hsp90 sequences from human and yeast species were aligned. Despite the degree of similarity between human and yeast N-terminal domain residues, the in vitro activities measured for the inhibitors interacting with this domain were not reproducible against all Candida species. Moreover, the inhibitors binding to the C-terminal domain (CTD) did not show any antifungal activity, with the exception of one of them. Given the greater sequence divergence in this domain, the identification of selective CTD inhibitors of fungal Hsp90 could be a promising strategy for the development of innovative antifungal drugs.
Collapse
Affiliation(s)
- Célia Rouges
- Nantes Université, CHU Nantes, Cibles et Médicaments des Infections et de l’Immunité, IICiMed, UR 1155, F-44000 Nantes, France; (C.R.); (P.M.)
| | - Mohammad Asad
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France
| | - Adèle D. Laurent
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France
| | - Pascal Marchand
- Nantes Université, CHU Nantes, Cibles et Médicaments des Infections et de l’Immunité, IICiMed, UR 1155, F-44000 Nantes, France; (C.R.); (P.M.)
| | - Patrice Le Pape
- Nantes Université, CHU Nantes, Cibles et Médicaments des Infections et de l’Immunité, IICiMed, UR 1155, F-44000 Nantes, France; (C.R.); (P.M.)
| |
Collapse
|
31
|
Day AW, Kumamoto CA. Selection of Ethanol Tolerant Strains of Candida albicans by Repeated Ethanol Exposure Results in Strains with Reduced Susceptibility to Fluconazole. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.13.557677. [PMID: 37745460 PMCID: PMC10515905 DOI: 10.1101/2023.09.13.557677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Candida albicans is a commensal yeast that has important impacts on host metabolism and immune function, and can establish life-threatening infections in immunocompromised individuals. Previously, C. albicans colonization has been shown to contribute to the progression and severity of alcoholic liver disease. However, relatively little is known about how C. albicans responds to changing environmental conditions in the GI tract of individuals with alcohol use disorder, namely repeated exposure to ethanol. In this study, we repeatedly exposed C. albicans to high concentrations (10% vol/vol) of ethanol-a concentration that can be observed in the upper GI tract of humans following consumption of alcohol. Following this repeated exposure protocol, ethanol small colony (Esc) variants of C. albicans isolated from these populations exhibited increased ethanol tolerance, altered transcriptional responses to ethanol, and cross-resistance/tolerance to the frontline antifungal fluconazole. These Esc strains exhibited chromosomal copy number variations and carried polymorphisms in genes previously associated with the acquisition of fluconazole resistance during human infection. This study identifies a selective pressure that can result in evolution of fluconazole tolerance and resistance without previous exposure to the drug.
Collapse
Affiliation(s)
- Andrew W. Day
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| |
Collapse
|
32
|
Stover KR, Hawkins BK, Keck JM, Barber KE, Cretella DA. Antifungal resistance, combinations and pipeline: oh my! Drugs Context 2023; 12:2023-7-1. [PMID: 38021410 PMCID: PMC10653594 DOI: 10.7573/dic.2023-7-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/26/2023] [Indexed: 12/01/2023] Open
Abstract
Invasive fungal infections are a strong contributor to healthcare costs, morbidity and mortality, especially amongst hospitalized patients. Historically, Candida was responsible for approximately 15% of all nosocomial bloodstream infections. In the past 10 years, the epidemiology of Candida species has altered, with increasing prevalence of resistant species. With rising fungal resistance, especially in Candida spp., the demand for novel antifungal therapies has exponentially increased over the last decade. Newer antifungal agents have become an attractive option for patients needing long-term therapy for infections or those requiring antifungal prophylaxis. Despite advances in coverage of non-Candida pathogens with newer agents, clinical scenarios involving multidrug-resistant fungal pathogens continue to arise in practice. Combination antifungal therapy can lead to a host of side-effects, some of which can be drug limiting. Additional antifungal therapies with enhanced fungal spectrum of activity and decreased rates of adverse effects are warranted. Fosmanogepix, ibrexafungerp, olorofim and rezafungin may help fill some of these gaps in the antifungal armamentarium. This article is part of the Challenges and strategies in the management of invasive fungal infections Special Issue: https://www.drugsincontext.com/special_issues/challenges-and-strategies-in-the-management-of-invasive-fungal-infections.
Collapse
Affiliation(s)
- Kayla R Stover
- Department of Pharmacy Practice, University of Mississippi School of Pharmacy, Jackson, MS, USA
| | - Brandon K Hawkins
- Department of Clinical Pharmacy and Translational Science, The University of Tennessee Health Science Center, Knoxville, TN, USA
| | - J Myles Keck
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Katie E Barber
- Department of Pharmacy Practice, University of Mississippi School of Pharmacy, Jackson, MS, USA
| | - David A Cretella
- Division of Infectious Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
33
|
Dumeaux V, Massahi S, Bettauer V, Mottola A, Dukovny A, Khurdia SS, Costa ACBP, Omran RP, Simpson S, Xie JL, Whiteway M, Berman J, Hallett MT. Candida albicans exhibits heterogeneous and adaptive cytoprotective responses to antifungal compounds. eLife 2023; 12:e81406. [PMID: 37888959 PMCID: PMC10699808 DOI: 10.7554/elife.81406] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/26/2023] [Indexed: 10/28/2023] Open
Abstract
Candida albicans, an opportunistic human pathogen, poses a significant threat to human health and is associated with significant socio-economic burden. Current antifungal treatments fail, at least in part, because C. albicans can initiate a strong drug tolerance response that allows some cells to grow at drug concentrations above their minimal inhibitory concentration. To better characterize this cytoprotective tolerance program at the molecular single-cell level, we used a nanoliter droplet-based transcriptomics platform to profile thousands of individual fungal cells and establish their subpopulation characteristics in the absence and presence of antifungal drugs. Profiles of untreated cells exhibit heterogeneous expression that correlates with cell cycle stage with distinct metabolic and stress responses. At 2 days post-fluconazole exposure (a time when tolerance is measurable), surviving cells bifurcate into two major subpopulations: one characterized by the upregulation of genes encoding ribosomal proteins, rRNA processing machinery, and mitochondrial cellular respiration capacity, termed the Ribo-dominant (Rd) state; and the other enriched for genes encoding stress responses and related processes, termed the Stress-dominant (Sd) state. This bifurcation persists at 3 and 6 days post-treatment. We provide evidence that the ribosome assembly stress response (RASTR) is activated in these subpopulations and may facilitate cell survival.
Collapse
Affiliation(s)
- Vanessa Dumeaux
- Department of Anatomy and Cell Biology, Western University, London, Canada
| | - Samira Massahi
- Department of Biology, Concordia University, Montreal, Canada
| | - Van Bettauer
- Department of Computer Science and Software Engineering, Concordia University, Montreal, Canada
| | - Austin Mottola
- Shmunis School of Biomedical and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Anna Dukovny
- Shmunis School of Biomedical and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | | | | | | | - Shawn Simpson
- Department of Computer Science and Software Engineering, Concordia University, Montreal, Canada
| | - Jinglin Lucy Xie
- Department of Chemical and Systems Biology, Stanford University, Stanford, United States
| | | | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | | |
Collapse
|
34
|
Robbins N, Cowen LE. Roles of Hsp90 in Candida albicans morphogenesis and virulence. Curr Opin Microbiol 2023; 75:102351. [PMID: 37399670 PMCID: PMC11016340 DOI: 10.1016/j.mib.2023.102351] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023]
Abstract
Hsp90 is a conserved molecular chaperone that facilitates the folding and function of hundreds of client proteins, many of which serve as core hubs of signal transduction networks. Hsp90 has a critical role in virulence of the opportunistic fungal pathogen Candida albicans, which exists as a natural commensal of the human microbiota and is a leading cause of invasive fungal infections, particularly in immunocompromised individuals. The ability of C. albicans to cause disease is tightly coupled to its capacity to undergo a morphogenetic transition between yeast and filamentous forms. Here, we describe the complex mechanisms by which Hsp90 regulates C. albicans morphogenesis and virulence, and explore the potential of targeting fungal Hsp90 as a therapeutic strategy to combat fungal infections.
Collapse
Affiliation(s)
- Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Prasad P, Jain N, Chaudhary J, Thakur RK, Savadi S, Bhardwaj SC, Gangwar OP, Lata C, Adhikari S, Kumar S, Balyan HS, Gupta PK. Candidate effectors for leaf rust resistance gene Lr28 identified through transcriptome and in-silico analysis. Front Microbiol 2023; 14:1143703. [PMID: 37789861 PMCID: PMC10543267 DOI: 10.3389/fmicb.2023.1143703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/31/2023] [Indexed: 10/05/2023] Open
Abstract
Puccinia spp. causing rust diseases in wheat and other cereals secrete several specialized effector proteins into host cells. Characterization of these proteins and their interaction with host's R proteins could greatly help to limit crop losses due to diseases. Prediction of effector proteins by combining the transcriptome analysis and multiple in-silico approaches is gaining importance in revealing the pathogenic mechanism. The present study involved identification of 13 Puccinia triticina (Pt) coding sequences (CDSs), through transcriptome analysis, that were differentially expressed during wheat-leaf rust interaction; and prediction of their effector like features using different in-silico tools. NCBI-BLAST and pathogen-host interaction BLAST (PHI-BLAST) tools were used to annotate and classify these sequences based on their most closely matched counterpart in both the databases. Homology between CDSs and the annotated sequences in the NCBI database ranged from 79 to 94% and with putative effectors of other plant pathogens in PHI-BLAST from 24.46 to 54.35%. Nine of the 13 CDSs had effector-like features according to EffectorP 3.0 (≥0.546 probability of these sequences to be effector). The qRT-PCR expression analysis revealed that the relative expression of all CDSs in compatible interaction (HD2329) was maximum at 11 days post inoculation (dpi) and that in incompatible interactions (HD2329 + Lr28) was maximum at 3 dpi in seven and 9 dpi in five CDSs. These results suggest that six CDSs (>0.8 effector probability as per EffectorP 3.0) could be considered as putative Pt effectors. The molecular docking and MD simulation analysis of these six CDSs suggested that candidate Lr28 protein binds more strongly to candidate effector c14094_g1_i1 to form more stable complex than the remaining five. Further functional characterization of these six candidate effectors should prove useful for a better understanding of wheat-leaf rust interaction. In turn, this should facilitate effector-based leaf rust resistance breeding in wheat.
Collapse
Affiliation(s)
- Pramod Prasad
- ICAR-Indian Institute of Wheat and Barley Research, Regional Station, Shimla, India
| | - Neelu Jain
- Division of Genetics, ICAR-Indian Agricultural Research Institute (IARI), New Delhi, India
| | - Jyoti Chaudhary
- Department of Genetics and Plant Breeding, Chaudhary Charan Singh University, Meerut, India
| | - Rajni Kant Thakur
- ICAR-Indian Institute of Wheat and Barley Research, Regional Station, Shimla, India
| | | | | | - Om Prakash Gangwar
- ICAR-Indian Institute of Wheat and Barley Research, Regional Station, Shimla, India
| | - Charu Lata
- ICAR-Indian Institute of Wheat and Barley Research, Regional Station, Shimla, India
| | - Sneha Adhikari
- ICAR-Indian Institute of Wheat and Barley Research, Regional Station, Shimla, India
- Division of Genetics, ICAR-Indian Agricultural Research Institute (IARI), New Delhi, India
| | - Subodh Kumar
- ICAR-Indian Institute of Wheat and Barley Research, Regional Station, Shimla, India
| | - Harindra Singh Balyan
- Department of Genetics and Plant Breeding, Chaudhary Charan Singh University, Meerut, India
| | - Pushpendra Kumar Gupta
- Department of Genetics and Plant Breeding, Chaudhary Charan Singh University, Meerut, India
| |
Collapse
|
36
|
Marquez L, Lee Y, Duncan D, Whitesell L, Cowen LE, Quave C. Potent Antifungal Activity of Penta- O-galloyl-β-d-Glucose against Drug-Resistant Candida albicans, Candida auris, and Other Non- albicans Candida Species. ACS Infect Dis 2023; 9:1685-1694. [PMID: 37607350 PMCID: PMC10496123 DOI: 10.1021/acsinfecdis.3c00113] [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: 03/10/2023] [Indexed: 08/24/2023]
Abstract
Among fungal pathogens, infections by drug-resistant Candida species continue to pose a major challenge to healthcare. This study aimed to evaluate the activity of the bioactive natural product, penta-O-galloyl-β-d-glucose (PGG) against multidrug-resistant (MDR) Candida albicans, MDR Candida auris, and other MDR non-albicans Candida species. Here, we show that PGG has a minimum inhibitory concentration (MIC) of 0.25-8 μg mL-1 (0.265-8.5 μM) against three clinical strains of C. auris and a MIC of 0.25-4 μg mL-1 (0.265-4.25 μM) against a panel of other MDR Candida species. Our cytotoxicity studies found that PGG was well tolerated by human kidney, liver, and epithelial cells with an IC50 > 256 μg mL-1 (>272 μM). We also show that PGG is a high-capacity iron chelator and that deletion of key iron homeostasis genes in C. albicans rendered strains hypersensitive to PGG. In conclusion, PGG displayed potent anti-Candida activity with minimal cytotoxicity for human cells. We also found that the antifungal activity of PGG is mediated through an iron-chelating mechanism, suggesting that the compound could prove useful as a topical treatment for superficial Candida infections.
Collapse
Affiliation(s)
- Lewis Marquez
- Molecular
and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, Georgia 30322, United States
- Jones
Center at Ichauway, Newton, Georgia 39870, United States
| | - Yunjin Lee
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Dustin Duncan
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
- Department
of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| | - Luke Whitesell
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Leah E. Cowen
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Cassandra Quave
- Center
for the Study of Human Health, Emory University, Atlanta, Georgia 30322, United States
- Department
of Dermatology, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
37
|
Wagner AS, Lumsdaine SW, Mangrum MM, Reynolds TB. Caspofungin-induced β(1,3)-glucan exposure in Candida albicans is driven by increased chitin levels. mBio 2023; 14:e0007423. [PMID: 37377417 PMCID: PMC10470516 DOI: 10.1128/mbio.00074-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/04/2023] [Indexed: 06/29/2023] Open
Abstract
To successfully induce disease, Candida albicans must effectively evade the host immune system. One mechanism used by C. albicans to achieve this is to mask immunogenic β(1,3)-glucan epitopes within its cell wall under an outer layer of mannosylated glycoproteins. Consequently, induction of β(1,3)-glucan exposure (unmasking) via genetic or chemical manipulation increases fungal recognition by host immune cells in vitro and attenuates disease during systemic infection in mice. Treatment with the echinocandin caspofungin is one of the most potent drivers of β(1,3)-glucan exposure. Several reports using murine infection models suggest a role for the immune system, and specifically host β(1,3)-glucan receptors, in mediating the efficacy of echinocandin treatment in vivo. However, the mechanism by which caspofungin-induced unmasking occurs is not well understood. In this report, we show that foci of unmasking co-localize with areas of increased chitin within the yeast cell wall in response to caspofungin, and that inhibition of chitin synthesis via nikkomycin Z attenuates caspofungin-induced β(1,3)-glucan exposure. Furthermore, we find that both the calcineurin and Mkc1 mitogen-activated protein kinase pathways work synergistically to regulate β(1,3)-glucan exposure and chitin synthesis in response to drug treatment. When either of these pathways are interrupted, it results in a bimodal population of cells containing either high or low chitin content. Importantly, increased unmasking correlates with increased chitin content within these cells. Microscopy further indicates that caspofungin-induced unmasking correlates with actively growing cells. Collectively, our work presents a model in which chitin synthesis induces unmasking within the cell wall in response to caspofungin in growing cells. IMPORTANCE Systemic candidiasis has reported mortality rates ranging from 20% to 40%. The echinocandins, including caspofungin, are first-line antifungals used to treat systemic candidiasis. However, studies in mice have shown that echinocandin efficacy relies on both its cidal impacts on Candida albicans, as well as a functional immune system to successfully clear invading fungi. In addition to direct C. albicans killing, caspofungin increases exposure (unmasking) of immunogenic β(1,3)-glucan moieties. To evade immune detection, β(1,3)-glucan is normally masked within the C. albicans cell wall. Consequently, unmasked β(1,3)-glucan renders these cells more visible to the host immune system and attenuates disease progression. Therefore, discovery of how caspofungin-induced unmasking occurs is needed to elucidate how the drug facilitates host immune system-mediated clearance in vivo. We report a strong and consistent correlation between chitin deposition and unmasking in response to caspofungin and propose a model in which altered chitin synthesis drives increased unmasking during drug exposure.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | | | - Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
38
|
Shen J, Ma M, Duan W, Huang Y, Shi B, Wu Q, Wei X. Autophagy Alters the Susceptibility of Candida albicans Biofilms to Antifungal Agents. Microorganisms 2023; 11:2015. [PMID: 37630575 PMCID: PMC10458732 DOI: 10.3390/microorganisms11082015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Candida albicans (C. albicans) reigns as a major cause of clinical candidiasis. C. albicans biofilms are known to increase resistance to antifungal agents, making biofilm-related infections particularly challenging to treat. Drug resistance is of particular concern due to the spread of multidrug-resistant fungal pathogens, while autophagy is crucial for the maintenance of cellular homeostasis. Therefore, this study aimed to investigate the effects of an activator and an inhibitor of autophagy on the susceptibility of C. albicans biofilms to antifungal agents and the related mechanisms. The susceptibility of C. albicans biofilms to different antifungal agents after treatment with or without the autophagy activator or inhibitor was evaluated using XTT assay. Alkaline phosphatase (ALP) activity and reactive oxygen species (ROS) level, as well as the expression of ROS-related and autophagy-related genes, were examined to evaluate the autophagic activity of C. albicans biofilms when treated with antifungal agents. The autophagosomes were observed by transmission electron microscopy (TEM). The susceptibility of C. albicans biofilms to antifungal agents changed when autophagy changed. The ALP activity and ROS level of C. albicans biofilms increased with the treatment of antifungal agents, and autophagosomes could be observed in C. albicans biofilms. Autophagy was involved in the susceptibility of C. albicans biofilms to antifungal agents.
Collapse
Affiliation(s)
- Jiadi Shen
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| | - Ming Ma
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| | - Wei Duan
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| | - Yun Huang
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| | - Banruo Shi
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| | - Qiaochu Wu
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| | - Xin Wei
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210000, China; (J.S.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
39
|
Sharma C, Kadosh D. Post-transcriptional control of antifungal resistance in human fungal pathogens. Crit Rev Microbiol 2023; 49:469-484. [PMID: 35634915 PMCID: PMC9766424 DOI: 10.1080/1040841x.2022.2080527] [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: 01/12/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 11/03/2022]
Abstract
Global estimates suggest that over 300 million individuals of all ages are affected by serious fungal infections every year, culminating in about 1.7 million deaths. The societal and economic burden on the public health sector due to opportunistic fungal pathogens is quite significant, especially among immunocompromised patients. Despite the high clinical significance of these infectious agents, treatment options are limited with only three major classes of antifungal drugs approved for use. Clinical management of fungal diseases is further compromised by the emergence of antifungal resistant strains. Transcriptional and genetic mechanisms that control drug resistance in human fungal pathogens are well-studied and include drug target alteration, upregulation of drug efflux pumps as well as changes in drug affinity and abundance of target proteins. In this review, we highlight several recently discovered novel post-transcriptional mechanisms that control antifungal resistance, which involve regulation at the translational, post-translational, epigenetic, and mRNA stability levels. The discovery of many of these novel mechanisms has opened new avenues for the development of more effective antifungal treatment strategies and new insights, perspectives, and future directions that will facilitate this process are discussed.
Collapse
Affiliation(s)
- Cheshta Sharma
- Department of Microbiology, Immunology and Molecular Genetics University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - David Kadosh
- Department of Microbiology, Immunology and Molecular Genetics University of Texas Health Science Center at San Antonio, San Antonio, TX
| |
Collapse
|
40
|
Lu H, Hong T, Jiang Y, Whiteway M, Zhang S. Candidiasis: From cutaneous to systemic, new perspectives of potential targets and therapeutic strategies. Adv Drug Deliv Rev 2023; 199:114960. [PMID: 37307922 DOI: 10.1016/j.addr.2023.114960] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Candidiasis is an infection caused by fungi from a Candida species, most commonly Candida albicans. C. albicans is an opportunistic fungal pathogen typically residing on human skin and mucous membranes of the mouth, intestines or vagina. It can cause a wide variety of mucocutaneous barrier and systemic infections; and becomes a severe health problem in HIV/AIDS patients and in individuals who are immunocompromised following chemotherapy, treatment with immunosuppressive agents or after antibiotic-induced dysbiosis. However, the immune mechanism of host resistance to C. albicans infection is not fully understood, there are a limited number of therapeutic antifungal drugs for candidiasis, and these have disadvantages that limit their clinical application. Therefore, it is urgent to uncover the immune mechanisms of the host protecting against candidiasis and to develop new antifungal strategies. This review synthesizes current knowledge of host immune defense mechanisms from cutaneous candidiasis to invasive C. albicans infection and documents promising insights for treating candidiasis through inhibitors of potential antifungal target proteins.
Collapse
Affiliation(s)
- Hui Lu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Ting Hong
- Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada.
| | - Shiqun Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China.
| |
Collapse
|
41
|
Iyer KR, Li SC, Revie NM, Lou JW, Duncan D, Fallah S, Sanchez H, Skulska I, Ušaj MM, Safizadeh H, Larsen B, Wong C, Aman A, Kiyota T, Yoshimura M, Kimura H, Hirano H, Yoshida M, Osada H, Gingras AC, Andes DR, Shapiro RS, Robbins N, Mazhab-Jafari MT, Whitesell L, Yashiroda Y, Boone C, Cowen LE. Identification of triazenyl indoles as inhibitors of fungal fatty acid biosynthesis with broad-spectrum activity. Cell Chem Biol 2023; 30:795-810.e8. [PMID: 37369212 PMCID: PMC11016341 DOI: 10.1016/j.chembiol.2023.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 04/17/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023]
Abstract
Rising drug resistance among pathogenic fungi, paired with a limited antifungal arsenal, poses an increasing threat to human health. To identify antifungal compounds, we screened the RIKEN natural product depository against representative isolates of four major human fungal pathogens. This screen identified NPD6433, a triazenyl indole with broad-spectrum activity against all screening strains, as well as the filamentous mold Aspergillus fumigatus. Mechanistic studies indicated that NPD6433 targets the enoyl reductase domain of fatty acid synthase 1 (Fas1), covalently inhibiting its flavin mononucleotide-dependent NADPH-oxidation activity and arresting essential fatty acid biosynthesis. Robust Fas1 inhibition kills Candida albicans, while sublethal inhibition impairs diverse virulence traits. At well-tolerated exposures, NPD6433 extended the lifespan of nematodes infected with azole-resistant C. albicans. Overall, identification of NPD6433 provides a tool with which to explore lipid homeostasis as a therapeutic target in pathogenic fungi and reveals a mechanism by which Fas1 function can be inhibited.
Collapse
Affiliation(s)
- Kali R Iyer
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Sheena C Li
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada; RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Nicole M Revie
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Jennifer W Lou
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Dustin Duncan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Sara Fallah
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hiram Sanchez
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Iwona Skulska
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Mojca Mattiazzi Ušaj
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
| | - Hamid Safizadeh
- Department of Computer Science and Engineering and Department of Electrical and Computer Engineering, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Brett Larsen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Cassandra Wong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Taira Kiyota
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Mami Yoshimura
- RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Hiromi Kimura
- RIKEN Center for Sustainable Resource Science, Wako, Japan
| | | | - Minoru Yoshida
- RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Hiroyuki Osada
- RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - David R Andes
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mohammad T Mazhab-Jafari
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Yoko Yashiroda
- RIKEN Center for Sustainable Resource Science, Wako, Japan.
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada; RIKEN Center for Sustainable Resource Science, Wako, Japan.
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
42
|
Lee Y, Robbins N, Cowen LE. Molecular mechanisms governing antifungal drug resistance. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:5. [PMID: 38686214 PMCID: PMC11057204 DOI: 10.1038/s44259-023-00007-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/17/2023] [Indexed: 05/02/2024]
Abstract
Fungal pathogens are a severe public health problem. The leading causative agents of systemic fungal infections include species from the Candida, Cryptococcus, and Aspergillus genera. As opportunistic pathogens, these fungi are generally harmless in healthy hosts; however, they can cause significant morbidity and mortality in immunocompromised patients. Despite the profound impact of pathogenic fungi on global human health, the current antifungal armamentarium is limited to only three major classes of drugs, all of which face complications, including host toxicity, unfavourable pharmacokinetics, or limited spectrum of activity. Further exacerbating this issue is the growing prevalence of antifungal-resistant infections and the emergence of multidrug-resistant pathogens. In this review, we discuss the diverse strategies employed by leading fungal pathogens to evolve antifungal resistance, including drug target alterations, enhanced drug efflux, and induction of cellular stress response pathways. Such mechanisms of resistance occur through diverse genetic alterations, including point mutations, aneuploidy formation, and epigenetic changes given the significant plasticity observed in many fungal genomes. Additionally, we highlight recent literature surrounding the mechanisms governing resistance in emerging multidrug-resistant pathogens including Candida auris and Candida glabrata. Advancing our knowledge of the molecular mechanisms by which fungi adapt to the challenge of antifungal exposure is imperative for designing therapeutic strategies to tackle the emerging threat of antifungal resistance.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| |
Collapse
|
43
|
Lin Y, Jung H, Bulman CA, Ng J, Vinck R, O'Beirne C, Moser MS, Tricoche N, Peguero R, Li RW, Urban JF, Pape PL, Pagniez F, Moretto M, Weil T, Lustigman S, Cariou K, Mitreva M, Sakanari JA, Gasser G. Discovery of New Broad-Spectrum Anti-Infectives for Eukaryotic Pathogens Using Bioorganometallic Chemistry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546819. [PMID: 37425761 PMCID: PMC10327022 DOI: 10.1101/2023.06.28.546819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Drug resistance observed with many anti-infectives clearly highlights the need for new broad-spectrum agents to treat especially neglected tropical diseases (NTDs) caused by eukaryotic parasitic pathogens including fungal infections. Since these diseases target the most vulnerable communities who are disadvantaged by health and socio-economic factors, new agents should be, if possible, easy-to-prepare to allow for commercialization based on their low cost. In this study, we show that simple modification of one of the most well-known antifungal drugs, fluconazole, with organometallic moieties not only improves the activity of the parent drug but also broadens the scope of application of the new derivatives. These compounds were highly effective in vivo against pathogenic fungal infections and potent against parasitic worms such as Brugia, which causes lymphatic filariasis and Trichuris, one of the soil-transmitted helminths that infects millions of people globally. Notably, the identified molecular targets indicate a mechanism of action that differs greatly from the parental antifungal drug, including targets involved in biosynthetic pathways that are absent in humans, offering great potential to expand our armamentarium against drug-resistant fungal infections and NTDs targeted for elimination by 2030. Overall, the discovery of these new compounds with broad-spectrum activity opens new avenues for the development of treatments for several current human infections, either caused by fungi or by parasites, including other NTDs, as well as newly emerging diseases. ONE-SENTENCE SUMMARY Simple derivatives of the well-known antifungal drug fluconazole were found to be highly effective in vivo against fungal infections, and also potent against the parasitic nematode Brugia, which causes lymphatic filariasis and against Trichuris, one of the soil-transmitted helminths that infects millions of people globally.
Collapse
|
44
|
Yang F, Wang Y, Yan D, Liu Z, Wei B, Chen J, He W. Binding Mechanism of Inhibitors to Heat Shock Protein 90 Investigated by Multiple Independent Molecular Dynamics Simulations and Prediction of Binding Free Energy. Molecules 2023; 28:4792. [PMID: 37375347 DOI: 10.3390/molecules28124792] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The heat shock protein (HSP90) has been an import target of drug design in the treatment of human disease. An exploration of the conformational changes in HSP90 can provide useful information for the development of efficient inhibitors targeting HSP90. In this work, multiple independent all-atom molecular dynamics (AAMD) simulations followed by calculations of the molecular mechanics generalized Born surface area (MM-GBSA) were performed to explore the binding mechanism of three inhibitors (W8Y, W8V, and W8S) to HSP90. The dynamics analyses verified that the presence of inhibitors impacts the structural flexibility, correlated movements, and dynamics behavior of HSP90. The results of the MM-GBSA calculations suggest that the selection of GB models and empirical parameters has important influences on the predicted results and verify that van der Waals interactions are the main forces that determine inhibitor-HSP90 binding. The contributions of separate residues to the inhibitor-HSP90 binding process indicate that hydrogen-bonding interactions (HBIs) and hydrophobic interactions play important roles in HSP90-inhibitor identifications. Moreover, residues L34, N37, D40, A41, D79, I82, G83, M84, F124, and T171 are recognized as hot spots of inhibitor-HSP90 binding and provide significant target sites of for the design of drugs related to HSP90. This study aims to contribute to the development of efficient inhibitors that target HSP90 by providing an energy-based and theoretical foundation.
Collapse
Affiliation(s)
- Fen Yang
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
| | - Yiwen Wang
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
- School of Aeronautics, Shandong Jiaotong University, Jinan 250357, China
| | - Dongliang Yan
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Zhongtao Liu
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
| | - Benzheng Wei
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China
| | - Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Weikai He
- School of Aeronautics, Shandong Jiaotong University, Jinan 250357, China
| |
Collapse
|
45
|
Rabaan AA, Sulaiman T, Al-Ahmed SH, Buhaliqah ZA, Buhaliqah AA, AlYuosof B, Alfaresi M, Al Fares MA, Alwarthan S, Alkathlan MS, Almaghrabi RS, Abuzaid AA, Altowaileb JA, Al Ibrahim M, AlSalman EM, Alsalman F, Alghounaim M, Bueid AS, Al-Omari A, Mohapatra RK. Potential Strategies to Control the Risk of Antifungal Resistance in Humans: A Comprehensive Review. Antibiotics (Basel) 2023; 12:antibiotics12030608. [PMID: 36978475 PMCID: PMC10045400 DOI: 10.3390/antibiotics12030608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/30/2023] Open
Abstract
Fungal infections are becoming one of the main causes of morbidity and mortality in people with weakened immune systems. Mycoses are becoming more common, despite greater knowledge and better treatment methods, due to the regular emergence of resistance to the antifungal medications used in clinical settings. Antifungal therapy is the mainstay of patient management for acute and chronic mycoses. However, the limited availability of antifungal drug classes limits the range of available treatments. Additionally, several drawbacks to treating mycoses include unfavourable side effects, a limited activity spectrum, a paucity of targets, and fungal resistance, all of which continue to be significant issues in developing antifungal drugs. The emergence of antifungal drug resistance has eliminated accessible drug classes as treatment choices, which significantly compromises the clinical management of fungal illnesses. In some situations, the emergence of strains resistant to many antifungal medications is a major concern. Although new medications have been developed to address this issue, antifungal drug resistance has grown more pronounced, particularly in patients who need long-term care or are undergoing antifungal prophylaxis. Moreover, the mechanisms that cause resistance must be well understood, including modifications in drug target affinities and abundances, along with biofilms and efflux pumps that diminish intracellular drug levels, to find novel antifungal drugs and drug targets. In this review, different classes of antifungal agents, and their resistance mechanisms, have been discussed. The latter part of the review focuses on the strategies by which we can overcome this serious issue of antifungal resistance in humans.
Collapse
Affiliation(s)
- Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Tarek Sulaiman
- Infectious Diseases Section, Medical Specialties Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Shamsah H Al-Ahmed
- Specialty Paediatric Medicine, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Zainab A Buhaliqah
- Department of Family Medicine, Primary Healthcare Center, Dammam 32433, Saudi Arabia
| | - Ali A Buhaliqah
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Buthina AlYuosof
- Directorate of Public Health, Dammam Network, Eastern Health Cluster, Dammam 31444, Saudi Arabia
| | - Mubarak Alfaresi
- Department of Pathology and Laboratory Medicine, Zayed Military Hospital, Abu Dhabi 3740, United Arab Emirates
- Department of Pathology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Mona A Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mohammed S Alkathlan
- Infectious Diseases Department, King Fahad Specialist Hospital, Buraydah 52382, Saudi Arabia
| | - Reem S Almaghrabi
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Abdulmonem A Abuzaid
- Medical Microbiology Department, Security Forces Hospital Programme, Dammam 32314, Saudi Arabia
| | - Jaffar A Altowaileb
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Maha Al Ibrahim
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Eman M AlSalman
- Department of Family Medicine, Primary Health Care Centers, Qatif Health Network, Qatif 31911, Saudi Arabia
| | - Fatimah Alsalman
- Department of Emergency Medicine, Oyun City Hospital, Al-Ahsa 36312, Saudi Arabia
| | | | - Ahmed S Bueid
- Microbiology Laboratory, King Faisal General Hospital, Al-Ahsa 31982, Saudi Arabia
| | - Awad Al-Omari
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Research Center, Dr. Sulaiman Al Habib Medical Group, Riyadh 11372, Saudi Arabia
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar 758002, India
| |
Collapse
|
46
|
Zhang Y, He K, Guo X, Jiang J, Qian L, Xu J, Che Z, Huang X, Liu S. Transcriptomic Profiling of Fusarium pseudograminearum in Response to Carbendazim, Pyraclostrobin, Tebuconazole, and Phenamacril. J Fungi (Basel) 2023; 9:jof9030334. [PMID: 36983502 PMCID: PMC10057576 DOI: 10.3390/jof9030334] [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: 01/12/2023] [Revised: 02/09/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Fusarium pseudograminearum has been identified as a significant pathogen. It causes Fusarium crown rot (FCR), which occurs in several major wheat-producing areas in China. Chemical control is the primary measure with which to control this disease. In this study, transcriptome sequencing (RNA-Seq) was used to determine the different mechanisms of action of four frequently used fungicides including carbendazim, pyraclostrobin, tebuconazole, and phenamacril on F. pseudograminearum. In brief, 381, 1896, 842, and 814 differentially expressed genes (DEGs) were identified under the carbendazim, pyraclostrobin, tebuconazole, and phenamacril treatments, respectively. After the joint analysis, 67 common DEGs were obtained, and further functional analysis showed that the ABC transported pathway was significantly enriched. Moreover, FPSE_04130 (FER6) and FPSE_11895 (MDR1), two important ABC multidrug transporter genes whose expression levels simultaneously increased, were mined under the different treatments, which unambiguously demonstrated the common effects. In addition, Mfuzz clustering analysis and WGCNA analysis revealed that the core DEGs are involved in several critical pathways in each of the four treatment groups. Taken together, these genes may play a crucial function in the mechanisms of F. pseudograminearum's response to the fungicides stress.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Kai He
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xuhao Guo
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Jia Jiang
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Le Qian
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Jianqiang Xu
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Zhiping Che
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Xiaobo Huang
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| | - Shengming Liu
- Department of Plant Protection, College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang 471023, China
| |
Collapse
|
47
|
Lu H, Li W, Whiteway M, Wang H, Zhu S, Ji Z, Feng Y, Yan L, Fang T, Li L, Ni T, Zhang X, Lv Q, Ding Z, Qiu L, Zhang D, Jiang Y. A Small Molecule Inhibitor of Erg251 Makes Fluconazole Fungicidal by Inhibiting the Synthesis of the 14α-Methylsterols. mBio 2023; 14:e0263922. [PMID: 36475771 PMCID: PMC9973333 DOI: 10.1128/mbio.02639-22] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Fluconazole (FLC) is widely used to prevent and treat invasive fungal infections. However, FLC is a fungistatic agent, allowing clinical FLC-susceptible isolates to tolerate FLC. Making FLC fungicidal in combination with adjuvants is a promising strategy to avoid FLC resistance and eliminate the persistence and recurrence of fungal infections. Here, we identify a new small molecule compound, CZ66, that can make FLC fungicidal. The mechanism of action of CZ66 is targeting the C-4 sterol methyl oxidase, encoded by the ERG251 gene, resulting in decreased content of sterols with the 14α-methyl group and ultimately eliminating FLC tolerance of Candida albicans. CZ66 most likely interacts with Erg251 through residues Glu195, Gly206, and Arg241. Establishing Erg251 as a synergistic lethal target protein of FLC should direct research to identify specific small molecule inhibitors of 14α-methylsterol synthesis and open the way to abolishing fungal FLC tolerance. IMPORTANCE Fluconazole (FLC) tolerance increases the frequency of acquired FLC resistance, and a high FLC tolerance level is associated with persistent candidemia. Multiple functional proteins, such as calcineurin, heat shock protein 90 (Hsp90), and ADP ribosylation factor, are essential for the survival of C. albicans exposed to FLC, but how these factors increase the fungicidal activity of FLC remains to be determined. In this study, we found that 14α-methylsterols replace ergosterol to allow C. albicans to survive FLC, but Erg251 inactivated by CZ66 results in loss of 14α-methylsterol synthesis and cell death of C. albicans treated with FLC. Establishing Erg251 as a synergistic lethal target protein of FLC should direct research to identify specific small molecule inhibitors of 14α-methylsterol synthesis and open the way to abolishing fungal FLC tolerance.
Collapse
Affiliation(s)
- Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wanqian Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Hongkang Wang
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shuo Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhe Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lan Yan
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Ting Fang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaolong Zhang
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Quanzhen Lv
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Zichao Ding
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Lijuan Qiu
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Dazhi Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
48
|
Deng H, Song J, Huang Y, Yang C, Zang X, Zhou Y, Li H, Dai B, Xue X. Combating increased antifungal drug resistance in Cryptococcus, what should we do in the future? Acta Biochim Biophys Sin (Shanghai) 2023; 55:540-547. [PMID: 36815374 PMCID: PMC10195138 DOI: 10.3724/abbs.2023011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/08/2022] [Indexed: 02/05/2023] Open
Abstract
Few therapeutic drugs and increased drug resistance have aggravated the current treatment difficulties of Cryptococcus in recent years. To better understand the antifungal drug resistance mechanism and treatment strategy of cryptococcosis. In this review, by combining the fundamental features of Cryptococcus reproduction leading to changes in its genome, we review recent research into the mechanism of four current anti-cryptococcal agents, coupled with new therapeutic strategies and the application of advanced technologies WGS and CRISPR-Cas9 in this field, hoping to provide a broad idea for the future clinical therapy of cryptococcosis.
Collapse
Affiliation(s)
- Hengyu Deng
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Jialin Song
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Yemei Huang
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Chen Yang
- Department of Laboratory Medicinethe First Medical CentreChinese PLA General HospitalBeijing100853China
| | - Xuelei Zang
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Yangyu Zhou
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Hongli Li
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Bin Dai
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Xinying Xue
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| |
Collapse
|
49
|
Branco J, Miranda IM, Rodrigues AG. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. J Fungi (Basel) 2023; 9:jof9010080. [PMID: 36675901 PMCID: PMC9862255 DOI: 10.3390/jof9010080] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Candida parapsilosis is the second most common Candida species isolated in Asia, Southern Europe, and Latin America and is often involved in invasive infections that seriously impact human health. This pathogen is part of the psilosis complex, which also includes Candida orthopsilosis and Candida metapsilosis. C. parapsilosis infections are particularly prevalent among neonates with low birth weights, individuals who are immunocompromised, and patients who require prolonged use of a central venous catheter or other indwelling devices, whose surfaces C. parapsilosis exhibits an enhanced capacity to adhere to and form biofilms. Despite this well-acknowledged prevalence, the biology of C. parapsilosis has not been as extensively explored as that of Candida albicans. In this paper, we describe the molecular mechanistic pathways of virulence in C. parapsilosis and show how they differ from those of C. albicans. We also describe the mode of action of antifungal drugs used for the treatment of Candida infections, namely, polyenes, echinocandins, and azoles, as well as the resistance mechanisms developed by C. parapsilosis to overcome them. Finally, we stress the importance of the ongoing search for species-specific features that may aid the development of effective control strategies and thus reduce the burden on patients and healthcare costs.
Collapse
Affiliation(s)
- Joana Branco
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Correspondence: ; Tel./Fax: +351-225513662
| | - Isabel M. Miranda
- Cardiovascular Research & Development Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Acácio G. Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
50
|
Wang H, Ji Z, Feng Y, Yan T, Cao Y, Lu H, Jiang Y. Myriocin enhances the antifungal activity of fluconazole by blocking the membrane localization of the efflux pump Cdr1. Front Pharmacol 2022; 13:1101553. [PMID: 36618949 PMCID: PMC9815617 DOI: 10.3389/fphar.2022.1101553] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction: Extrusion of azoles from the cell, mediated by an efflux pump Cdr1, is one of the most frequently used strategies for developing azole resistance in pathogenic fungi. The efflux pump Cdr1 is predominantly localized in lipid rafts within the plasma membrane, and its localization is sensitive to changes in the composition of lipid rafts. Our previous study found that the calcineurin signal pathway is important in transferring sphingolipids from the inner to the outer membrane. Methods: We investigated multiple factors that enhance the antifungal activity of fluconazole (FLC) using minimum inhibitory concentration (MIC) assays and disk diffusion assays. We studied the mechanism of action of myriocin through qRT-PCR analysis and confocal microscopy analysis. We tested whether myriocin enhanced the antifungal activity of FLC and held therapeutic potential using a mouse infection model. Results: We found that this signal pathway has no function in the activity of Cdr1. We found that inhibiting sphingolipid biosynthesis by myriocin remarkably increased the antifungal activity of FLC with a broad antifungal spectrum and held therapeutic potential. We further found that myriocin potently enhances the antifungal activity of FLC against C. albicans by blocking membrane localization of the Cdr1 rather than repressing the expression of Cdr1. In addition, we found that myriocin enhanced the antifungal activity of FLC and held therapeutic potential. Discussion: Our study demonstrated that blocking the membrane location and inactivating Cdr1 by inhibiting sphingolipids biogenesis is beneficial for enhancing the antifungal activity of azoles against azole-resistant C. albicans due to Cdr1 activation.
Collapse
Affiliation(s)
- Hongkang Wang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe Ji
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanru Feng
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianhua Yan
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongbing Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Lu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanying Jiang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|