1
|
Takemura K, Kolasinski V, Del Poeta M, Vieira de Sa NF, Garg A, Ojima I, Del Poeta M, Pereira de Sa N. Iron acquisition strategies in pathogenic fungi. mBio 2025:e0121125. [PMID: 40391928 DOI: 10.1128/mbio.01211-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
Iron plays a crucial role in various biological processes, including enzyme function, DNA replication, energy production, oxygen transport, lipid, and carbon metabolism. Although it is abundant in the Earth's crust, its bioavailability is restricted by the insolubility of ferric iron (Fe³+) and the auto-oxidation of ferrous iron (Fe²+) in oxygen-rich environments. This limitation poses significant challenges for all organisms, including fungi, which have developed intricate mechanisms for iron acquisition and utilization. These mechanisms include reductive iron uptake, siderophore production/transport, and heme utilization. Fungi employ a variety of enzymes-such as ferric reductases, ferroxidases, permeases, and transporters-to regulate intracellular iron levels effectively. The challenge is heightened for pathogenic fungi during infection, as they must compete with the host's iron-binding proteins like transferrin and lactoferrin, which sequester iron to restrict pathogen growth. This review delves into the iron acquisition strategies of medically important fungi, emphasizing the roles of reductive iron uptake and siderophore pathways. Understanding these mechanisms is vital for enhancing our knowledge of fungal pathogenesis and developing effective treatments. By targeting these iron acquisition processes, new antifungal therapies can be formulated more effectively to combat fungal infections.
Collapse
Affiliation(s)
- Kathryn Takemura
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Vanessa Kolasinski
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Matteo Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | | | - Ashna Garg
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Maurizio Del Poeta
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Veterans Affairs Medical Center, Northport, New York, USA
| | - Nivea Pereira de Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
2
|
Arekar T, Katikaneni D, Kasem S, Desai D, Acharya T, Cole A, Khodayari N, Vaulont S, Hube B, Nemeth E, Drakesmith A, Lionakis MS, Mehrad B, Scindia Y. Essential role of hepcidin in host resistance to disseminated candidiasis. Cell Rep 2025; 44:115649. [PMID: 40333187 DOI: 10.1016/j.celrep.2025.115649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/20/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Candida albicans is a leading cause of life-threatening invasive infection despite antifungal therapy. Patients with chronic liver disease are at increased risk of candidemia, but the mechanisms underlying this susceptibility are incompletely defined. One consequence of chronic liver disease is an attenuated ability to produce hepcidin and maintain organismal control of iron homeostasis. To address the biology underlying this critical clinical problem, we demonstrate the mechanistic link between hepcidin insufficiency and candida infection using genetic and inducible hepcidin knockout mice. Hepcidin deficiency led to unrestrained fungal growth and increased transition to the invasive hypha morphology with exposed 1,3-β-glucan, which exacerbated kidney injury, independent of the fungal pore-forming toxin candidalysin in immunocompetent mice. Of translational relevance, the therapeutic administration of PR-73, a hepcidin mimetic, improved the outcome of infection. Thus, we identify hepcidin deficiency as a host susceptibility factor against C. albicans and hepcidin mimetics as a potential intervention.
Collapse
Affiliation(s)
- Tanmay Arekar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Divya Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Sadat Kasem
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Dhruv Desai
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Thrisha Acharya
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Augustina Cole
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Nazli Khodayari
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Sophie Vaulont
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Elizabeta Nemeth
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexander Drakesmith
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Yogesh Scindia
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA; Center for Integrated Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
3
|
Lortal L, Lyon CM, Sprague JL, Sonnberger J, Paulin OKA, Wickramasinghe DN, Richardson JP, Hube B, Naglik JR. Candidalysin biology and activation of host cells. mBio 2025:e0060324. [PMID: 40293285 DOI: 10.1128/mbio.00603-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can cause life-threatening systemic infections and distressing mucosal infections. A major breakthrough in understanding C. albicans pathogenicity was the discovery of candidalysin, the first cytolytic peptide toxin identified in a human pathogenic fungus. Secreted by C. albicans hyphae and encoded by the ECE1 gene, this 31-amino acid peptide integrates into and permeabilizes host cell membranes, causing damage across diverse cell types. Beyond its cytolytic activity, candidalysin can trigger potent innate immune responses in epithelial cells, macrophages, and neutrophils. Additionally, candidalysin plays a key role in nutrient acquisition during infection. This review explores the biology of candidalysin, its role in host cell activation, and extends the discussion to non-candidalysin Ece1p peptides, shedding light on their emerging significance.
Collapse
Affiliation(s)
- Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Claire M Lyon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Johannes Sonnberger
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Olivia K A Paulin
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Don N Wickramasinghe
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
4
|
Chai X, Jiang Y, Lu H, Huang X. Integrating ensemble machine learning and multi-omics approaches to identify Dp44mT as a novel anti- Candida albicans agent targeting cellular iron homeostasis. Front Pharmacol 2025; 16:1574990. [PMID: 40342996 PMCID: PMC12058677 DOI: 10.3389/fphar.2025.1574990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Introduction Candidiasis, mainly caused by Candida albicans, poses a serious threat to human health. The escalating drug resistance in C. albicans and the limited antifungal options highlight the critical need for novel therapeutic strategies. Methods We evaluated 12 machine learning models on a self-constructed dataset with known anti-C. albicans activity. Based on their performance, the optimal model was selected to screen our separate in-house compound library with unknown anti-C. albicans activity for potential antifungal agents. The anti-C. albicans activity of the selected compounds was confirmed through in vitro drug susceptibility assays, hyphal growth assays, and biofilm formation assays. Through transcriptomics, proteomics, iron rescue experiments, CTC staining, JC-1 staining, DAPI staining, molecular docking, and molecular dynamics simulations, we elucidated the mechanism underlying the anti-C. albicans activity of the compound. Result Among the evaluated machine learning models, the best predictive model was an ensemble learning model constructed from Random Forests and Categorical Boosting using soft voting. It predicts that Dp44mT exhibits potent anti-C. albicans activity. The in vitro tests further verified this finding that Dp44mT can inhibit planktonic growth, hyphal formation, and biofilm formation of C. albicans. Mechanistically, Dp44mT exerts antifungal activity by disrupting cellular iron homeostasis, leading to a collapse of mitochondrial membrane potential and ultimately causing apoptosis. Conclusion This study presents a practical approach for predicting the antifungal activity of com-pounds using machine learning models and provides new insights into the development of antifungal compounds by disrupting iron homeostasis in C. albicans.
Collapse
Affiliation(s)
- Xiaowei Chai
- Department of Dermatology, Hair Medical Center of Shanghai Tongji Hospital, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Lu
- Department of Pharmacy, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Huang
- Department of Dermatology, Hair Medical Center of Shanghai Tongji Hospital, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Yang MC, Huang WL, Chen HY, Lin SH, Chang YS, Tseng KY, Lo HJ, Wang IC, Lin CJ, Lan CY. Deletion of RAP1 affects iron homeostasis, azole resistance, and virulence in Candida albicans. mSphere 2025:e0015525. [PMID: 40265929 DOI: 10.1128/msphere.00155-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/29/2025] [Indexed: 04/24/2025] Open
Abstract
Rap1 is a DNA-binding protein conserved from yeast to mammals for its role in telomeric maintenance. Here, to explore additional functions of Candida albicans Rap1, we performed RNA sequencing analysis. Experimental validations further showed that Rap1 plays a role in iron regulation, especially under low-iron conditions. Moreover, Rap1 was involved in iron acquisition and modulation of iron-related genes. Rap1 was found to be associated with fluconazole resistance in a low-iron condition. Finally, we demonstrated that the deletion of RAP1 leads to reduced C. albicans virulence in a mouse model of infection. Together, this study reveals new functions of C. albicans Rap1, particularly in iron homeostasis, azole resistance, and virulence. IMPORTANCE Candida albicans is an important pathogenic fungus that can cause superficial to life-threatening infections. Iron is essential for almost all organisms, yet it is highly restricted within the human host to defend against pathogens. To grow and survive in the iron-limited host environment, C. albicans has evolved multiple iron acquisition mechanisms. Understanding the regulation of iron homeostasis is, therefore, critical for elucidating C. albicans pathogenesis and virulence. This study explores the novel functions of C. albicans Rap1, with a focus on its contribution to iron acquisition and utilization. Our findings further highlight how iron availability impacts antifungal resistance and virulence through Rap1, providing insight into the complex iron regulatory machinery of C. albicans.
Collapse
Affiliation(s)
- Min-Chi Yang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Wei-Luen Huang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsuan-Yu Chen
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shin-Huey Lin
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Shan Chang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Kuo-Yun Tseng
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Taiwan Mycology Reference Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsiu-Jung Lo
- Taiwan Mycology Reference Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - I-Ching Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Jan Lin
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Chung-Yu Lan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
6
|
Schille TB, Sprague JL, Naglik JR, Brunke S, Hube B. Commensalism and pathogenesis of Candida albicans at the mucosal interface. Nat Rev Microbiol 2025:10.1038/s41579-025-01174-x. [PMID: 40247134 DOI: 10.1038/s41579-025-01174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
Fungi are important and often underestimated human pathogens. Infections with fungi mostly originate from the environment, from soil or airborne spores. By contrast, Candida albicans, one of the most common and clinically important fungal pathogens, permanently exists in the vast majority of healthy individuals as a member of the human mucosal microbiota. Only under certain circumstances will these commensals cause infections. However, although the pathogenic behaviour and disease manifestation of C. albicans have been at the centre of research for many years, its asymptomatic colonization of mucosal surfaces remains surprisingly understudied. In this Review, we discuss the interplay of the fungus, the host and the microbiome on the dualism of commensal and pathogenic life of C. albicans, and how commensal growth is controlled and permitted. We explore hypotheses that could explain how the mucosal environment shapes C. albicans adaptations to its commensal lifestyle, while still maintaining or even increasing its pathogenic potential.
Collapse
Affiliation(s)
- Tim B Schille
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
7
|
Lee CH, Wu CJ, Yen FY, Chiang JY, Shen TJ, Leu SJ, Chang CR, Lo HJ, Tsai BY, Mao YC, Andriani V, Thenaka PC, Wang WC, Chao YP, Yang YY. Identification of chicken-derived antibodies targeting the Candida albicans Als3 protein. Appl Microbiol Biotechnol 2025; 109:85. [PMID: 40198376 PMCID: PMC11978541 DOI: 10.1007/s00253-025-13469-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
Candida albicans is a major opportunistic pathogen, responsible for nearly half of clinical candidemia cases. The rising prevalence of azole-resistant Candida species represents a significant clinical challenge, underscoring the urgent need for alternative therapeutic strategies. Monoclonal antibody-based therapies have emerged as a promising and cost-effective approach to combating Candida infections. Agglutinin-like sequence protein 3 (Als3), a key cell surface protein of C. albicans, plays a pivotal role in adherence and biofilm formation, both of which are essential for its pathogenesis. In this study, recombinant Als3 protein was purified and utilized to immunize chickens, resulting in the production of Als3-specific immunoglobulin Y (IgY) antibodies. Two single-chain variable fragment (scFv) antibody libraries were subsequently constructed using phage display technology, yielding transformant counts of 5.3 × 107 and 2.8 × 107, respectively. Phage-based enzyme-linked immunosorbent assay (ELISA) revealed enhanced signals following bio-panning, enabling the identification and sequence validation of three scFv antibodies. These scFv antibodies exhibited strong binding activities to Als3, as confirmed through ELISA and western blot analyses. Binding affinities were determined to be ~ 10⁻⁸ M via serial titration ELISA and competitive ELISA. Additionally, the selected scFv antibodies specifically recognized endogenous Als3 protein in C. albicans, as demonstrated by western blot and cell-based ELISA assays. In conclusion, this study successfully generated and characterized high-affinity scFv antibodies targeting Als3, which exhibited exceptional specificity and binding activity. These findings highlight their potential as promising immunotherapeutic candidates for the treatment of C. albicans infections. KEY POINTS: • The Als3 protein of C. albicans is a critical biomarker and therapeutic target • Chicken-derived scFv antibodies against Als3 were developed via phage display • The scFv antibodies showed strong binding to endogenous Als3 in C. albicans.
Collapse
Affiliation(s)
- Chi-Hsin Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University Inc., Taipei, 110301, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chao-Jung Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University Inc., Taipei, 110301, Taiwan
| | - Fang-Yi Yen
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
| | - Jia-Yun Chiang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
| | - Ting-Jing Shen
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University Inc., Taipei, 110301, Taiwan
| | - Sy-Jye Leu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chuang-Rung Chang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Hsiu-Jung Lo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 350401, Taiwan
| | - Bor-Yu Tsai
- Navi Bio-Therapeutics Inc., Taipei, 10351, Taiwan
| | - Yan-Chiao Mao
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung, 407219, Taiwan
| | - Valencia Andriani
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
| | - Priskila Cherisca Thenaka
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan
| | - Wei-Chu Wang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University Inc., Taipei, 110301, Taiwan
| | - Yu-Pin Chao
- iReal Biotechnology Inc., Hsinchu, 30060, Taiwan
| | - Yi-Yuan Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235, Taiwan.
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University Inc., Taipei, 110301, Taiwan.
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, 110301, Taiwan.
| |
Collapse
|
8
|
Chiang HS, Chen JH, Liao YT, Peng YC, Hsu CC, Ke CL, Chung CT, Yeh YC, Tsai HY, Lin CH. MNN45 is involved in Zcf31-mediated cell surface integrity and chitosan susceptibility in Candida albicans. Med Mycol 2025; 63:myaf025. [PMID: 40118513 DOI: 10.1093/mmy/myaf025] [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: 01/08/2025] [Revised: 02/17/2025] [Accepted: 03/20/2025] [Indexed: 03/23/2025] Open
Abstract
Candida albicans is a major human fungal pathogen; however, limited antifungal agents, undesirable drug side effects, and ineffective prevention of drug-resistant strains have become serious problems. Chitosan is a nontoxic, biodegradable, and biocompatible linear polysaccharide made from the deacetylation of chitin. In this study, a ZCF31 putative transcription factor gene was selected from a previous mutant library screen, as zcf31Δ strains exhibited defective cell growth in response to chitosan. Furthermore, chitosan caused notable damage to zcf31Δ cells; however, ZCF31 expression was not significantly changed by chitosan, suggesting that zcf31Δ is sensitive to chitosan could be due to changes in the physical properties of C. albicans. Indeed, zcf31Δ cells displayed significant increases in cell wall thickness. Consistent with the previous study, zcf31Δ strains were resistant to calcofluor white but highly susceptible to SDS (sodium dodecyl sulfate). These results implied that chitosan mainly influences membrane function, as zcf31Δ strengthens the stress resistance of the fungal cell wall but lessens cell membrane function. Interestingly, this effect on the cell surface mechanics of the C. albicans zcf31Δ strains was not responsible for the virulence-associated function. RNA-seq analysis further revealed that six mannosyltransferase-related genes were upregulated in zcf31Δ. Although five mannosyltransferase-related mutant strains in the zcf31Δ background partially reduced the cell wall thickness, only zcf31Δ/mnn45Δ showed the recovery of chitosan resistance. Our findings suggest that Zcf31 mediates a delicate and complicated dynamic balance between the cell membrane and cell wall architectures through the mannosyltransferase genes in C. albicans, leading to altered chitosan susceptibility.
Collapse
Affiliation(s)
- Hao-Sen Chiang
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Ji-Hong Chen
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Yu-Ting Liao
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Yu-Chun Peng
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Chih-Chieh Hsu
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Cai-Ling Ke
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Chi-Ting Chung
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Yu-Chiao Yeh
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Hsiao-Yen Tsai
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| | - Ching-Hsuan Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan, 10617
| |
Collapse
|
9
|
Chougule S, Patil S, Gavandi T, Basrani S, Jadhav AK, Karuppayil SM. Alpha-bisabolol inhibits yeast to hyphal form transition and biofilm development in Candida albicans: in vitro and in silico studies. In Silico Pharmacol 2025; 13:53. [PMID: 40182396 PMCID: PMC11961840 DOI: 10.1007/s40203-025-00335-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
In recent years, there has been growing concern about infections caused by Candida albicans, which pose a significant threat to human health. This intensifies the concern that can be largely attributed to the increasing number of people with compromised immune systems and the emergence of drug-resistant strains. Natural molecules are considered to be good alternatives to synthetic antifungal agents. The present study explored the effectiveness of alpha-bisabolol as an antifungal agent and its mechanism of action against C. albicans ATCC90028. α-bisabolol effectively inhibited various pathogenic traits of C. albicans like, adhesion, yeast to hyphal switching, and development of biofilm at 1 mg/ml, 0.25 mg/ml, and 0.125 mg/ml concentration, respectively. In addition, α-bisabolol demonstrated inhibition of cell cycle propagation at the G1 phase. Ergosterol production in the C. albicans was suppressed by α-bisabolol treatment in a dose-dependent manner. The molecular docking study revealed α-bisabolol has a good binding energy of - 7.11 kcal/mol with 14-α-demethylase enzyme, which is crucial for ergosterol synthesis. Therefore, the cell membrane integrity may be affected by treatment with α-bisabolol. qRT-PCR studies proved that α-bisabolol treatment affects gene expression in C. albicans. In silico binding affinity was also analyzed for RAS1, TUP1 and CST20 in the signal transduction pathway and exhibited binding affinities for at - 7.7 kcal/mol, - 8.21 kcal/mol, and for - 5.79 kcal/mol respectively. In conclusion, α-bisabolol caused reduced biofilm, ergosterol synthesis along with altered gene expressions in C. albicans with no hemolysis. This study proposed α-bisabolol as an alternative antifungal agent.
Collapse
Affiliation(s)
- Sayali Chougule
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra 416003 India
| | - Shivani Patil
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra 416003 India
| | - Tanjila Gavandi
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra 416003 India
| | - Sargun Basrani
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra 416003 India
| | - Ashwini K. Jadhav
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra 416003 India
| | - S. Mohan Karuppayil
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra 416003 India
| |
Collapse
|
10
|
Jong T, Stack CM, Moffitt MC, Morton CO. An Introduction to the Influence of Nutritional Factors on the Pathogenesis of Opportunist Fungal Pathogens in Humans. Pathogens 2025; 14:335. [PMID: 40333109 PMCID: PMC12030028 DOI: 10.3390/pathogens14040335] [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: 01/17/2025] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Fungi such as Aspergillus fumigatus, Candida albicans, and Cryptococcus neoformans are opportunistic pathogens in humans. They usually infect individuals whose immune system is compromised due to either a primary infection, e.g., HIV/AIDS, or as part of treatment for another condition, e.g., stem cell or solid organ transplant. In hosts with a weakened immune system, these fungi can cause life-threatening infections. Unlike true pathogens, opportunistic pathogens do not have specific mechanisms to overcome a healthy host, requiring a different approach to understand how they cause infection. The ability of fungi to adapt to various environmental conditions, including the human host, is critical for virulence. In humans, micronutrient metals, such as iron, are sequestered to reduce serum concentrations, which helps to inhibit microbial growth. Other human tissues may increase metal concentrations to toxic levels to prevent infection by pathogens. The ability of fungi to acquire or detoxify nutrients, such as iron or copper, from the host is essential for the establishment of infection. In this review, the role of fungal nutrition will be discussed in relation to opportunistic fungal pathogens. It will focus on the acquisition of micronutrients, e.g., iron, copper, and zinc, and how this enables these fungi to circumvent host nutritional immunity.
Collapse
Affiliation(s)
| | | | | | - Charles Oliver Morton
- Western Sydney University, School of Science, Campbelltown, NSW 2560, Australia (C.M.S.); (M.C.M.)
| |
Collapse
|
11
|
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
|
12
|
Li Y, Pham T, Hipsher K, Lee CWJ, Jiao J, Penninger JM, Kronstad JW, Fan Y, Zhao Y, Ambati S, Meagher RB, Xie X, Lin X. Identification of a protective antigen reveals the trade-off between iron acquisition and antigen exposure in a global fungal pathogen. Proc Natl Acad Sci U S A 2025; 122:e2420898122. [PMID: 39946532 PMCID: PMC11848283 DOI: 10.1073/pnas.2420898122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/09/2025] [Indexed: 02/26/2025] Open
Abstract
Systemic infections caused by Cryptococcus claim over 161,000 lives annually, with global mortality rate close to 70% despite antifungal therapies. Currently, no vaccine is available. To develop an effective multivalent vaccine against this free-living opportunistic eukaryotic pathogen, it is critical to identify protective antigens. We previously discovered ZNF2oe strains elicit protective host immune responses and increase the abundance of antigens present in the capsule, which is required for its immunoprotection. Capsule is a defining feature of Cryptococcus species and composed of polysaccharides and mannoproteins. Here, we found increased levels of exposed mannoproteins in ZNF2oe cells. As mannoproteins are the primary components recognized by anticryptococcal cell-mediated immune responses and few have been characterized, we systemically screened all 49 predicted GPI-mannoproteins in Cryptococcus neoformans for enhanced host recognition. We identified those highly present in ZNF2oe cells and found Cig1 to be a protective antigen against cryptococcosis either as a recombinant protein vaccine or an mRNA vaccine. Cig1 is induced by iron limitation and is highly expressed by this fungus in infected mice and in patients with cryptococcal meningitis. Remarkably, iron restriction by the host induces cryptococcal cells to express iron-uptake proteins including Cig1, which act as cryptococcal antigens and in turn enhance host detection. Our results highlight an arms race between the pathogen and the host centered on iron competition, and the trade-off between cryptococcal iron acquisition and antigen exposure. These findings demonstrate the potential of leveraging this host-pathogen interaction for vaccine development.
Collapse
Affiliation(s)
- Yeqi Li
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Tuyetnhu Pham
- Department of Plant Biology, University of Georgia, Athens, GA30602
| | - Kenton Hipsher
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Christopher W. J. Lee
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Jie Jiao
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Josef M. Penninger
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
- Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Vienna1090, Austria
- Helmholtz Centre for Infection Research, Braunschweig38124, Germany
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna1030, Austria
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Yumeng Fan
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Youbao Zhao
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Suresh Ambati
- Department of Genetics, University of Georgia, Athens, GA30602
| | | | - Xiaofeng Xie
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA30602
- Department of Plant Biology, University of Georgia, Athens, GA30602
| |
Collapse
|
13
|
Valentine M, Wilson D, Gresnigt MS, Hube B. Vaginal Candida albicans infections: host-pathogen-microbiome interactions. FEMS Microbiol Rev 2025; 49:fuaf013. [PMID: 40347186 PMCID: PMC12071381 DOI: 10.1093/femsre/fuaf013] [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: 09/20/2024] [Revised: 04/14/2025] [Accepted: 05/09/2025] [Indexed: 05/12/2025] Open
Abstract
Candida albicans is a fungus that colonizes the gut, oral, and vaginal mucosae of most humans without causing disease. However, under certain predisposing conditions this fungus can cause disease. Candida albicans has several factors and attributes that facilitate its commensal and pathogenic lifestyles including the transition from a yeast to a hyphal morphology, which is accompanied by the expression of virulence factors. These factors are central in candidiasis that can range from invasive to superficial. This review focuses on one example of a superficial disease, i.e. vulvovaginal candidiasis (VVC) that affects ~75% of women at least once with some experiencing four or more symptomatic infections per year (RVVC). During VVC, fungal factors trigger inflammation, which is maintained by a dysregulated innate immune response. This in turn leads to immunopathology and symptoms. Another unique characteristic of the vaginal niche, is its Lactobacillus-dominated microbiota with low species diversity that is believed to antagonize C. albicans pathogenicity. The importance of the interactions between C. albicans, the host, and vaginal microbiota during commensalism and (R)VVC is discussed in this review, which also addresses the application of this knowledge to identify novel treatment strategies and to study vaginal C. albicans infections.
Collapse
Affiliation(s)
- Marisa Valentine
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 23 Adolf-Reichwein-Straße, 07745, Jena, Germany
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology at the University of Exeter, University of Exeter, Geoffrey Pope Building Stocker Road, Exeter EX4 4QD, United Kingdom
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 23 Adolf-Reichwein-Straße, 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 23 Adolf-Reichwein-Straße, 07745, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 25 Neugasse, 07743, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 1 Fürstengraben, 07743, Jena, Germany
| |
Collapse
|
14
|
Hitzler SUJ, Fernández-Fernández C, Montaño DE, Dietschmann A, Gresnigt MS. Microbial adaptive pathogenicity strategies to the host inflammatory environment. FEMS Microbiol Rev 2025; 49:fuae032. [PMID: 39732621 PMCID: PMC11737513 DOI: 10.1093/femsre/fuae032] [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: 09/11/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 12/30/2024] Open
Abstract
Pathogenic microorganisms can infect a variety of niches in the human body. During infection, these microbes can only persist if they adapt adequately to the dynamic host environment and the stresses imposed by the immune system. While viruses entirely rely on host cells to replicate, bacteria and fungi use their pathogenicity mechanisms for the acquisition of essential nutrients that lie under host restriction. An inappropriate deployment of pathogenicity mechanisms will alert host defence mechanisms that aim to eradicate the pathogen. Thus, these adaptations require tight regulation to guarantee nutritional access without eliciting strong immune activation. To work efficiently, the immune system relies on a complex signalling network, involving a myriad of immune mediators, some of which are quite directly associated with imminent danger for the pathogen. To manipulate the host immune system, viruses have evolved cytokine receptors and viral cytokines. However, among bacteria and fungi, selected pathogens have evolved the capacity to use these inflammatory response-specific signals to regulate their pathogenicity. In this review, we explore how bacterial and fungal pathogens can sense the immune system and use adaptive pathogenicity strategies to evade and escape host defence to ensure their persistence in the host.
Collapse
Affiliation(s)
- Sophia U J Hitzler
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Candela Fernández-Fernández
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Dolly E Montaño
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| |
Collapse
|
15
|
Gómez-Gaviria M, Baruch-Martínez DA, Mora-Montes HM. Exploring the Biology, Virulence, and General Aspects of Candida dubliniensis. Infect Drug Resist 2024; 17:5755-5773. [PMID: 39722735 PMCID: PMC11669290 DOI: 10.2147/idr.s497862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Fungal infections have become a growing public health concern, aggravated by the emergence of new pathogenic species and increasing resistance to antifungal drugs. The most common candidiasis is caused by Candida albicans; however, Candida dubliniensis has become an emerging opportunistic pathogen, and although less prevalent, it can cause superficial and systemic infections, especially in immunocompromised individuals. This yeast can colonize the oral cavity, skin, and other tissues, and has been associated with oral infections in patients with human immunodeficiency virus (HIV) and acquired immunodeficiency syndrome (AIDS), making it difficult to treat. The special interest in the study of this species lies in its ability to evade commonly used antifungal drugs, such as fluconazole, under different concentrations. In addition, it is difficult to identify because it can be confused with the species C. albicans, which could interfere with adequate treatment. Although the study of virulence factors in C. dubliniensis is limited, proteomic comparisons with C. albicans indicate that these virulence factors could be similar between the two species. However, differences could exist considering the evolutionary processes and lifestyle of each species. In this study, a detailed review of the current literature on C. dubliniensis was conducted, considering aspects such as biology, possible virulence factors, immune response, pathogen-host interaction, diagnosis, and treatment.
Collapse
Affiliation(s)
- Manuela Gómez-Gaviria
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto, Mexico
| | - Dario A Baruch-Martínez
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto, Mexico
| | - Héctor M Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto, Mexico
| |
Collapse
|
16
|
Brown GD, Ballou ER, Bates S, Bignell EM, Borman AM, Brand AC, Brown AJP, Coelho C, Cook PC, Farrer RA, Govender NP, Gow NAR, Hope W, Hoving JC, Dangarembizi R, Harrison TS, Johnson EM, Mukaremera L, Ramsdale M, Thornton CR, Usher J, Warris A, Wilson D. The pathobiology of human fungal infections. Nat Rev Microbiol 2024; 22:687-704. [PMID: 38918447 DOI: 10.1038/s41579-024-01062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
Human fungal infections are a historically neglected area of disease research, yet they cause more than 1.5 million deaths every year. Our understanding of the pathophysiology of these infections has increased considerably over the past decade, through major insights into both the host and pathogen factors that contribute to the phenotype and severity of these diseases. Recent studies are revealing multiple mechanisms by which fungi modify and manipulate the host, escape immune surveillance and generate complex comorbidities. Although the emergence of fungal strains that are less susceptible to antifungal drugs or that rapidly evolve drug resistance is posing new threats, greater understanding of immune mechanisms and host susceptibility factors is beginning to offer novel immunotherapeutic options for the future. In this Review, we provide a broad and comprehensive overview of the pathobiology of human fungal infections, focusing specifically on pathogens that can cause invasive life-threatening infections, highlighting recent discoveries from the pathogen, host and clinical perspectives. We conclude by discussing key future challenges including antifungal drug resistance, the emergence of new pathogens and new developments in modern medicine that are promoting susceptibility to infection.
Collapse
Affiliation(s)
- Gordon D Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Elizabeth R Ballou
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Steven Bates
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elaine M Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Andrew M Borman
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alexandra C Brand
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Carolina Coelho
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Peter C Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rhys A Farrer
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Nelesh P Govender
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - William Hope
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - J Claire Hoving
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rachael Dangarembizi
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Thomas S Harrison
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elizabeth M Johnson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Liliane Mukaremera
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark Ramsdale
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
17
|
Arekar T, Katikaneni D, Kasem S, Desai D, Acharya T, Cole A, Khodayari N, Vaulont S, Hube B, Nemeth E, Drakesmith A, Lionakis MS, Mehrad B, Scindia Y. Essential role of Hepcidin in host resistance to disseminated candidiasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620511. [PMID: 39553949 PMCID: PMC11565830 DOI: 10.1101/2024.10.29.620511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Candida albicans is a leading cause of life-threatening invasive infections with up to 40% mortality rates in hospitalized individuals despite antifungal therapy. Patients with chronic liver disease are at an increased risk of candidemia, but the mechanisms underlying this susceptibility are incompletely defined. One consequence of chronic liver disease is attenuated ability to produce hepcidin and maintain organismal control of iron homeostasis. To address the biology underlying this critical clinical problem, we demonstrate the mechanistic link between hepcidin insufficiency and candida infection using genetic and inducible hepcidin knockout mice. Hepcidin deficiency led to unrestrained fungal growth and increased transition to the invasive hypha morphology with exposed 1,3, β-glucan that exacerbated kidney injury, independent of the fungal pore-forming toxin candidalysin in immunocompetent mice. Of translational relevance, the therapeutic administration of PR-73, a hepcidin mimetic, improved the outcomes of infection. Thus, we identify hepcidin deficiency as a novel host susceptibility factor against C. albicans and hepcidin mimetics as a potential intervention.
Collapse
|
18
|
Gao Y, Cao Q, Xiao Y, Wu Y, Ding L, Huang H, Li Y, Yang J, Meng L. The progress and future of the treatment of Candida albicans infections based on nanotechnology. J Nanobiotechnology 2024; 22:568. [PMID: 39285480 PMCID: PMC11406819 DOI: 10.1186/s12951-024-02841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024] Open
Abstract
Systemic infection with Candida albicans poses a significant risk for people with weakened immune systems and carries a mortality rate of up to 60%. However, current therapeutic options have several limitations, including increasing drug tolerance, notable off-target effects, and severe adverse reactions. Over the past four decades, the progress in developing drugs to treat Candida albicans infections has been sluggish. This comprehensive review addresses the limitations of existing drugs and summarizes the efforts made toward redesigning and innovating existing or novel drugs through nanotechnology. The discussion explores the potential applications of nanomedicine in Candida albicans infections from four perspectives: nano-preparations for anti-biofilm therapy, innovative formulations of "old drugs" targeting the cell membrane and cell wall, reverse drug resistance therapy targeting subcellular organelles, and virulence deprivation therapy leveraging the unique polymorphism of Candida albicans. These therapeutic approaches are promising to address the above challenges and enhance the efficiency of drug development for Candida albicans infections. By harnessing nano-preparation technology to transform existing and preclinical drugs, novel therapeutic targets will be uncovered, providing effective solutions and broader horizons to improve patient survival rates.
Collapse
Affiliation(s)
- Yang Gao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Qinyan Cao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yuyang Xiao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yue Wu
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Liang Ding
- Nanjing Stomatological Hospital, Nanjing, 210008, China
| | - He Huang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yanan Li
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Jingpeng Yang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Lingtong Meng
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
19
|
Murante D, Hogan DA. Drivers of diversification in fungal pathogen populations. PLoS Pathog 2024; 20:e1012430. [PMID: 39264909 PMCID: PMC11392411 DOI: 10.1371/journal.ppat.1012430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Abstract
To manage and treat chronic fungal diseases effectively, we require an improved understanding of their complexity. There is an increasing appreciation that chronic infection populations are often heterogeneous due to diversification and drift, even within a single microbial species. Genetically diverse populations can contribute to persistence and resistance to treatment by maintaining cells with different phenotypes capable of thriving in these dynamic environments. In chronic infections, fungal pathogens undergo prolonged challenges that can drive trait selection to convergent adapted states through restricted access to critical nutrients, assault by immune effectors, competition with other species, and antifungal drugs. This review first highlights the various genetic and epigenetic mechanisms that promote diversity in pathogenic fungal populations and provide an additional barrier to assessing the actual heterogeneity of fungal infections. We then review existing studies of evolution and genetic heterogeneity in fungal populations from lung infections associated with the genetic disease cystic fibrosis. We conclude with a discussion of open research questions that, once answered, may aid in diagnosing and treating chronic fungal infections.
Collapse
Affiliation(s)
- Daniel Murante
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Deborah Ann Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
20
|
Lim SJ, Noor NDM, Sabri S, Ali MSM, Salleh AB, Oslan SN. Extracellular BSA-degrading SAPs in the rare pathogen Meyerozyma guilliermondii strain SO as potential virulence factors in candidiasis. Microb Pathog 2024; 193:106773. [PMID: 38960213 DOI: 10.1016/j.micpath.2024.106773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 05/08/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Meyerozyma guilliermondii (Candida guilliermondii) is one of the Candida species associated with invasive candidiasis. With the potential for expressing industrially important enzymes, M. guilliermondii strain SO possessed 99 % proteome similarity with the clinical ATCC 6260 isolate and showed pathogenicity towards zebrafish embryos. Recently, three secreted aspartyl proteinases (SAPs) were computationally identified as potential virulence factors in this strain without in vitro verification of SAP activity. The quantification of Candida SAPs activity in liquid broth were also scarcely reported. Thus, this study aimed to characterize M. guilliermondii strain SO's ability to produce SAPs (MgSAPs) in different conditions (morphology and medium) besides analyzing its growth profile. MgSAPs' capability to cleave bovine serum albumin (BSA) was also determined to propose that MgSAPs as the potential virulence factors compared to the avirulent Saccharomyces cerevisiae. M. guilliermondii strain SO produced more SAPs (higher activity) in yeast nitrogen base-BSA-dextrose broth compared to yeast extract-BSA-dextrose broth despite insignificantly different SAP activity in both planktonic and biofilm cells. FeCl3 supplementation significantly increased the specific protein activity (∼40 %). The BSA cleavage by MgSAPs at an acidic pH was proven through semi-quantitative SDS-PAGE, sharing similar profile with HIV-1 retropepsin. The presented work highlighted the MgSAPs on fungal cell wall and extracellular milieu during host infection could be corroborated to the quantitative production in different growth modes presented herein besides shedding lights on the potential usage of retropepsin's inhibitors in treating candidiasis. Molecular and expression analyses of MgSAPs and their deletion should be further explored to attribute their respective virulence effects.
Collapse
Affiliation(s)
- Si Jie Lim
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Noor Dina Muhd Noor
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Suriana Sabri
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Mohd Shukuri Mohamad Ali
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Abu Bakar Salleh
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Siti Nurbaya Oslan
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
21
|
Van Genechten W, Vergauwen R, Van Dijck P. The intricate link between iron, mitochondria and azoles in Candida species. FEBS J 2024; 291:3568-3580. [PMID: 37846606 DOI: 10.1111/febs.16977] [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: 05/21/2023] [Revised: 09/19/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Invasive fungal infections are rapidly increasing, and the opportunistic pathogenic Candida species are the fourth most common cause of nosocomial systemic infections. The current antifungal classes, of which azoles are the most widely used, all have shortcomings. Azoles are generally considered fungistatic rather than fungicidal, they do not actively kill fungal cells and therefore resistance against azoles can be rapidly acquired. Combination therapies with azoles provide an interesting therapeutic outlook and agents limiting iron are excellent candidates. We summarize how iron is acquired by the host and transported towards both storage and iron-utilizing organelles. We indicate whether these pathways alter azole susceptibility and/or tolerance, to finally link these transport mechanisms to mitochondrial iron availability. In this review, we highlight putative novel intracellular iron shuffling mechanisms and indicate that mitochondrial iron dynamics in relation to azole treatment and iron limitation is a significant knowledge gap.
Collapse
Affiliation(s)
- Wouter Van Genechten
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| | - Rudy Vergauwen
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| |
Collapse
|
22
|
Xiong L, Goerlich K, Do E, Mitchell AP. Strain variation in the Candida albicans iron limitation response. mSphere 2024; 9:e0037224. [PMID: 38980069 PMCID: PMC11288005 DOI: 10.1128/msphere.00372-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/16/2024] [Indexed: 07/10/2024] Open
Abstract
Iron acquisition is critical for pathogens to proliferate during invasive infection, and the human fungal pathogen Candida albicans is no exception. The iron regulatory network, established in reference strain SC5314 and derivatives, includes the central player Sef1, a transcription factor that activates iron acquisition genes in response to iron limitation. Here, we explored potential variation in this network among five diverse C. albicans strains through mutant analysis, Nanostring gene expression profiling, and, for two strains, RNA-Seq. Our findings highlight four features that may inform future studies of natural variation and iron acquisition in this species. (i) Conformity: In all strains, major iron acquisition genes are upregulated during iron limitation, and a sef1Δ/Δ mutation impairs that response and growth during iron limitation. (ii) Response variation: Some aspects of the iron limitation response vary among strains, notably the activation of hypha-associated genes. As this gene set is tied to tissue damage and virulence, variation may impact the progression of infection. (iii) Genotype-phenotype variation: The impact of a sef1Δ/Δ mutation on cell wall integrity varies, and for the two strains examined the phenotype correlated with sef1Δ/Δ impact on several cell wall integrity genes. (iv) Phenotype discovery: DNA repair genes were induced modestly by iron limitation in sef1Δ/Δ mutants, with fold changes we would usually ignore. However, the response occurred in both strains tested and was reminiscent of a much stronger response described in Cryptococcus neoformans, a suggestion that it may have biological meaning. In fact, we observed that the iron limitation of a sef1Δ/Δ mutant caused recessive phenotypes to emerge at two heterozygous loci. Overall, our results show that a network that is critical for pathogen proliferation presents variation outside of its core functions.IMPORTANCEA key virulence factor of Candida albicans is the ability to maintain iron homeostasis in the host where iron is scarce. We focused on a central iron regulator, SEF1. We found that iron regulator Sef1 is required for growth, cell wall integrity, and genome integrity during iron limitation. The novel aspect of this work is the characterization of strain variation in a circuit that is required for survival in the host and the connection of iron acquisition to genome integrity in C. albicans.
Collapse
Affiliation(s)
- Liping Xiong
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | | | - Eunsoo Do
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
23
|
Katsipoulaki M, Stappers MHT, Malavia-Jones D, Brunke S, Hube B, Gow NAR. Candida albicans and Candida glabrata: global priority pathogens. Microbiol Mol Biol Rev 2024; 88:e0002123. [PMID: 38832801 PMCID: PMC11332356 DOI: 10.1128/mmbr.00021-23] [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] [Indexed: 06/05/2024] Open
Abstract
SUMMARYA significant increase in the incidence of Candida-mediated infections has been observed in the last decade, mainly due to rising numbers of susceptible individuals. Recently, the World Health Organization published its first fungal pathogen priority list, with Candida species listed in medium, high, and critical priority categories. This review is a synthesis of information and recent advances in our understanding of two of these species-Candida albicans and Candida glabrata. Of these, C. albicans is the most common cause of candidemia around the world and is categorized as a critical priority pathogen. C. glabrata is considered a high-priority pathogen and has become an increasingly important cause of candidemia in recent years. It is now the second most common causative agent of candidemia in many geographical regions. Despite their differences and phylogenetic divergence, they are successful as pathogens and commensals of humans. Both species can cause a broad variety of infections, ranging from superficial to potentially lethal systemic infections. While they share similarities in certain infection strategies, including tissue adhesion and invasion, they differ significantly in key aspects of their biology, interaction with immune cells, host damage strategies, and metabolic adaptations. Here we provide insights on key aspects of their biology, epidemiology, commensal and pathogenic lifestyles, interactions with the immune system, and antifungal resistance.
Collapse
Affiliation(s)
- Myrto Katsipoulaki
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Mark H. T. Stappers
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Dhara Malavia-Jones
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
24
|
Bose S, Sahu SR, Dutta A, Acharya N. A chemically induced attenuated strain of Candida albicans generates robust protective immune responses and prevents systemic candidiasis development. eLife 2024; 13:RP93760. [PMID: 38787374 PMCID: PMC11126311 DOI: 10.7554/elife.93760] [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] [Indexed: 05/25/2024] Open
Abstract
Despite current antifungal therapy, invasive candidiasis causes >40% mortality in immunocompromised individuals. Therefore, developing an antifungal vaccine is a priority. Here, we could for the first time successfully attenuate the virulence of Candida albicans by treating it with a fungistatic dosage of EDTA and demonstrate it to be a potential live whole cell vaccine by using murine models of systemic candidiasis. EDTA inhibited the growth and biofilm formation of C. albicans. RNA-seq analyses of EDTA-treated cells (CAET) revealed that genes mostly involved in metal homeostasis and ribosome biogenesis were up- and down-regulated, respectively. Consequently, a bulky cell wall with elevated levels of mannan and β-glucan, and reduced levels of total monosomes and polysomes were observed. CAET was eliminated faster than the untreated strain (Ca) as found by differential fungal burden in the vital organs of the mice. Higher monocytes, granulocytes, and platelet counts were detected in Ca- vs CAET-challenged mice. While hyper-inflammation and immunosuppression caused the killing of Ca-challenged mice, a critical balance of pro- and anti-inflammatory cytokines-mediated immune responses are the likely reasons for the protective immunity in CAET-infected mice.
Collapse
Affiliation(s)
- Swagata Bose
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| | - Satya Ranjan Sahu
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| | - Abinash Dutta
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| |
Collapse
|
25
|
Hang S, Lu H, Jiang Y. Marine-Derived Metabolites Act as Promising Antifungal Agents. Mar Drugs 2024; 22:180. [PMID: 38667797 PMCID: PMC11051449 DOI: 10.3390/md22040180] [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: 03/20/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
The incidence of invasive fungal diseases (IFDs) is on the rise globally, particularly among immunocompromised patients, leading to significant morbidity and mortality. Current clinical antifungal agents, such as polyenes, azoles, and echinocandins, face increasing resistance from pathogenic fungi. Therefore, there is a pressing need for the development of novel antifungal drugs. Marine-derived secondary metabolites represent valuable resources that are characterized by varied chemical structures and pharmacological activities. While numerous compounds exhibiting promising antifungal activity have been identified, a comprehensive review elucidating their specific underlying mechanisms remains lacking. In this review, we have compiled a summary of antifungal compounds derived from marine organisms, highlighting their diverse mechanisms of action targeting various fungal cellular components, including the cell wall, cell membrane, mitochondria, chromosomes, drug efflux pumps, and several biological processes, including vesicular trafficking and the growth of hyphae and biofilms. This review is helpful for the subsequent development of antifungal drugs due to its summary of the antifungal mechanisms of secondary metabolites from marine organisms.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 200092 Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 200092 Shanghai, China
| |
Collapse
|
26
|
Chen Y, Gao Y, Li Y, Yin J. Anti-Biofilm Activity of Assamsaponin A, Theasaponin E1, and Theasaponin E2 against Candida albicans. Int J Mol Sci 2024; 25:3599. [PMID: 38612411 PMCID: PMC11011434 DOI: 10.3390/ijms25073599] [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: 01/31/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Biofilm formation plays a crucial role in the pathogenesis of Candida albicans and is significantly associated with resistance to antifungal agents. Tea seed saponins, a class of non-ionic triterpenes, have been proven to have fungicidal effects on planktonic C. albicans. However, their anti-biofilm activity and mechanism of action against C. albicans remain unclear. In this study, the effects of three Camellia sinensis seed saponin monomers, namely, theasaponin E1 (TE1), theasaponin E2 (TE2), and assamsaponin A (ASA), on the metabolism, biofilm development, and expression of the virulence genes of C. albicans were evaluated. The results of the XTT reduction assay and crystal violet (CV) staining assay demonstrated that tea seed saponin monomers concentration-dependently suppressed the adhesion and biofilm formation of C. albicans and were able to eradicate mature biofilms. The compounds were in the following order in terms of their inhibitory effects: ASA > TE1 > TE2. The mechanisms were associated with reductions in multiple crucial virulence factors, including cell surface hydrophobicity (CSH), adhesion ability, hyphal morphology conversion, and phospholipase activity. It was further demonstrated through qRT-PCR analysis that the anti-biofilm activity of ASA and TE1 against C. albicans was attributed to the inhibition of RAS1 activation, which consequently suppressed the cAMP-PKA and MAPK signaling pathways. Conversely, TE2 appeared to regulate the morphological turnover and hyphal growth of C. albicans via a pathway that was independent of RAS1. These findings suggest that tea seed saponin monomers are promising innovative agents against C. albicans.
Collapse
Affiliation(s)
- Yuhong Chen
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
- Tea Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Ying Gao
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
| | - Yifan Li
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
| | - Junfeng Yin
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
| |
Collapse
|
27
|
Liang SH, Sircaik S, Dainis J, Kakade P, Penumutchu S, McDonough LD, Chen YH, Frazer C, Schille TB, Allert S, Elshafee O, Hänel M, Mogavero S, Vaishnava S, Cadwell K, Belenky P, Perez JC, Hube B, Ene IV, Bennett RJ. The hyphal-specific toxin candidalysin promotes fungal gut commensalism. Nature 2024; 627:620-627. [PMID: 38448595 PMCID: PMC11230112 DOI: 10.1038/s41586-024-07142-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
The fungus Candida albicans frequently colonizes the human gastrointestinal tract, from which it can disseminate to cause systemic disease. This polymorphic species can transition between growing as single-celled yeast and as multicellular hyphae to adapt to its environment. The current dogma of C. albicans commensalism is that the yeast form is optimal for gut colonization, whereas hyphal cells are detrimental to colonization but critical for virulence1-3. Here, we reveal that this paradigm does not apply to multi-kingdom communities in which a complex interplay between fungal morphology and bacteria dictates C. albicans fitness. Thus, whereas yeast-locked cells outcompete wild-type cells when gut bacteria are absent or depleted by antibiotics, hyphae-competent wild-type cells outcompete yeast-locked cells in hosts with replete bacterial populations. This increased fitness of wild-type cells involves the production of hyphal-specific factors including the toxin candidalysin4,5, which promotes the establishment of colonization. At later time points, adaptive immunity is engaged, and intestinal immunoglobulin A preferentially selects against hyphal cells1,6. Hyphal morphotypes are thus under both positive and negative selective pressures in the gut. Our study further shows that candidalysin has a direct inhibitory effect on bacterial species, including limiting their metabolic output. We therefore propose that C. albicans has evolved hyphal-specific factors, including candidalysin, to better compete with bacterial species in the intestinal niche.
Collapse
Affiliation(s)
- Shen-Huan Liang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Shabnam Sircaik
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Joseph Dainis
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Pallavi Kakade
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Liam D McDonough
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Ying-Han Chen
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Corey Frazer
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Tim B Schille
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute (HKI), Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute (HKI), Jena, Germany
| | - Osama Elshafee
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute (HKI), Jena, Germany
| | - Maria Hänel
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute (HKI), Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute (HKI), Jena, Germany
| | - Shipra Vaishnava
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Ken Cadwell
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - J Christian Perez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute (HKI), Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.
| | - Iuliana V Ene
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
| | - Richard J Bennett
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA.
| |
Collapse
|
28
|
Hefny ZA, Ji B, Elsemman IE, Nielsen J, Van Dijck P. Transcriptomic meta-analysis to identify potential antifungal targets in Candida albicans. BMC Microbiol 2024; 24:66. [PMID: 38413885 PMCID: PMC10898158 DOI: 10.1186/s12866-024-03213-8] [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: 09/29/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Candida albicans is a fungal pathogen causing human infections. Here we investigated differential gene expression patterns and functional enrichment in C. albicans strains grown under different conditions. METHODS A systematic GEO database search identified 239 "Candida albicans" datasets, of which 14 were selected after rigorous criteria application. Retrieval of raw sequencing data from the ENA database was accompanied by essential metadata extraction from dataset descriptions and original articles. Pre-processing via the tailored nf-core pipeline for C. albicans involved alignment, gene/transcript quantification, and diverse quality control measures. Quality assessment via PCA and DESeq2 identified significant genes (FDR < = 0.05, log2-fold change > = 1 or <= -1), while topGO conducted GO term enrichment analysis. Exclusions were made based on data quality and strain relevance, resulting in the selection of seven datasets from the SC5314 strain background for in-depth investigation. RESULTS The meta-analysis of seven selected studies unveiled a substantial number of genes exhibiting significant up-regulation (24,689) and down-regulation (18,074). These differentially expressed genes were further categorized into 2,497 significantly up-regulated and 2,573 significantly down-regulated Gene Ontology (GO) IDs. GO term enrichment analysis clustered these terms into distinct groups, providing insights into the functional implications. Three target gene lists were compiled based on previous studies, focusing on central metabolism, ion homeostasis, and pathogenicity. Frequency analysis revealed genes with higher occurrence within the identified GO clusters, suggesting their potential as antifungal targets. Notably, the genes TPS2, TPS1, RIM21, PRA1, SAP4, and SAP6 exhibited higher frequencies within the clusters. Through frequency analysis within the GO clusters, several key genes emerged as potential targets for antifungal therapies. These include RSP5, GLC7, SOD2, SOD5, SOD1, SOD6, SOD4, SOD3, and RIM101 which exhibited higher occurrence within the identified clusters. CONCLUSION This comprehensive study significantly advances our understanding of the dynamic nature of gene expression in C. albicans. The identification of genes with enhanced potential as antifungal drug targets underpins their value for future interventions. The highlighted genes, including TPS2, TPS1, RIM21, PRA1, SAP4, SAP6, RSP5, GLC7, SOD2, SOD5, SOD1, SOD6, SOD4, SOD3, and RIM101, hold promise for the development of targeted antifungal therapies.
Collapse
Affiliation(s)
- Zeinab Abdelmoghis Hefny
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Kasteelpark Arenberg 31, Leuven, B-3001, Belgium
| | - Boyang Ji
- BioInnovation Institute, Ole Maaløes Vej 3, Copenhagen, DK2200, Denmark
| | - Ibrahim E Elsemman
- Department of Information Systems, Faculty of Computers and Information, Assiut University, Assiut, 2071515, Egypt
| | - Jens Nielsen
- BioInnovation Institute, Ole Maaløes Vej 3, Copenhagen, DK2200, Denmark.
- Department of Life Sciences, Chalmers University of Technology, Kemivägen 10, SE41296, Gothenburg, SE41296, Sweden.
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Kasteelpark Arenberg 31, Leuven, B-3001, Belgium.
| |
Collapse
|
29
|
Feng Z, Lu H, Jiang Y. Promising immunotherapeutic targets for treating candidiasis. Front Cell Infect Microbiol 2024; 14:1339501. [PMID: 38404288 PMCID: PMC10884116 DOI: 10.3389/fcimb.2024.1339501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
In the last twenty years, there has been a significant increase in invasive fungal infections, which has corresponded with the expanding population of individuals with compromised immune systems. As a result, the mortality rate linked to these infections remains unacceptably high. The currently available antifungal drugs, such as azoles, polyenes, and echinocandins, face limitations in terms of their diversity, the escalating resistance of fungi and the occurrence of significant adverse effects. Consequently, there is an urgent need to develop new antifungal medications. Vaccines and antibodies present a promising avenue for addressing fungal infections due to their targeted antifungal properties and ability to modulate the immune response. This review investigates the structure and function of cell wall proteins, secreted proteins, and functional proteins within C. albicans. Furthermore, it seeks to analyze the current advancements and challenges in macromolecular drugs to identify new targets for the effective management of candidiasis.
Collapse
Affiliation(s)
| | - 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
|
Pijuan J, Moreno DF, Yahya G, Moisa M, Ul Haq I, Krukiewicz K, Mosbah R, Metwally K, Cavalu S. Regulatory and pathogenic mechanisms in response to iron deficiency and excess in fungi. Microb Biotechnol 2023; 16:2053-2071. [PMID: 37804207 PMCID: PMC10616654 DOI: 10.1111/1751-7915.14346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023] Open
Abstract
Iron is an essential element for all eukaryote organisms because of its redox properties, which are important for many biological processes such as DNA synthesis, mitochondrial respiration, oxygen transport, lipid, and carbon metabolism. For this reason, living organisms have developed different strategies and mechanisms to optimally regulate iron acquisition, transport, storage, and uptake in different environmental responses. Moreover, iron plays an essential role during microbial infections. Saccharomyces cerevisiae has been of key importance for decrypting iron homeostasis and regulation mechanisms in eukaryotes. Specifically, the transcription factors Aft1/Aft2 and Yap5 regulate the expression of genes to control iron metabolism in response to its deficiency or excess, adapting to the cell's iron requirements and its availability in the environment. We also review which iron-related virulence factors have the most common fungal human pathogens (Aspergillus fumigatus, Cryptococcus neoformans, and Candida albicans). These factors are essential for adaptation in different host niches during pathogenesis, including different fungal-specific iron-uptake mechanisms. While being necessary for virulence, they provide hope for developing novel antifungal treatments, which are currently scarce and usually toxic for patients. In this review, we provide a compilation of the current knowledge about the metabolic response to iron deficiency and excess in fungi.
Collapse
Affiliation(s)
- Jordi Pijuan
- Laboratory of Neurogenetics and Molecular MedicineInstitut de Recerca Sant Joan de DéuBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadridSpain
| | - David F. Moreno
- Department of Molecular Cellular and Developmental BiologyYale UniversityNew HavenConnecticutUSA
- Systems Biology InstituteYale UniversityWest HavenConnecticutUSA
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of PharmacyZagazig UniversityAl SharqiaEgypt
| | - Mihaela Moisa
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| | - Ihtisham Ul Haq
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Programa de Pós‐graduação em Inovação TecnológicaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Centre for Organic and Nanohybrid ElectronicsSilesian University of TechnologyGliwicePoland
| | - Rasha Mosbah
- Infection Control UnitHospitals of Zagazig UniversityZagazigEgypt
| | - Kamel Metwally
- Department of Medicinal Chemistry, Faculty of PharmacyUniversity of TabukTabukSaudi Arabia
- Department of Pharmaceutical Medicinal Chemistry, Faculty of PharmacyZagazig UniversityZagazigEgypt
| | - Simona Cavalu
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| |
Collapse
|
31
|
Schena NC, Baker KM, Stark AA, Thomas DP, Cleary IA. Constitutive ALS3 expression in Candida albicans enhances adhesion and biofilm formation of efg1, but not cph1 mutant strains. PLoS One 2023; 18:e0286547. [PMID: 37440498 PMCID: PMC10343153 DOI: 10.1371/journal.pone.0286547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/17/2023] [Indexed: 07/15/2023] Open
Abstract
Adhesion to living and non-living surfaces is an important virulence trait of the fungal pathogen Candida albicans. Biofilm formation in this organism depends on the expression of a number of cell surface proteins including the hypha-specific protein Als3p. Loss of ALS3 impairs biofilm formation and decreases cell-cell adhesion. We wanted to test whether constitutively expressing ALS3 could compensate for defects in adhesion and biofilm formation observed in mutant strains that lack key transcriptional regulators of biofilm formation Efg1p and Cph1p. We found that ALS3 improved adhesion and biofilm formation in the efg1Δ and efg1Δ cph1Δ mutant strains, but had less effect on the cph1Δ strain.
Collapse
Affiliation(s)
- Nicholas C. Schena
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Kassandra M. Baker
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Anna A. Stark
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Derek P. Thomas
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Ian A. Cleary
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| |
Collapse
|
32
|
de Souza PC, Corrêa AEDN, Gameiro JG, de Oliveira Júnior AG, Panagio LA, Venancio EJ, Almeida RS. Production of IgY against iron permease Ftr1 from Candida albicans and evaluation of its antifungal activity using Galleria mellonella as a model of systemic infection. Microb Pathog 2023:106166. [PMID: 37290729 DOI: 10.1016/j.micpath.2023.106166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/07/2023] [Accepted: 05/19/2023] [Indexed: 06/10/2023]
Abstract
Candida albicans is one of the leading pathological agents of mucosal and deep tissue infections. Considering that the variety of antifungals is restricted and that toxicity limits their use, immunotherapies against pathogenic fungi have been viewed as alternatives with reduced adverse effects. In this context, C. albicans has a protein used to capture iron from the environment and the host, known as the high-affinity iron permease Ftr1. This protein may be a new target of action for novel antifungal therapies, as it influences the virulence of this yeast. Thus, the aim of the present study was to produce and conduct the biological characterization of IgY antibodies against C. albicans Ftr1. Immunization of laying hens with an Ftr1-derived peptide resulted in IgY antibodies extracted from egg yolks capable of binding to the antigen with high affinity (avidity index = 66.6 ± 0.3%). These antibodies reduced the growth and even eliminated C. albicans under iron restriction, a favorable condition for the expression of Ftr1. This also occurred with a mutant strain that does not produce Ftr1 in the presence of iron, a circumstance in which the protein analog of iron permease, Ftr2, is expressed. Furthermore, the survival of G. mellonella larvae infected with C. albicans and treated with the antibodies was 90% higher than the control group, which did not receive treatment (p < 0.0001). Therefore, our data suggest that IgY antibodies against Ftr1 from C. albicans can inhibit yeast propagation by blocking iron uptake.
Collapse
Affiliation(s)
- Patricia Canteri de Souza
- Department of Microbiology, Center of Biological Science, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil
| | - Alana Elke do Nascimento Corrêa
- Department of Microbiology, Center of Biological Science, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil
| | - Juliana Gutschow Gameiro
- Department of Pathology, Clinical and Toxicological Analysis, Center of Health Sciences, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil
| | - Admilton Gonçalves de Oliveira Júnior
- Department of Microbiology, Center of Biological Science, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil
| | - Luciano Aparecido Panagio
- Department of Microbiology, Center of Biological Science, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil
| | - Emerson José Venancio
- Department of Pathological Sciences, Center of Biological Science, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil
| | - Ricardo Sergio Almeida
- Department of Microbiology, Center of Biological Science, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445, Km 380, Londrina, 86.057-970, Paraná, Brazil.
| |
Collapse
|
33
|
Bose S, Singh DV, Adhya TK, Acharya N. Escherichia coli, but Not Staphylococcus aureus, Functions as a Chelating Agent That Exhibits Antifungal Activity against the Pathogenic Yeast Candida albicans. J Fungi (Basel) 2023; 9:jof9030286. [PMID: 36983454 PMCID: PMC10057578 DOI: 10.3390/jof9030286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 02/24/2023] Open
Abstract
Humans are colonized by diverse populations of microbes. Infections by Candida albicans, an opportunistic fungal pathogen, are a result of imbalances in the gut microbial ecosystem and are due to the suppressed immunity of the host. Here, we explored the potential effects of the polymicrobial interactions of C. albicans with Staphylococcus aureus, a Gram-positive bacterium, and Escherichia coli, a Gram-negative bacterium, in dual and triple in vitro culture systems on their respective growth, morphology, and biofilms. We found that S. aureus promoted the fungal growth and hyphal transition of C. albicans through cell-to-cell contacts; contrarily, both the cell and cell-free culture filtrate of E. coli inhibited fungal growth. A yet to be identified secretory metabolite of E. coli functionally mimicked EDTA and EGTA to exhibit antifungal activity. These findings suggested that E. coli, but not S. aureus, functions as a chelating agent and that E. coli plays a dominant role in regulating excessive growth and, potentially, the commensalism of C. albicans. Using animal models of systemic candidiasis, we found that the E. coli cell-free filtrate suppressed the virulence of C. albicans. In general, this study unraveled a significant antimicrobial activity and a potential role in the nutritional immunity of E. coli, and further determining the underlying processes behind the E. coli–C. albicans interaction could provide critical information in understanding the pathogenicity of C. albicans.
Collapse
Affiliation(s)
- Swagata Bose
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar 751023, India
- KIIT School of Biotechnology, Bhubaneswar 751021, India
| | - Durg Vijai Singh
- Department of Biotechnology, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya 824236, India
| | | | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar 751023, India
- Correspondence: ; Tel.: +91-674-230-4278; Fax: +91-674-230-0728
| |
Collapse
|
34
|
Candidalysin Is the Hemolytic Factor of Candida albicans. Toxins (Basel) 2022; 14:toxins14120874. [PMID: 36548771 PMCID: PMC9785678 DOI: 10.3390/toxins14120874] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Candida albicans produces an important virulence factor, the hypha-associated Ece1-derived secreted peptide toxin candidalysin, which is crucial for the establishment of mucosal and systemic infections. C. albicans has also long been known to be hemolytic, yet the hemolytic factor has not been clearly identified. Here, we show that candidalysin is the hemolytic factor of C. albicans. Its hemolytic activity is modulated by fragments of another Ece1 peptide, P7. Hemolysis by candidalysin can be neutralized by the purinergic receptor antagonist pyridoxal-phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS). PPADS also affects candidalysin's ability to intercalate into synthetic membranes. We also describe the neutralization potential of two anti-candidalysin nanobodies, which are promising candidates for future anti-Candida therapy. This work provides evidence that the historically proposed hemolytic factor of C. albicans is in fact candidalysin and sheds more light on the complex roles of this toxin in C. albicans biology and pathogenicity.
Collapse
|
35
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
36
|
Sen S, Bal SK, Yadav S, Mishra P, G VV, Rastogi R, Mukhopadhyay CK. Intracellular pathogen Leishmania intervenes in iron loading into ferritin by cleaving chaperones in host macrophages as an iron acquisition strategy. J Biol Chem 2022; 298:102646. [PMID: 36309090 PMCID: PMC9700016 DOI: 10.1016/j.jbc.2022.102646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Iron (Fe) sequestration is one of the most important strategies of the host to control the growth and survival of invading pathogens. Ferritin (Ft) plays a pivotal role in the sequestration mechanism of mammalian hosts by storing Fe. Recent evidence suggests that poly(rC)-binding proteins (PCBPs) act as chaperones for loading Fe into Ft. Incidentally, modulation of host PCBPs in respect to storing Fe in Ft during any infection remains unexplored. Among PCBPs, PCBP1 and PCBP2 are present in every cell type and involved in interacting with Ft for Fe loading. Leishmania donovani (LD) resides within macrophages during the mammalian stage of infection, causing life-threatening visceral leishmaniasis. Here, we reveal the ability of LD to cleave PCBP1 and PCBP2 in host monocytes/macrophages. LD cleaves PCBP1-FLAG into two fragments and PCBP2-FLAG into multiple fragments, thus affecting their interactions with Ft and resulting in decreased Fe loading into Ft. LD-derived culture supernatant or exosome-enriched fractions are also able to cleave PCBPs, suggesting involvement of a secreted protease of the parasite. Using an immune-depletion experiment and transgenic mutants, we confirmed the involvement of zinc-metalloprotease GP63 in cleaving PCBPs. We further revealed that by cleaving host PCBPs, Leishmania could inhibit Fe loading into Ft to accumulate available Fe for higher intracellular growth. This is the first report of a novel strategy adopted by a mammalian pathogen to interfere with Fe sequestration into Ft by cleaving chaperones for its survival advantage within the host.
Collapse
|
37
|
Pokhrel S, Boonmee N, Tulyaprawat O, Pharkjaksu S, Thaipisutikul I, Chairatana P, Ngamskulrungroj P, Mitrpant C. Assessment of Biofilm Formation by Candida albicans Strains Isolated from Hemocultures and Their Role in Pathogenesis in the Zebrafish Model. J Fungi (Basel) 2022; 8:jof8101014. [PMID: 36294579 PMCID: PMC9605499 DOI: 10.3390/jof8101014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/04/2022] Open
Abstract
Candida albicans, an opportunistic pathogen, has the ability to form biofilms in the host or within medical devices in the body. Biofilms have been associated with disseminated/invasive disease with increased severity of infection by disrupting the host immune response and prolonging antifungal treatment. In this study, the in vivo virulence of three strains with different biofilm formation strengths, that is, non-, weak-, and strong biofilm formers, was evaluated using the zebrafish model. The survival assay and fungal tissue burden were measured. Biofilm-related gene expressions were also investigated. The survival of zebrafish, inoculated with strong biofilms forming C. albicans,, was significantly shorter than strains without biofilms forming C. albicans. However, there were no statistical differences in the burden of viable colonogenic cell number between the groups of the three strains tested. We observed that the stronger the biofilm formation, the higher up-regulation of biofilm-associated genes. The biofilm-forming strain (140 and 57), injected into zebrafish larvae, possessed a higher level of expression of genes associated with adhesion, attachment, filamentation, and cell proliferation, including eap1, als3, hwp1, bcr1, and mkc1 at 8 h. The results suggested that, despite the difference in genetic background, biofilm formation is an important virulence factor for the pathogenesis of C. albicans. However, the association between biofilm formation strength and in vivo virulence is controversial and needs to be further studied.
Collapse
Affiliation(s)
- Sabi Pokhrel
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nawarat Boonmee
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Orawan Tulyaprawat
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sujiraphong Pharkjaksu
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Iyarit Thaipisutikul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Phoom Chairatana
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Popchai Ngamskulrungroj
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chalermchai Mitrpant
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Correspondence:
| |
Collapse
|
38
|
Arvizu-Rubio VJ, García-Carnero LC, Mora-Montes HM. Moonlighting proteins in medically relevant fungi. PeerJ 2022; 10:e14001. [PMID: 36117533 PMCID: PMC9480056 DOI: 10.7717/peerj.14001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/13/2022] [Indexed: 01/19/2023] Open
Abstract
Moonlighting proteins represent an intriguing area of cell biology, due to their ability to perform two or more unrelated functions in one or many cellular compartments. These proteins have been described in all kingdoms of life and are usually constitutively expressed and conserved proteins with housekeeping functions. Although widely studied in pathogenic bacteria, the information about these proteins in pathogenic fungi is scarce, but there are some reports of their functions in the etiological agents of the main human mycoses, such as Candida spp., Paracoccidioides brasiliensis, Histoplasma capsulatum, Aspergillus fumigatus, Cryptococcus neoformans, and Sporothrix schenckii. In these fungi, most of the described moonlighting proteins are metabolic enzymes, such as enolase and glyceraldehyde-3-phosphate dehydrogenase; chaperones, transcription factors, and redox response proteins, such as peroxiredoxin and catalase, which moonlight at the cell surface and perform virulence-related processes, contributing to immune evasion, adhesions, invasion, and dissemination to host cells and tissues. All moonlighting proteins and their functions described in this review highlight the limited information about this biological aspect in pathogenic fungi, representing this a relevant opportunity area that will contribute to expanding our current knowledge of these organisms' pathogenesis.
Collapse
|
39
|
Song M, Thak EJ, Kang HA, Kronstad JW, Jung WH. Cryptococcus neoformans can utilize ferritin as an iron source. Med Mycol 2022; 60:myac056. [PMID: 35943215 PMCID: PMC9387142 DOI: 10.1093/mmy/myac056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/11/2022] [Accepted: 08/06/2022] [Indexed: 01/21/2023] Open
Abstract
UNLABELLED Ferritin, a major iron storage protein in vertebrates, supplies iron upon iron deficiency. Ferritin is also found extracellularly, and acts as an iron carrier and a contributor to the immune response to invading microbes. Some microbial pathogens take advantage of ferritin as an iron source upon infection. However, no information is currently available on whether the human fungal pathogen Cryptococcus neoformans can acquire iron from ferritin. Here, we found that C. neoformans grew well in the presence of ferritin as a sole iron source. We showed that the binding of ferritin to the surface of C. neoformans is necessary and that acidification may contribute to ferritin-iron utilization by the fungus. Our data also revealed that the high-affinity reductive iron uptake system in C. neoformans is required for ferritin-iron acquisition. Furthermore, phagocytosis of C. neoformans by macrophages led to increased intracellular ferritin levels, suggesting that iron is sequestered by ferritin in infected macrophages. The increase in intracellular ferritin levels was reversed upon infection with a C. neoformans mutant deficient in the high-affinity reductive iron uptake system, indicating that this system plays a major role in iron acquisition in the phagocytosed C. neoformans in macrophages. LAY SUMMARY Cryptococcus neoformans is an opportunistic fungal pathogen causing life-threatening pulmonary disease and cryptococcal meningitis, mainly in immunocompromised patients. In this study, we found that C. neoformans can use ferritin, a major iron storage protein in vertebrates, as a sole iron source.
Collapse
Affiliation(s)
- Moonyong Song
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| | - Eun Jung Thak
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Hyun Ah Kang
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - James W Kronstad
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Won Hee Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| |
Collapse
|
40
|
Role of Iron and Iron Overload in the Pathogenesis of Invasive Fungal Infections in Patients with Hematological Malignancies. J Clin Med 2022; 11:jcm11154457. [PMID: 35956074 PMCID: PMC9369168 DOI: 10.3390/jcm11154457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Iron is an essential trace metal necessary for the reproduction and survival of fungal pathogens. The latter have developed various mechanisms to acquire iron from their mammalian hosts, with whom they participate in a continuous struggle for dominance over iron. Invasive fungal infections are an important problem in the treatment of patients with hematological malignancies, and they are associated with significant morbidity and mortality. The diagnosis of invasive clinical infections in these patients is complex, and the treatment, which must occur as early as possible, is difficult. There are several studies that have shown a possible link between iron overload and an increased susceptibility to infections. This link is also relevant for patients with hematological malignancies and for those treated with allogeneic hematopoietic stem cell transplantation. The role of iron and its metabolism in the virulence and pathogenesis of various invasive fungal infections is intriguing, and so far, there is some evidence linking invasive fungal infections to iron or iron overload. Clarifying the possible association of iron and iron overload with susceptibility to invasive fungal infections could be important for a better prevention and treatment of these infections in patients with hematological malignancies.
Collapse
|
41
|
Gonçalves SM, Ferreira AV, Cunha C, Carvalho A. Targeting immunometabolism in host-directed therapies to fungal disease. Clin Exp Immunol 2022; 208:158-166. [PMID: 35641161 PMCID: PMC9188340 DOI: 10.1093/cei/uxab014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/03/2021] [Accepted: 10/29/2021] [Indexed: 01/21/2024] Open
Abstract
Fungal infections affect over a billion people and are responsible for more than 1.5 million deaths each year. Despite progress in diagnostic and therapeutic approaches, the management of severe fungal infections remains a challenge. Recently, the reprogramming of cellular metabolism has emerged as a central mechanism through which the effector functions of immune cells are supported to promote antifungal activity. An improved understanding of the immunometabolic signatures that orchestrate antifungal immunity, together with the dissection of the mechanisms that underlie heterogeneity in individual immune responses, may therefore unveil new targets amenable to adjunctive host-directed therapies. In this review, we highlight recent advances in the metabolic regulation of host-fungus interactions and antifungal immune responses, and outline targetable pathways and mechanisms with promising therapeutic potential.
Collapse
Affiliation(s)
- Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Anaísa V Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| |
Collapse
|
42
|
Ramírez-Zavala B, Krüger I, Dunker C, Jacobsen ID, Morschhäuser J. The protein kinase Ire1 has a Hac1-independent essential role in iron uptake and virulence of Candida albicans. PLoS Pathog 2022; 18:e1010283. [PMID: 35108336 PMCID: PMC8846550 DOI: 10.1371/journal.ppat.1010283] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/14/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022] Open
Abstract
Protein kinases play central roles in virtually all signaling pathways that enable organisms to adapt to their environment. Microbial pathogens must cope with severely restricted iron availability in mammalian hosts to invade and establish themselves within infected tissues. To uncover protein kinase signaling pathways that are involved in the adaptation of the pathogenic yeast Candida albicans to iron limitation, we generated a comprehensive protein kinase deletion mutant library of a wild-type strain. Screening of this library revealed that the protein kinase Ire1, which has a conserved role in the response of eukaryotic cells to endoplasmic reticulum stress, is essential for growth of C. albicans under iron-limiting conditions. Ire1 was not necessary for the activity of the transcription factor Sef1, which regulates the response of the fungus to iron limitation, and Sef1 target genes that are induced by iron depletion were normally upregulated in ire1Δ mutants. Instead, Ire1 was required for proper localization of the high-affinity iron permease Ftr1 to the cell membrane. Intriguingly, iron limitation did not cause increased endoplasmic reticulum stress, and the transcription factor Hac1, which is activated by Ire1-mediated removal of the non-canonical intron in the HAC1 mRNA, was dispensable for Ftr1 localization to the cell membrane and growth under iron-limiting conditions. Nevertheless, expression of a pre-spliced HAC1 copy in ire1Δ mutants restored Ftr1 localization and rescued the growth defects of the mutants. Both ire1Δ and hac1Δ mutants were avirulent in a mouse model of systemic candidiasis, indicating that an appropriate response to endoplasmic reticulum stress is important for the virulence of C. albicans. However, the specific requirement of Ire1 for the functionality of the high-affinity iron permease Ftr1, a well-established virulence factor, even in the absence of endoplasmic reticulum stress uncovers a novel Hac1-independent essential role of Ire1 in iron acquisition and virulence of C. albicans.
Collapse
Affiliation(s)
| | - Ines Krüger
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Christine Dunker
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Ilse D. Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Joachim Morschhäuser
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
43
|
Shichiri-Negoro Y, Tsutsumi-Arai C, Arai Y, Satomura K, Arakawa S, Wakabayashi N. Ozone ultrafine bubble water inhibits the early formation of Candida albicans biofilms. PLoS One 2021; 16:e0261180. [PMID: 34890423 PMCID: PMC8664219 DOI: 10.1371/journal.pone.0261180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/24/2021] [Indexed: 11/19/2022] Open
Abstract
This study aimed to investigate the effect of ozone ultrafine bubble water (OUFBW) on the formation and growth of Candida albicans (C. albicans) biofilms and surface properties of denture base resins. OUFBWs were prepared under concentrations of 6 (OUFBW6), 9 (OUFBW9), and 11 ppm (OUFBW11). Phosphate buffered saline and ozone-free electrolyte aqueous solutions (OFEAS) were used as controls. Acrylic resin discs were made according to manufacturer instructions, and C. albicans was initially cultured on the discs for 1.5 h. A colony forming unit (CFU) assay was performed by soaking the discs in OUFBW for 5 min after forming a 24-h C. albicans biofilm. The discs after initial attachment for 1.5 h were immersed in OUFBW and then cultured for 0, 3, and 5 h. CFUs were subsequently evaluated at each time point. Moreover, a viability assay, scanning electron microscopy (SEM), Alamar Blue assay, and quantitative real-time polymerase chain reaction (qRT-PCR) test were performed. To investigate the long-term effects of OUFBW on acrylic resin surface properties, Vickers hardness (VH) and surface roughness (Ra) were measured. We found that OUFBW9 and OUFBW11 significantly degraded the formed 24-h biofilm. The time point CFU assay showed that C. albicans biofilm formation was significantly inhibited due to OUFBW11 exposure. Interestingly, fluorescence microscopy revealed that almost living cells were observed in all groups. In SEM images, the OUFBW group had lesser number of fungi and the amount of non-three-dimensional biofilm than the control group. In the Alamar Blue assay, OUFBW11 was found to suppress Candida metabolic function. The qRT-PCR test showed that OUFBW down-regulated ALS1 and ALS3 expression regarding cell-cell, cell-material adhesion, and biofilm formation. Additionally, VH and Ra were not significantly different between the two groups. Overall, our data suggest that OUFBW suppressed C. albicans growth and biofilm formation on polymethyl methacrylate without impairing surface properties.
Collapse
Affiliation(s)
- Yuka Shichiri-Negoro
- Department of Removable Partial Prosthodontics, Graduate School, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chiaki Tsutsumi-Arai
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa, Japan
| | - Yuki Arai
- Department of Removable Partial Prosthodontics, Graduate School, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazuhito Satomura
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa, Japan
| | - Shinichi Arakawa
- Department of Lifetime Oral Health Care Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Noriyuki Wakabayashi
- Department of Removable Partial Prosthodontics, Graduate School, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
44
|
A. L. Bataineh MT, Soares NC, Semreen MH, Cacciatore S, Dash NR, Hamad M, Mousa MK, Salam JSA, Al Gharaibeh MF, Zerbini LF, Hamad M. Candida albicans PPG1, a serine/threonine phosphatase, plays a vital role in central carbon metabolisms under filament-inducing conditions: A multi-omics approach. PLoS One 2021; 16:e0259588. [PMID: 34874940 PMCID: PMC8651141 DOI: 10.1371/journal.pone.0259588] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/21/2021] [Indexed: 01/01/2023] Open
Abstract
Candida albicans is the leading cause of life-threatening bloodstream candidiasis, especially among immunocompromised patients. The reversible morphological transition from yeast to hyphal filaments in response to host environmental cues facilitates C. albicans tissue invasion, immune evasion, and dissemination. Hence, it is widely considered that filamentation represents one of the major virulence properties in C. albicans. We have previously characterized Ppg1, a PP2A-type protein phosphatase that controls filament extension and virulence in C. albicans. This study conducted RNA sequencing analysis of samples obtained from C. albicans wild type and ppg1Δ/Δ strains grown under filament-inducing conditions. Overall, ppg1Δ/Δ strain showed 1448 upregulated and 710 downregulated genes, representing approximately one-third of the entire annotated C. albicans genome. Transcriptomic analysis identified significant downregulation of well-characterized genes linked to filamentation and virulence, such as ALS3, HWP1, ECE1, and RBT1. Expression analysis showed that essential genes involved in C. albicans central carbon metabolisms, including GDH3, GPD1, GPD2, RHR2, INO1, AAH1, and MET14 were among the top upregulated genes. Subsequent metabolomics analysis of C. albicans ppg1Δ/Δ strain revealed a negative enrichment of metabolites with carboxylic acid substituents and a positive enrichment of metabolites with pyranose substituents. Altogether, Ppg1 in vitro analysis revealed a link between metabolites substituents and filament formation controlled by a phosphatase to regulate morphogenesis and virulence.
Collapse
Affiliation(s)
- Mohammad Tahseen A. L. Bataineh
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
- Department of Genetics and Molecular Biology, College Of Medicine And Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Nelson Cruz Soares
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Mohammad Harb Semreen
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Stefano Cacciatore
- Cancer Genomics Group, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
- Institute for Reproductive and Developmental Biology, Imperial College, London, United Kingdom
| | | | - Mohamad Hamad
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, UAE
| | - Muath Khairi Mousa
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | | | | | - Luiz F. Zerbini
- Cancer Genomics Group, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Mawieh Hamad
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, UAE
| |
Collapse
|
45
|
Wang ZL, Pan HB, Li MY, Wu W, Yu XP. Comprehensive insights into host-pathogen interaction between brown planthopper and a fungal entomopathogen by dual RNA sequencing. PEST MANAGEMENT SCIENCE 2021; 77:4903-4914. [PMID: 34171165 DOI: 10.1002/ps.6529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/20/2021] [Accepted: 06/25/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The brown planthopper (BPH) is one of the most destructive pests of rice, causing tremendous yield and economic losses every year. The fungal entomopathogen Metarhizium anisopliae was previously proved to have great potential for BPH biocontrol. Genome-wide insight into the insect-fungus interaction is crucial for genetic improvement of M. anisopliae to enhance its virulence to BPH but still has been poorly explored. RESULTS Using dual RNA-seq approach, we present here a global view of host and fungal gene expressions in BPH adults during the fungal infection. The results revealed that BPH could initiate strong defense responses against the fungal attack by upregulating the expressions of a large number of genes, including genes involved in cuticle formation, immune response, cell detoxification and biomacromolecule metabolism. Correspondingly, the fungal entomopathogen could induce a series of genes to infect and modulate BPH, including genes involved in fungal penetration, invasive growth, stress resistance and virulence. Three host defense-related genes (NlPCE4, NlPOD1 and NlCYP4DE1) were chosen for further function analysis. RNAi-mediated knockdown of NlPCE4 caused a significant decrease in BPH survival, but no obvious effects on the survival rates were detected by the suppression of NlPOD1 and NlCYP4DE1. Combination of dsRNA injection and fungal infection could significantly enhance the BPH-killing speed, as synergistic mortalities were observed in co-treatments of RNAi and M. anisopliae infection. CONCLUSION Our study provides a comprehensive insight into molecular mechanisms of host-pathogen interaction between BPH and M. anisopliae and contributes to future development of new efficient biocontrol strategies for BPH biocontrol.
Collapse
Affiliation(s)
- Zheng-Liang Wang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Sciences, China Jiliang University, Zhejiang, China
| | - Hai-Bo Pan
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Sciences, China Jiliang University, Zhejiang, China
| | - Mu-Yu Li
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Sciences, China Jiliang University, Zhejiang, China
| | - Wei Wu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Sciences, China Jiliang University, Zhejiang, China
| | - Xiao-Ping Yu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Sciences, China Jiliang University, Zhejiang, China
| |
Collapse
|
46
|
Ibe C, Munro CA. Fungal Cell Wall Proteins and Signaling Pathways Form a Cytoprotective Network to Combat Stresses. J Fungi (Basel) 2021; 7:jof7090739. [PMID: 34575777 PMCID: PMC8466366 DOI: 10.3390/jof7090739] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/13/2022] Open
Abstract
Candida species are part of the normal flora of humans, but once the immune system of the host is impaired and they escape from commensal niches, they shift from commensal to pathogen causing candidiasis. Candida albicans remains the primary cause of candidiasis, accounting for about 60% of the global candidiasis burden. The cell wall of C. albicans and related fungal pathogens forms the interface with the host, gives fungal cells their shape, and also provides protection against stresses. The cell wall is a dynamic organelle with great adaptive flexibility that allows remodeling, morphogenesis, and changes in its components in response to the environment. It is mainly composed of the inner polysaccharide rich layer (chitin, and β-glucan) and the outer protein coat (mannoproteins). The highly glycosylated protein coat mediates interactions between C. albicans cells and their environment, including reprograming of wall architecture in response to several conditions, such as carbon source, pH, high temperature, and morphogenesis. The mannoproteins are also associated with C. albicans adherence, drug resistance, and virulence. Vitally, the mannoproteins contribute to cell wall construction and especially cell wall remodeling when cells encounter physical and chemical stresses. This review describes the interconnected cell wall integrity (CWI) and stress-activated pathways (e.g., Hog1, Cek1, and Mkc1 mediated pathways) that regulates cell wall remodeling and the expression of some of the mannoproteins in C. albicans and other species. The mannoproteins of the surface coat is of great importance to pathogen survival, growth, and virulence, thus understanding their structure and function as well as regulatory mechanisms can pave the way for better management of candidiasis.
Collapse
Affiliation(s)
- Chibuike Ibe
- Department of Microbiology, Faculty of Biological Sciences, Abia State University, Uturu 441107, Nigeria
- Correspondence:
| | - Carol A. Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB24 3FX, UK;
| |
Collapse
|
47
|
CO 2 enhances the formation, nutrient scavenging and drug resistance properties of C. albicans biofilms. NPJ Biofilms Microbiomes 2021; 7:67. [PMID: 34385462 PMCID: PMC8361082 DOI: 10.1038/s41522-021-00238-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
C. albicans is the predominant human fungal pathogen and frequently colonises medical devices, such as voice prostheses, as a biofilm. It is a dimorphic yeast that can switch between yeast and hyphal forms in response to environmental cues, a property that is essential during biofilm establishment and maturation. One such cue is the elevation of CO2 levels, as observed in exhaled breath for example. However, despite the clear medical relevance, the effect of CO2 on C. albicans biofilm growth has not been investigated to date. Here we show that physiologically relevant CO2 elevation enhances each stage of the C. albicans biofilm-forming process: from attachment through maturation to dispersion. The effects of CO2 are mediated via the Ras/cAMP/PKA signalling pathway and the central biofilm regulators Efg1, Brg1, Bcr1 and Ndt80. Biofilms grown under elevated CO2 conditions also exhibit increased azole resistance, increased Sef1-dependent iron scavenging and enhanced glucose uptake to support their rapid growth. These findings suggest that C. albicans has evolved to utilise the CO2 signal to promote biofilm formation within the host. We investigate the possibility of targeting CO2-activated processes and propose 2-deoxyglucose as a drug that may be repurposed to prevent C. albicans biofilm formation on medical airway management implants. We thus characterise the mechanisms by which CO2 promotes C. albicans biofilm formation and suggest new approaches for future preventative strategies.
Collapse
|
48
|
Hameed S, Hans S, Singh S, Dhiman R, Monasky R, Pandey RP, Thangamani S, Fatima Z. Revisiting the Vital Drivers and Mechanisms of β-Glucan Masking in Human Fungal Pathogen, Candida albicans. Pathogens 2021; 10:942. [PMID: 34451406 PMCID: PMC8399646 DOI: 10.3390/pathogens10080942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/14/2021] [Accepted: 07/22/2021] [Indexed: 12/31/2022] Open
Abstract
Among the several human fungal pathogens, Candida genus represents one of the most implicated in the clinical scenario. There exist several distinctive features that govern the establishment of Candida infections in addition to their capacity to adapt to multiple stress conditions inside humans which also include evasion of host immune responses. The complex fungal cell wall of the prevalent pathogen, Candida albicans, is one of the main targets of antifungal drugs and recognized by host immune cells. The wall consists of tiered arrangement of an outer thin but dense covering of mannan and inner buried layers of β-glucan and chitin. However, the pathogenic fungi adopt strategies to evade immune recognition by masking these molecules. This capacity to camouflage the immunogenic polysaccharide β-glucan from the host is a key virulence factor of C. albicans. The present review is an attempt to collate various underlying factors and mechanisms involved in Candida β-glucan masking from the available pool of knowledge and provide a comprehensive understanding. This will further improve therapeutic approaches to candidiasis by identifying new antifungal targets that blocks fungal immune evasion.
Collapse
Affiliation(s)
- Saif Hameed
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram 122413, India; (S.H.); (S.H.); (S.S.)
| | - Sandeep Hans
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram 122413, India; (S.H.); (S.H.); (S.S.)
| | - Shweta Singh
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram 122413, India; (S.H.); (S.H.); (S.S.)
| | - Ruby Dhiman
- Centre for Drug Design Discovery and Development (C4D), SRM University, Sonepat 131029, India; (R.D.); (R.P.P.)
| | - Ross Monasky
- Department of Pathology and Population Medicine, College of Veterinary Medicine, Midwestern University, 19555 N. 59th Ave., Glendale, AZ 85308, USA; (R.M.); (S.T.)
| | - Ramendra Pati Pandey
- Centre for Drug Design Discovery and Development (C4D), SRM University, Sonepat 131029, India; (R.D.); (R.P.P.)
| | - Shankar Thangamani
- Department of Pathology and Population Medicine, College of Veterinary Medicine, Midwestern University, 19555 N. 59th Ave., Glendale, AZ 85308, USA; (R.M.); (S.T.)
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Zeeshan Fatima
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram 122413, India; (S.H.); (S.H.); (S.S.)
| |
Collapse
|
49
|
Liu C, Xu C, Du Y, Liu J, Ning Y. Role of agglutinin-like sequence protein 3 (Als3) in the structure and antifungal resistance of Candida albicans biofilms. FEMS Microbiol Lett 2021; 368:6316779. [PMID: 34232317 DOI: 10.1093/femsle/fnab089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Agglutinin-like sequence protein 3 (Als3) is a cell surface glycoprotein of Candida albicans that plays essential roles in the processes of adherence and biofilm formation in vitro. In this study, we focused on the contribution of Als3 to the structure and drug susceptibility of biofilms. The C. albicans wild-type (WT) strain DAY185, the als3Δ/Δ null strain and the als3Δ/Δ + pALS3 complemented strain were used. Colony-forming unit enumeration, crystal violet and cell surface hydrophobicity assays, scanning electron microscopy and confocal laser scanning microscopy coupled with analyses using COMSTAT software were performed to evaluate the biomass and architecture of the biofilms. The detailed architectural analysis showed a significant variation in the biofilm parameters of the als3Δ/Δ biofilms compared with those of the WT biofilms. Fluconazole, miconazole and amphotericin B were selected as the antifungal agents for the antimycotic susceptibility test, and increased susceptibility was found with the ALS3 deletion biofilms. A quantitative real-time polymerase chain reaction analysis showed downregulation of biofilm formation-related genes (ALS1, EFG1, HWP1 and CSH1) and drug resistance-related genes (ERG11, CDR1, CDR2 and MDR1) in the als3Δ/Δ biofilms. We concluded that Als3 contributes to biofilm formation by changing the biofilm architecture and is involved in the antifungal resistance of C. albicans biofilms.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Cheng Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Yu Du
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Jia Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Yang Ning
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| |
Collapse
|
50
|
Swetha TK, Subramenium GA, Kasthuri T, Sharumathi R, Pandian SK. 5-hydroxymethyl-2-furaldehyde impairs Candida albicans - Staphylococcus epidermidis interaction in co-culture by suppressing crucial supportive virulence traits. Microb Pathog 2021; 158:104990. [PMID: 34048889 DOI: 10.1016/j.micpath.2021.104990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 04/25/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Polymicrobial biofilms involving fungal-bacterial interactions are stated to modulate host immune response and exhibit enhanced antibiotic resistance. In this milieu, clinically important opportunistic pathogens Candida albicans and Staphylococcus epidermidis associate synergistically and instigate implant and blood stream infections. Impediment of virulence traits that support successive pathogenic lifestyle and inter-kingdom interactions without altering the microbial growth represents an attractive alternate strategy. To accomplish this objective, 5-hydroxymethyl-2-furaldehyde (5HM2F), a reported antibiofilm agent against C. albicans, was considered for this study. 5HM2F significantly repressed the biofilm formation of S. epidermidis and mixed-species at 300 μg/mL and 400 μg/mL, respectively without modulating the growth. Microscopic analyses and phenotypic assays explicated the competency of 5HM2F to impede biofilm formation, hyphal growth, initial attachment, intercellular adhesion, and fungal-bacterial interaction. Further, 5HM2F greatly reduced the secreted hydrolases production. Reduced content of biofilm matrix components upon 5HM2F treatment was believed to be the underlying reason for enhanced antibiotic and/antifungal susceptibility. Additionally, qPCR analysis correlated well with in vitro bioassays wherein, 5HM2F was identified to repress the expression of important genes associated with hyphal morphogenesis, adhesion, biofilm formation and virulence in both mono-species and mixed-species. Reduced virulence and colonization of mono-species and mixed-species in 5HM2F treated Caenorhabditis elegans substantiated the antibiofilm and antivirulence potential of 5HM2F. Overall, this study proposes 5HM2F as a potent therapeutic candidate against single and mixed-species biofilm infections of C. albicans and S. epidermidis.
Collapse
Affiliation(s)
| | | | - Thirupathi Kasthuri
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630 003, Tamil Nadu, India
| | - Rajendran Sharumathi
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630 003, Tamil Nadu, India
| | - Shunmugiah Karutha Pandian
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630 003, Tamil Nadu, India.
| |
Collapse
|