1
|
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
|
2
|
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
|
3
|
Liu S, Lin K, Tian Y, He Y, Zhang Z, Li C, Luo Y, Liang Y, Ma Y, Deng W. Candidalysin induces Tim-3 expression in macrophages to suppress antifungal immunity in oropharyngeal candidiasis. Int Immunopharmacol 2025; 156:114672. [PMID: 40252466 DOI: 10.1016/j.intimp.2025.114672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
Oropharyngeal candidiasis significantly impacts patients' quality of life. Although the immune mechanisms underlying OPC have been explored, the roles of immune checkpoints and macrophages in this context remain unclear. We employed single-cell techniques to analyze changes in the oral immune microenvironment and explored the role of Tim-3 in OPC. Our single-cell analysis identified distinct macrophage subpopulations in OPC tissue, each exhibiting heterogeneous antifungal immune responses. Further investigation revealed that candidalysin induces Tim-3, which primarily reduces the proportion of macrophage subpopulations 1 and 2 in OPCs. Additionally, it suppresses the inflammatory response during infection. Experimental research showed that Tim-3 inhibited the P65 inflammatory signaling pathway in macrophages, thereby reducing the secretion of inflammatory cytokines such as l1b, Il6, Tnf, Il23, and chemokines including Ccl2, Cxcl1, Cxcl2, Cxcl3, and Cxcl5. Additionally, Tim-3 promoted macrophage apoptosis, potentially contributing to the decreased proportion of macrophages. We also found that Tim-3 suppressed the phagocytic and killing functions of macrophages against C.albicans. Notably, our research revealed that Tim-3's ligand, Galectin-9, also exhibits functional similarities to Tim-3. Specifically, the Galectin-9 gene, Lgals9, is also induced by candidalysin. This study identifies candidalysin in inhibiting macrophage responses to C.albicans through the induction of Galectin-9/Tim-3 in oral mucosal immunity, potentially representing a novel mechanism of immune evasion by C.albicans in OPC. Therefore, our research provides a basis for considering this axis as a potential therapeutic target.
Collapse
Affiliation(s)
- Siqi Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Kefan Lin
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yuyang Tian
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yuchen He
- Department of Clinical Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhiwen Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Chunyan Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yurong Luo
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yunsheng Liang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China.
| | - Yubo Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Weiwei Deng
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
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
|
5
|
Sharma A, Homayoon A, Weyler M, Frazer C, Ramírez-Zavala B, Morschhäuser J, Bennett RJ. Transcriptional control of C. albicans white-opaque switching and modulation by environmental cues and strain background. mBio 2025:e0058125. [PMID: 40202334 DOI: 10.1128/mbio.00581-25] [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: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
The opportunistic fungal pathogen Candida albicans can undergo cellular transitions in response to environmental cues that impact its lifestyle and its interactions with the human host. This is exemplified by the white-opaque switch, which is a heritable transition between two phenotypic states that is regulated by a highly interconnected network of transcription factors (TFs). To obtain greater understanding of the transcriptional regulation of the switch, we generated a genome-wide, tetracycline-inducible TF library in the WO-1 strain background and identified those TFs whose forced expression induces white cells to switch to the opaque state. This set of opaque-inducing TFs was also evaluated for their ability to induce switching in a second strain background, that of the standard reference strain SC5314, as well as during growth on different laboratory media. These experiments identify 14 TFs that can drive white-to-opaque switching when overexpressed but that do so in a highly strain- and media-specific manner. In particular, changes in pH, amino acids, and zinc concentrations had marked effects on the ability of TFs to drive phenotypic switching. These results provide insights into the complex transcriptional regulation of switching in C. albicans and reveal that an interplay between genetic and environmental factors determines TF function and cell fate.IMPORTANCEThe white-opaque switch in Candida albicans represents a model system for understanding an epigenetic switch in a eukaryotic pathogen. Here, we generated an inducible library of the set of transcription factors (TFs) present in C. albicans and identify 14 TFs that can drive the white-to-opaque transition when ectopically expressed. We demonstrate that several of these TFs induce the switch in a highly strain- and media-specific manner. This highlights that both strain background and changes in experimental conditions (including different water sources) can profoundly impact the phenotypic consequences of TF overexpression. Moreover, the inducible TF library provides an invaluable tool for the further analysis of TF function in this important human pathogen.
Collapse
Affiliation(s)
- Anupam Sharma
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Ameen Homayoon
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Michael Weyler
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Corey Frazer
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Bernardo Ramírez-Zavala
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Joachim Morschhäuser
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Richard J Bennett
- Department of Molecular and Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
6
|
Wu Q, Cen F, Xie Y, Ning X, Wang J, Lin Z, Huang J. Nanoparticle-based antifungal therapies innovations mechanisms and future prospects. PeerJ 2025; 13:e19199. [PMID: 40226540 PMCID: PMC11988106 DOI: 10.7717/peerj.19199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Fungal infections present an increasing global health challenge, with a substantial annual mortality rate of 1.6 million deaths each year in certain situations. The emergence of antifungal resistance has further complicated treatment strategies, underscoring the urgent need for novel therapeutic approaches. This review explores recent advances in nanoparticle-based therapies targeting fungal infections, emphasizing their unique potential to enhance drug solubility, bioavailability, and targeted delivery. Nanoparticles offer the ability to penetrate biological barriers, improve drug stability, and act as direct antifungal agents by disrupting fungal cell walls and generating reactive oxygen species. Despite their promising applications, challenges such as potential toxicity, scalability of production, and the need for controlled drug release remain. Future research should focus on optimizing nanoparticle properties, evaluating long-term safety profiles, developing environmentally sustainable synthesis methods, and exploring synergistic approaches with existing antifungal drugs. Nanotechnology offers a transformative opportunity in the management of fungal diseases, paving the way for more effective and targeted treatments.
Collapse
Affiliation(s)
- Qinglin Wu
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Fulan Cen
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Ying Xie
- Graduate School of Public Administration, Seoul National University, Seoul, Republic of South Korea
| | - Xianjia Ning
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jinghua Wang
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Zhenghao Lin
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jia Huang
- Department of Intensive Care Unit, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
7
|
Schmidt S, Schwerk C, Schroten H, Ishikawa H, Schubert R, Lehrnbecher T, Rudolph H. Hyphal penetration is the major pathway of translocation of Candida albicans across the blood-cerebrospinal fluid barrier. Fluids Barriers CNS 2025; 22:34. [PMID: 40181464 PMCID: PMC11969880 DOI: 10.1186/s12987-025-00644-x] [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: 01/29/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Despite the availability of potent antifungal compounds, invasive fungal disease poses significant morbidity and mortality in immunocompromised patients. Candida albicans is one of the leading pathogens in this setting, and may affect the central nervous system (CNS), which is an extremely severe form of the infection. As the exact pathogenesis of Candida CNS infection is not clear, we investigated the mechanisms and effects of C. albicans transmigration into the CNS, which will be helpful for diagnosis, prevention and treatment. METHODS We used a human in vitro model of the Blood-Cerebrospinal Fluid Barrier (BCSFB), and we investigated the mechanisms of Candida albicans translocation into the CNS. Translocation was evaluated using immunofluorescence analysis focusing on tight and adherens junctions and the actin cytoskeleton. Barrier integrity was monitored via measurement of transepithelial resistance and the paracellular permeability of dextran. LIVE/DEAD assays were applied for viability controls and a cytometric bead array was performed to detect cytokine secretion of plexus epithelial cells. RESULTS Translocation at low doses occurs transcellularly in the absence of cytotoxicity or secretion of proinflammatory cytokines. This is accomplished by the formation of a tunnel-like structure exploiting the actin cytoskeleton. With higher infection doses of Candida albicans, a reduction in barrier integrity due to disruption of tight and adherens junctions was observed and cytotoxicity also increased. CONCLUSION Our findings reveal that Candida albicans can use transcellular translocation to invade into the CNS and is able to circumvent major host immune response, which may impact on diagnostic and preventive strategies.
Collapse
Affiliation(s)
- S Schmidt
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - C Schwerk
- Pediatric Infectious Diseases, Medical Faculty Mannheim, University Children's Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - H Schroten
- Pediatric Infectious Diseases, Medical Faculty Mannheim, University Children's Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - H Ishikawa
- Laboratory of Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - R Schubert
- Department of Pediatrics, Division of Pneumology, Allergology, Infectious Diseases and Gastroenterology, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - T Lehrnbecher
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - H Rudolph
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt/Main, Germany.
| |
Collapse
|
8
|
Blair BA, Bragdon E, Dhillon G, Baker N, Stasiak L, Muthig M, Miramon P, Lorenz MC, Wheeler RT. Forward genetic screen in zebrafish identifies new fungal regulators that limit host-protective Candida-innate immune interaction. mBio 2025:e0052925. [PMID: 40172223 DOI: 10.1128/mbio.00529-25] [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: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/04/2025] Open
Abstract
Candida is one of the most frequent causes of bloodstream infections, and our first line of defense against these invasive infections is the innate immune system. The early immune response is critical in controlling Candida albicans infection, but C. albicans has several strategies to evade host immune attack. Phagocytosis of C. albicans blocks hyphal growth, limiting host damage and virulence, but how C. albicans limits early recruitment and phagocytosis in vertebrate infection is poorly understood. To study innate immune evasion by intravital imaging, we utilized the transparent larval zebrafish infection model to screen 131 C. albicans mutants for altered virulence and phagocyte response. Infections with each of the seven hypovirulent mutants led to altered phagocyte recruitment and/or phagocytosis, falling into four categories. Of particular interest among these is NMD5, a predicted β-importin and newly identified virulence factor. The nmd5∆/∆ mutant fails to limit phagocytosis, and its virulence defects are eliminated when phagocyte activity is compromised, suggesting that its role in virulence is limited to immune evasion. These quantitative intravital imaging experiments are the first to document altered Candida-phagocyte interactions for several additional mutants and clearly distinguish recruitment from phagocytic uptake, suggesting that Candida modulates both events. This initial large-scale screen of individual C. albicans mutants in a vertebrate, coupled with high-resolution imaging of Candida-phagocyte interactions, provides a more nuanced view of how diverse mutations can lead to more effective phagocytosis, a key immune process that blocks germination and drives anti-fungal immunity. IMPORTANCE Candida albicans is part of the human microbial community and is a dangerous opportunistic pathogen, able to prevent its elimination by the host immune system. Although Candida avoids immune attack through several strategies, we still understand little about how it regulates when immune phagocytes get recruited to the infection site and when they engulf fungal cells. We tested over 130 selected Candida mutants for their ability to cause lethal infection and found several hypovirulent mutants, which provoked altered innate immune responses, resulting in lower overall inflammation and greater host survival. Of particular interest is NMD5, which acts to limit fungal phagocytosis and is predicted to regulate the activity of stress-associated transcription factors. Our high-content screening was enabled by modeling Candida infection in transparent vertebrate zebrafish larva. Our findings help us understand how Candida survives immune attack during commensal and pathogenic growth, and may eventually inform new strategies for controlling disease.
Collapse
Affiliation(s)
- Bailey A Blair
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Emma Bragdon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Gursimran Dhillon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Nnamdi Baker
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Lena Stasiak
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Mya Muthig
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Pedro Miramon
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael C Lorenz
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Robert T Wheeler
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| |
Collapse
|
9
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [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: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
10
|
Tamai R, Kiyoura Y. Candida Infections: The Role of Saliva in Oral Health-A Narrative Review. Microorganisms 2025; 13:717. [PMID: 40284554 PMCID: PMC12029948 DOI: 10.3390/microorganisms13040717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Candida species, particularly Candida albicans, are causative agents of oral infections to which immunocompromised patients are especially susceptible. Reduced saliva flow (xerostomia) can lead to Candida overgrowth, as saliva contains antibacterial components such as histatins and β-defensins that inhibit fungal growth and adhesion to the oral mucosa. Candida adheres to host tissues, forms biofilms, and secretes enzymes required for tissue invasion and immune evasion. Secretory asparaginyl proteinases (Saps) and candidalysin, a cytolytic peptide toxin, are vital to Candida virulence, and agglutinin-like sequence (Als) proteins are crucial for adhesion, invasion, and biofilm formation. C. albicans is a risk factor for dental caries and may increase periodontal disease virulence when it coexists with Porphyromonas gingivalis. Candida infections have been suggested to heighten the risk of oral cancer based on a relationship between Candida species and oral squamous cell carcinoma (OSCC) or oral potentially malignant disorder (OPMD). Meanwhile, β-glucan in the Candida cell wall has antitumor effects. In addition, Candida biofilms protect viruses such as herpesviruses and coxsackieviruses. Understanding the intricate interactions between Candida species, host immune responses, and coexisting microbial communities is essential for developing preventive and therapeutic strategies against oral Candida infections, particularly in immunocompromised individuals.
Collapse
Affiliation(s)
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| |
Collapse
|
11
|
Gangiah TK, Alisoltani A, Potgieter M, Bell L, Ross E, Iranzadeh A, McDonald Z, Allali I, Dabee S, Barnabas S, Blackburn JM, Tabb DL, Bekker LG, Jaspan HB, Passmore JAS, Mulder N, Masson L. Exploring the female genital tract mycobiome in young South African women using metaproteomics. MICROBIOME 2025; 13:76. [PMID: 40108637 PMCID: PMC11921665 DOI: 10.1186/s40168-025-02066-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Female genital tract (FGT) diseases such as bacterial vaginosis (BV) and sexually transmitted infections are prevalent in South Africa, with young women being at an increased risk. Since imbalances in the FGT microbiome are associated with FGT diseases, it is vital to investigate the factors that influence FGT health. The mycobiome plays an important role in regulating mucosal health, especially when the bacterial component is disturbed. However, we have a limited understanding of the FGT mycobiome since many studies have focused on bacterial communities and have neglected low-abundance taxonomic groups, such as fungi. To reduce this knowledge deficit, we present the first large-scale metaproteomic study to define the taxonomic composition and potential functional processes of the FGT mycobiome in South African reproductive-age women. RESULTS We examined FGT fungal communities present in 123 women by collecting lateral vaginal wall swabs for liquid chromatography-tandem mass spectrometry. From this, 39 different fungal genera were identified, with Candida dominating the mycobiome (53.2% relative abundance). We observed changes in relative abundance at the protein, genus, and functional (gene ontology biological processes) level between BV states. In women with BV, Malassezia and Conidiobolus proteins were more abundant, while Candida proteins were less abundant compared to BV-negative women. Correspondingly, Nugent scores were negatively associated with total fungal protein abundance. The clinical variables, Nugent score, pro-inflammatory cytokines, chemokines, vaginal pH, Chlamydia trachomatis, and the presence of clue cells were associated with fungal community composition. CONCLUSIONS The results of this study revealed the diversity of FGT fungal communities, setting the groundwork for understanding the FGT mycobiome. Video Abstract.
Collapse
Affiliation(s)
- Tamlyn K Gangiah
- Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, 7925, South Africa
- Department of Soil and Environment, Swedish University of Agricultural Sciences, 750 07, Uppsala, Sweden
| | - Arghavan Alisoltani
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
| | - Matthys Potgieter
- Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, 7925, South Africa
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, University of Cape Town, Cape Town, 7925, South Africa
| | - Liam Bell
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Elizabeth Ross
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Arash Iranzadeh
- Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, 7925, South Africa
| | - Zac McDonald
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Imane Allali
- Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, 7925, South Africa
- Laboratory of Human Pathologies Biology, Department of Biology and Genomic Center of Human Pathologies, Mohammed V University in Rabat, Rabat, Morocco
| | - Smritee Dabee
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Seattle Children'S Research Institute, University of Washington, Seattle, WA, 98101, USA
| | - Shaun Barnabas
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
| | - Jonathan M Blackburn
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
| | - David L Tabb
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Bioinformatics Unit, South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Stellenbosch, 7602, South Africa
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Stellenbosch, 7602, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Desmond Tutu HIV Centre, Cape Town, University of Cape Town, Cape Town, 7925, South Africa
| | - Heather B Jaspan
- Seattle Children'S Research Institute, University of Washington, Seattle, WA, 98101, USA
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
| | - Jo-Ann S Passmore
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, 4013, South Africa
- National Health Laboratory Service, Cape Town, 7925, South Africa
| | - Nicola Mulder
- Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for Infectious Diseases Research (CIDRI) in Africa Wellcome Trust Centre, University of Cape Town, Cape Town, 7925, South Africa
| | - Lindi Masson
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa.
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa.
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, 4013, South Africa.
- Women's, Children's and Adolescents' Health and Disease Elimination Programs, Life Sciences Discipline, Burnet Institute, Melbourne, 3004, Australia.
- Central Clinical School, Monash University, Melbourne, 3004, Australia.
| |
Collapse
|
12
|
Singh R, Shukla J, Ali M, Dubey AK. A Novel Benzopyrone Derivative from Streptomyces chrestomyceticus ADP4 Inhibits Growth and Virulence Factors of Candida albicans. Curr Microbiol 2025; 82:201. [PMID: 40100410 DOI: 10.1007/s00284-025-04169-2] [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: 09/24/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025]
Abstract
Antimicrobial resistance (AMR) poses a serious threat to human health globally. Expeditious discovery and development of new drugs has become indispensable for addressing this challenge. In this context, a novel benzopyrone derivative, designated as 82B1, has been isolated from S. chrestomyceticus strain ADP4. This compound exhibited significant inhibitory activity against different Candida species including C. albicans, C. tropicalis, C. krusei, C. parapsilosis and C. auris with minimum inhibitory concentration (MIC90) values in the concentration range of 25-125 µg/mL. The structure of 82B1 was elucidated through analyses of the spectral data obtained using liquid chromatography-tandem mass spectrometry (LCMS/MS), Nuclear Magnetic Resonance (NMR), Fourier Transform Infrared Spectroscopy (FTIR) and Ultraviolet (UV) spectroscopy, that led to its identity as 7, 13, 14-trihydroxy-6H-benzo-[c]-chromen-6-one8-[1'β-carboxycyclopentanyl]-2'β-[8'β-ethylcyclopentane]. It significantly inhibited the major virulence factors of C. albicans such as yeast to hyphae transition, biofilm formation, and secretion of hydrolytic enzymes at its subinhibitory concentrations. It did not display cytotoxicity on human hepatoblastoma cell line (HepG2 cells), signifying its potential as a candidate for anti-Candida drug development.
Collapse
Affiliation(s)
- Radha Singh
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, 110078, India
| | - Jyoti Shukla
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, 110078, India
| | - Mohd Ali
- Faculty of Pharmacy, Hamdard University, New Delhi, 110062, India
| | - Ashok K Dubey
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, 110078, India.
| |
Collapse
|
13
|
Sun YY, Liu NN. Mycobiome: an underexplored kingdom in cancer. Microbiol Mol Biol Rev 2025:e0026124. [PMID: 40084887 DOI: 10.1128/mmbr.00261-24] [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: 03/16/2025] Open
Abstract
SUMMARYThe human microbiome, including bacteria, fungi, archaea, and viruses, is intimately linked to both health and disease. The relationship between bacteria and disease has received much attention and intensive investigation, while that of the fungal microbiome, also known as mycobiome, has lagged far behind bacteria. There is growing evidence showing mycobiome dysbiosis in cancer patients, and certain cancer-specific fungi may contribute to cancer progression by interacting with both host and bacteria. It was also demonstrated that the role of fungi-derived products in cancer should also not be underestimated. Therefore, investigating how fungal pathogenesis contributes to the onset and spread of cancer would yield crucial information for cancer diagnosis, prevention, and anti-cancer therapy.
Collapse
Affiliation(s)
- Yan-Yan Sun
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Kontoyiannis DP, Casadevall A. Would global warming bring an increase of invertebrate-associated cutaneous invasive fungal infections? mBio 2025; 16:e0344724. [PMID: 39907444 PMCID: PMC11898599 DOI: 10.1128/mbio.03447-24] [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] [Indexed: 02/06/2025] Open
Abstract
Invasive mold-associated cutaneous disease is a rare but potentially catastrophic consequence of trauma. However, invertebrate bites are not well recognized as a mechanism for the inoculation of fungi into subcutaneous tissue that can also result in severe infections. Invertebrates often carry fungi with human pathogenic potential as part of their microbiome, and bites break the skin, providing a conduit for them to penetrate subcutaneous tissues where the establishment of infection can produce serious skin and soft tissue fungal diseases. In this essay, we review the existing data for invertebrate bite-associated cutaneous invasive fungal infections (IBA-cIFIs) and consider the potential consequences of global warming on their epidemiology. Climate changes will be associated with changes in the range of invertebrates and adaptation of their associated microbes to warmer temperatures. Fungal adaptation to higher temperatures can defeat the mammalian protective barrier and be associated with both more and different IBA-cIFIs.
Collapse
Affiliation(s)
- Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Iliev ID, Ananthakrishnan AN, Guo CJ. Microbiota in inflammatory bowel disease: mechanisms of disease and therapeutic opportunities. Nat Rev Microbiol 2025:10.1038/s41579-025-01163-0. [PMID: 40065181 DOI: 10.1038/s41579-025-01163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 03/26/2025]
Abstract
Perturbations in the intestinal microbiome are strongly linked to the pathogenesis of inflammatory bowel disease (IBD). Bacteria, fungi and viruses all make up part of a complex multi-kingdom community colonizing the gastrointestinal tract, often referred to as the gut microbiome. They can exert various effects on the host that can contribute to an inflammatory state. Advances in screening, multiomics and experimental approaches have revealed insights into host-microbiota interactions in IBD and have identified numerous mechanisms through which the microbiota and its metabolites can exert a major influence on the gastrointestinal tract. Looking into the future, the microbiome and microbiota-associated processes will be likely to provide unparalleled opportunities for novel diagnostic, therapeutic and diet-inspired solutions for the management of IBD through harnessing rationally designed microbial communities, powerful bacterial and fungal metabolites, individually or in combination, to foster intestinal health. In this Review, we examine the current understanding of the cross-kingdom gut microbiome in IBD, focusing on bacterial and fungal components and metabolites. We examine therapeutic and diagnostic opportunities, the microbial metabolism, immunity, neuroimmunology and microbiome-inspired interventions to link mechanisms of disease and identify novel research and therapeutic opportunities for IBD.
Collapse
Affiliation(s)
- Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology and Hepatology Division, Weill Cornell Medicine, New York, NY, USA.
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Chun-Jun Guo
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology and Hepatology Division, Weill Cornell Medicine, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
16
|
Tosiano MA, Lanni F, Mitchell AP, McManus CJ. Roles of P-body factors in Candida albicans filamentation and stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.09.602714. [PMID: 40161774 PMCID: PMC11952329 DOI: 10.1101/2024.07.09.602714] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Hyphal growth is strongly associated with virulence in the human fungal pathogen Candida albicans. While hyphal transcriptional networks have been the subject of intense study, relatively little is known about post-transcriptional regulation. Previous work reported that P-Body (PB) factors Dhh1 and Edc3 were required for C. albicans virulence and filamentation, suggesting an essential role for post-transcriptional regulation of these processes. However, the molecular roles of these factors have not been determined. To further study the function of PB factors in filamentation, we generated homozygous deletions of DHH1 and EDC3 in diverse prototrophic clinical strains using transient CRISPR-Cas9. Homozygous DHH1 deletion strongly impaired growth, altered filamentation, and exhibited unusual colony morphology in response to heat stress in five strain backgrounds. Using RNA-seq, we found DHH1 deletion disrupts the regulation of thousands of genes under both yeast and hyphal growth conditions in SC5314 and P57055. This included upregulation of many stress response genes in the absence of external stress, similar to deletion of the S. cerevisiae DHH1 homolog. In contrast, we found EDC3 was not required for heat tolerance or filamentation in diverse strains. These results support a model in which DHH1, but not EDC3, represses hyphal stress response transcripts in yeast and remodels the transcriptome during filamentation. Our work supports distinct requirements for specific mRNA decay factors, bolstering evidence for post-transcriptional regulation of filamentation in C. albicans.
Collapse
Affiliation(s)
- Melissa A. Tosiano
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
17
|
Wronowska E, Guevara-Lora I, Brankiewicz A, Bras G, Zawrotniak M, Satala D, Karkowska-Kuleta J, Budziaszek J, Koziel J, Rapala-Kozik M. Synergistic effects of Candida albicans and Porphyromonas gingivalis biofilms on epithelial barrier function in a 3D aspiration pneumonia model. Front Cell Infect Microbiol 2025; 15:1552395. [PMID: 40125517 PMCID: PMC11925950 DOI: 10.3389/fcimb.2025.1552395] [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: 12/28/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Polymicrobial infections involving Candida albicans and Porphyromonas gingivalis represent a significant challenge in maintaining epithelial barrier integrity. This study explores their synergistic effects on epithelial cells using an air-liquid interface (ALI) model. Methods Mixed-species biofilms were developed and analyzed for their impact on epithelial permeability and tight junction proteins. The effects of biofilm supernatants on IL-8 secretion and oxidative stress markers were also evaluated. The role of P. gingivalis proteases was assessed using a gingipain-null mutant (ΔKΔRAB) compared to the wild-type strain (W83). Survival experiments were conducted using Galleria mellonella larvae to examine the pathogenicity of dual-species biofilms. Results Mixed-species biofilms significantly increased epithelial permeability and disrupted tight junction proteins, as evidenced by reduced levels of ZO-1 and E-cadherin. These changes were accompanied by oxidative stress, characterized by decreased HO-1 expression and enhanced Bax/Bcl-xL ratios, indicating increased pro-apoptotic activity. Supernatants from dual-species biofilms demonstrated a pronounced effect on epithelial cells, modulating IL-8 secretion and exacerbating oxidative damage. C. albicans was identified as the dominant driver of pro-inflammatory responses, while P. gingivalis contributed through immune modulation and enzymatic activity, primarily via gingipains. The ΔKΔRAB mutant biofilms caused less epithelial disruption and oxidative stress compared to the wild-type, highlighting the critical role of gingipains in pathogenesis. Discussion Survival experiments using Galleria mellonella larvae supported these findings, highlighting the reduced survival associated with dual-species biofilms and the potential for high-dose antimicrobial therapies to mitigate this effect. These results emphasize the cooperative mechanisms of C. albicans and P. gingivalis in compromising epithelial barriers and underline the importance of combination therapies targeting both fungal and bacterial components in polymicrobial infections.
Collapse
Affiliation(s)
- Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Ibeth Guevara-Lora
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Brankiewicz
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Grazyna Bras
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satala
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Joanna Budziaszek
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
18
|
Molaeitabari A, Dahms TES. Blocking the shikimate pathway amplifies the impact of carvacrol on biofilm formation in Candida albicans. Microbiol Spectr 2025; 13:e0275424. [PMID: 39918333 DOI: 10.1128/spectrum.02754-24] [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: 11/05/2024] [Accepted: 12/14/2024] [Indexed: 03/05/2025] Open
Abstract
Candida albicans typically thrives in a commensal relationship with humans but is also an opportunistic fungal pathogen. As an opportunistic pathogen, C. albicans relies heavily on its ability to assimilate nutrients, for which it must compete with the host and other microorganisms. Amino acid biosynthesis, sensing, and uptake play pivotal roles in C. albicans growth and pathogenicity. C. albicans biosynthesizes aromatic amino acids and co-enzyme Q de novo through the shikimate pathway, including the Aro1, Aro2, and Aro7 enzymes, but also has amino acid transporters for uptake from the environment. Thus, antifungal approaches targeting aromatic amino acid biosynthesis must simultaneously inhibit amino acid biosynthesis and uptake. Herein, we investigate the plant-based antifungal, carvacrol, in conjunction with aromatic amino acid biosynthetic mutants, as a potential anti-candidal strategy. Growth of the WT, ARO2, and ARO7 strains were inhibited by 150 µg/mL carvacrol, whereas the ARO1 mutant was slightly more sensitive (with MIC 125 µg/mL). All repressed mutants exposed to carvacrol are partially rescued in the presence of para-aminobenzoic acid (PABA) (CoQ precursor), indicating that blocking the shikimate pathway impacts both aromatic amino acid and CoQ biosynthesis. Moreover, carvacrol at sublethal concentrations significantly inhibits ARO1 adhesion and hyphal formation, along with pre-attached and pre-formed hyphae, ultimately impacting biofilm metabolic activity and biomass accumulation and significantly reducing biofilm growth. In summary, carvacrol increases the sensitivity of C. albicans to ARO1 repression, with attenuated adhesion, hyphal formation, mycelial growth and biofilm formation, likely by blocking aromatic amino acid uptake.IMPORTANCEThe opportunistic pathogen Candida albicans remains the leading cause of candidemia and invasive candidiasis (IC), causing significant morbidity and mortality in immunocompromised patients. Our current arsenal of effective antifungal drugs is limited in number, mechanistic diversity, and efficacy, are cytotoxic and associated with antifungal resistance, necessitating the development of novel antifungals and combination therapies. Here, we show how simultaneously blocking the shikimate pathway, through ARO1 repression, and disrupting aromatic amino acid uptake by carvacrol prevent C. albicans biofilm formation. Thus, inhibitors of the Aro1 enzyme in combination with carvacrol are expected to shut down C. albicans biofilm formation and virulence.
Collapse
Affiliation(s)
- Ali Molaeitabari
- Department of Chemistry and Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Tanya E S Dahms
- Department of Chemistry and Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| |
Collapse
|
19
|
Deng X, Li H, Wu A, He J, Mao X, Dai Z, Tian G, Cai J, Tang J, Luo Y. Composition, Influencing Factors, and Effects on Host Nutrient Metabolism of Fungi in Gastrointestinal Tract of Monogastric Animals. Animals (Basel) 2025; 15:710. [PMID: 40075993 PMCID: PMC11898470 DOI: 10.3390/ani15050710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Intestinal fungi, collectively referred to as mycobiota, constitute a small (0.01-2%) but crucial component of the overall intestinal microbiota. While fungi are far less abundant than bacteria in the gut, the volume of an average fungal cell is roughly 100-fold greater than that of an average bacterial cell. They play a vital role in nutrient metabolism and maintaining intestinal health. The composition and spatial organization of mycobiota vary across different animal species and are influenced by a multitude of factors, including age, diet, and the host's physiological state. At present, quantitative research on the composition of mycobiota in monogastric animals remains scarce, and investigations into the mechanisms underlying their metabolic functions are also relatively restricted. This review delves into the distribution characteristics of mycobiota, including Candida albicans, Saccharomyces cerevisiae, Kazachstania slooffiae, in monogastric animals, the factors influencing their composition, and the consequent impacts on host metabolism and health. The objective is to offer insights for a deeper understanding of the nutritional significance of intestinal fungi in monogastric animals and to explore the mechanisms by which they affect host health in relation to inflammatory bowel disease (IBD), diarrhea, and obesity. Through a systematic evaluation of their functional contributions, this review shifts our perception of intestinal fungi from overlooked commensals to key components in gut ecosystem dynamics, emphasizing their potential in personalized metabolic control regulation and the enhancement of disease prevention and treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.D.); (H.L.); (A.W.); (J.H.); (X.M.); (Z.D.); (G.T.); (J.C.); (J.T.)
| |
Collapse
|
20
|
Tosiano MA, Lanni F, Mitchell AP, McManus CJ. Roles of P-body factors in Candida albicans filamentation and stress response. PLoS Genet 2025; 21:e1011632. [PMID: 40096135 PMCID: PMC11975087 DOI: 10.1371/journal.pgen.1011632] [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: 07/10/2024] [Revised: 04/07/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
Hyphal growth is strongly associated with virulence in the human fungal pathogen Candida albicans. While hyphal transcriptional networks have been the subject of intense study, relatively little is known about post-transcriptional regulation. Previous work reported that P-Body (PB) factors Dhh1 and Edc3 were required for C. albicans virulence and filamentation, suggesting an essential role for post-transcriptional regulation of these processes. However, the molecular roles of these factors have not been determined. To further study the function of PB factors in filamentation, we generated homozygous deletions of DHH1 and EDC3 in diverse prototrophic clinical strains using transient CRISPR-Cas9. Homozygous DHH1 deletion strongly impaired growth, altered filamentation, and exhibited unusual colony morphology in response to heat stress in five strain backgrounds. Using RNA-seq, we found DHH1 deletion disrupts the regulation of thousands of genes under both yeast and hyphal growth conditions in SC5314 and P57055. This included upregulation of many stress response genes in the absence of external stress, similar to deletion of the S. cerevisiae DHH1 homolog. In contrast, we found EDC3 was not required for heat tolerance or filamentation in diverse strains. These results support a model in which DHH1, but not EDC3, represses hyphal stress response transcripts in yeast and remodels the transcriptome during filamentation. Our work supports distinct requirements for specific mRNA decay factors, bolstering evidence for post-transcriptional regulation of filamentation in C. albicans.
Collapse
Affiliation(s)
- Melissa A. Tosiano
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
21
|
Reitler P, DeJarnette CA, Kumar R, Tucker KM, Peters TL, Twarog NR, Shelat AA, Palmer GE. A screen to identify antifungal antagonists reveals a variety of pharmacotherapies induce echinocandin tolerance in Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638903. [PMID: 40027746 PMCID: PMC11870487 DOI: 10.1101/2025.02.18.638903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Through screening a comprehensive collection of drugs approved for human use, we identified over 20 that oppose the antifungal activity of the echinocandins upon the infectious yeast, Candida albicans . More detailed evaluation of five such drugs, including the atypical antipsychotic aripiprazole and the tyrosine kinase inhibitor ponatinib, indicated they promote C. albicans survival following exposure to the echinocandin antifungals. The activity of the five selected antagonists was dependent upon the Mkc1p MAPK pathway, however, ponatinib was paradoxically shown to suppress phosphorylation and therefore activation of Mkc1p itself. Components of several other signaling pathways are also required, including those of calcineurin and casein kinase-2, suggesting the observed antagonism required much of the cell wall stress responses previously described for C. albicans . Transcriptome analysis revealed that the antagonists stimulated the expression of genes involved in xenobiotic and antifungal resistance, and suppressed the expression of genes associated with hyphal growth. Thus, the echinocandin antagonistic drugs modulate C. albicans physiology in ways that could impact its pathogenicity and/or response to therapeutic intervention. Finally, a mutant lacking the Efg1p transcription factor, which has a central role in the activation of C. albicans hyphal growth was found to have intrinsically high levels of echinocandin tolerance, suggesting a link between modulation of morphogenesis related signaling and echinocandin tolerance. Importance We report a substantial number of previously unknown drug interactions that modulate the echinocandin sensitivity of one of the most prevalent human fungal pathogens, Candida albicans . The echinocandins are the first line therapy for treating disseminated and often lethal Candida infections, that account for >75% of invasive fungal infections in the U.S.. For largely unknown reasons, a substantial number of patients with invasive candidiasis fail to respond to treatment with these drugs. The finding of this study suggest that co-administered medications have the potential to influence the therapeutic outcomes of invasive fungal infections through modulating antifungal drug tolerance and/or fungal pathogenicity. The potential for echinocandin antagonistic medications to influence therapeutic outcomes is discussed.
Collapse
Affiliation(s)
- Parker Reitler
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Christian A. DeJarnette
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Ravinder Kumar
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Katie M. Tucker
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Tracy L. Peters
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Nathaniel R Twarog
- Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Anang A. Shelat
- Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Glen E. Palmer
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| |
Collapse
|
22
|
Blair BA, Bragdon E, Dhillon G, Baker N, Stasiak L, Muthig M, Miramon P, Lorenz MC, Wheeler RT. Forward genetic screen in zebrafish identifies new fungal regulators that limit host-protective Candida-innate immune interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638315. [PMID: 39990375 PMCID: PMC11844468 DOI: 10.1101/2025.02.14.638315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Candida is one of the most frequent causes of bloodstream infections, and our first line of defense against these invasive infections is the innate immune system. The early immune response is critical in controlling C. albicans infection, but C. albicans has several strategies to evade host immune attack. Phagocytosis of C. albicans blocks hyphal growth, limiting host damage and virulence, but how C. albicans limits early recruitment and phagocytosis in vertebrate infection is poorly understood. To study innate immune evasion by intravital imaging, we utilized the transparent larval zebrafish infection model to screen 131 C. albicans mutants for altered virulence and phagocyte response. Infections with each of seven hypovirulent mutants led to altered phagocyte recruitment and/or phagocytosis, falling into four categories. Of particular interest among these is NMD5, a predicted β-importin and newly-identified virulence factor. The nmd5∆/∆ mutant fails to limit phagocytosis and its virulence defects are eliminated when phagocyte activity is compromised, suggesting that its role in virulence is limited to immune evasion. These quantitative intravital imaging experiments are the first to document altered Candida-phagocyte interactions for several additional mutants, and clearly distinguish recruitment from phagocytic uptake, suggesting that Candida modulates both events. This initial large-scale screen of individual C. albicans mutants in a vertebrate, coupled with high-resolution imaging of Candida-phagocyte interactions, provides a more nuanced view of how diverse mutations can lead to more effective phagocytosis, a key immune process which blocks germination and drives anti-fungal immunity.
Collapse
Affiliation(s)
- Bailey A. Blair
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469
| | - Emma Bragdon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Gursimran Dhillon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Nnamdi Baker
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Lena Stasiak
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Mya Muthig
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Pedro Miramon
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | - Robert T. Wheeler
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469
| |
Collapse
|
23
|
Morelli M, Queiroz K. Breaking Barriers: Candidalysin Disrupts Epithelial Integrity and Induces Inflammation in a Gut-on-Chip Model. Toxins (Basel) 2025; 17:89. [PMID: 39998106 PMCID: PMC11861147 DOI: 10.3390/toxins17020089] [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: 01/20/2025] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Candida albicans is an opportunistic pathogenic yeast commonly found in the gastrointestinal tract of healthy humans. Under certain conditions, it can become invasive and cause life-threatening systemic infections. One mechanism used by C.albicans to breach the epithelial barrier is the secretion of candidalysin, a cytolytic peptide toxin. Candidalysin damages epithelial membranes and activates the innate immune response, making it key to C.albicans' pathogenicity and a promising therapeutic target. Although candidalysin mediates C. albicans translocation through intestinal layers, its impact on epithelial responses is not fully understood. This study aims to characterize this response and develop scalable, quantitative methodologies to assess candidalysin's toxicological effects using gut-on-chip models. We used the OrganoPlate® platform to expose Caco-2 tubules to candidalysin and evaluated their response with trans-epithelial electrical resistance (TEER), protein detection, and immunostaining. We then validated our findings in a proof-of-concept experiment using human intestinal organoid tubules. Candidalysin impaired barrier integrity, induced actin remodeling, and increased cell permeability. It also induced the release of LDH, cytokines, and the antimicrobial peptide LL37, suggesting cellular damage, inflammation, and antimicrobial activity. This study strengthens our understanding of candidalysin's role in C. albicans pathogenesis and suggests new therapeutic strategies targeting this toxin. Moreover, patient-derived organoids show promise for capturing patient heterogeneity and developing personalized treatments.
Collapse
Affiliation(s)
- Moran Morelli
- MIMETAS B.V., De Limes 7, 2342 DH Oegstgeest, The Netherlands
| | | |
Collapse
|
24
|
Zhou T, Solis NV, Marshall M, Yao Q, Pearlman E, Filler SG, Liu H. Fungal Als proteins hijack host death effector domains to promote inflammasome signaling. Nat Commun 2025; 16:1562. [PMID: 39939579 PMCID: PMC11821908 DOI: 10.1038/s41467-025-56657-5] [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: 07/03/2024] [Accepted: 01/27/2025] [Indexed: 02/14/2025] Open
Abstract
High-damaging Candida albicans strains tend to form hyphae and exacerbate intestinal inflammation in ulcerative colitis patients through IL-1β-dependent mechanisms. Fungal agglutinin-like sequence (Als) proteins worsen DSS-induced colitis in mouse models. FADD and caspase-8 are important regulators of gut homeostasis and inflammation. However, whether they link directly to fungal proteins is not fully understood. Here, we report that Als proteins induce IL-1β release in immune cells. We show that hyphal Als3 is internalized in macrophages and interacts with caspase-8 and the inflammasome adaptor apoptosis-associated speck-like protein containing a CARD (ASC). Caspase-8 is essential for Als3-mediated ASC oligomerization and IL-1β processing. In non-immune cells, Als3 is associated with cell death core components FADD and caspase-8. N-terminal Als3 (N-Als3) expressed in Jurkat cells partially inhibits apoptosis. Mechanistically, N-Als3 promotes oligomerization of FADD and caspase-8 through their death effector domains (DEDs). N-Als3 variants with a mutation in the peptide-binding cavity or amyloid-forming region are impaired in DED oligomerization. Together, these results demonstrate that DEDs are intracellular sensors of Als3. This study identifies additional potential targets to control hypha-induced inflammation.
Collapse
Affiliation(s)
- Tingting Zhou
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Norma V Solis
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Michaela Marshall
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Qing Yao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Gilead Sciences Inc, Foster City, CA, USA
| | - Eric Pearlman
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Scott G Filler
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Haoping Liu
- Department of Biological Chemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
25
|
Álvaro-Moya M, Blesa A, Prieto D, Román E, Pla J, Alonso-Monge R. Identification of Candida albicans Antigens Recognized by Murine Intestinal IgAs by a Gel-Independent Immunoproteomic Approach. J Proteome Res 2025; 24:657-671. [PMID: 39804704 DOI: 10.1021/acs.jproteome.4c00691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2025]
Abstract
As part of the intestinal microbiota, Candida albicans can elicit a humoral response in the gastrointestinal tract (GIT) that is mainly directed toward hyphal antigens. This response has been implicated in controlling the invasive form of the fungus and maintaining the yeast as an innocuous commensal. However, the specific targets of this response are still unknown. Here, we used a gel-free immunoproteomic methodology to identify C. albicans gut immunogens. For this goal, we previously obtained specific secreted IgA from mice colonized with C. albicans. Then, secretome and surfome from C. albicans wild-type filaments were obtained and incubated with magnetic beads linked to antimouse IgA antibodies. sIgA targets were immunoprecipitated and analyzed by mass spectrometry. A third sample bearing the C. albicans antigen-sIgA complex was also examined. Those identified proteins that exhibited a higher percentile of relative abundance were considered for further analysis. From those, 35 proteins coincided among the three samples. Remarkably, about 40% of the identified proteins appeared in the databases as uncharacterized. The results were then validated by immunofluorescence assays overexpressing some of the genes identified in a yeast-lock C. albicans mutant. Adhesins such as Als3, Als1, and Hwp1 were corroborated to be IgA targets, as well as the chaperone Ssa2. Therefore, this gel-free immunoproteomic approach has been useful to identify new C. albicans antigens that generate a specific humoral response in the murine gut.
Collapse
Affiliation(s)
- Marina Álvaro-Moya
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Alba Blesa
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Daniel Prieto
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Elvira Román
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Jesús Pla
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Rebeca Alonso-Monge
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| |
Collapse
|
26
|
Xu B, Luo Z, Niu X, Li Z, Lu Y, Li J. Fungi, immunosenescence and cancer. Semin Cancer Biol 2025; 109:67-82. [PMID: 39788169 DOI: 10.1016/j.semcancer.2025.01.002] [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: 11/05/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Fungal microbes are a small but immunoreactive component of the human microbiome, which may influence cancer development, progression and therapeutic response. Immunosenescence is a process of immune dysfunction that occurs with aging, including lymphoid organ remodeling, contributing to alterations in the immune system in the elderly, which plays a critical role in many aspects of cancer. There is evidence for the interactions between fungi and immunosenescence in potentially regulating cancer progression and remodeling the tumor microenvironment (TME). In this review, we summarize potential roles of commensal and pathogenic fungi in modulating cancer-associated processes and provide more-detailed discussions on the mechanisms of which fungi affect tumor biology, including local and distant regulation of the TME, modulating antitumor immune responses and interactions with neighboring bacterial commensals. We also delineate the features of immunosenescence and its influence on cancer development and treatment, and highlight the interactions between fungi and immunosenescence in cancer. We discuss the prospects and challenges for harnessing fungi and immunosenescence in cancer diagnosis and/or treatment. Considering the limited understanding and techniques in conducting such research, we also provide our view on how to overcome challenges faced by the exploration of fungi, immunosenescence and their interactions on tumor biology.
Collapse
Affiliation(s)
- Bin Xu
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi 330029, China
| | - Zan Luo
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi 330029, China
| | - Xing Niu
- Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, China; Voylin Institute for Translation Medicine, Xiamen, Fujian 361000, China
| | - Zhi Li
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Yeping Lu
- Department of Neurosurgery, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, China.
| | - Junyu Li
- Department of Radiation Oncology, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi 330029, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
27
|
Bilal H, Khan MN, Khan S, Shafiq M, Fang W, Zeng Y, Guo Y, Li X, Zhao B, Lv QL, Xu B. Fungal Influences on Cancer Initiation, Progression, and Response to Treatment. Cancer Res 2025; 85:413-423. [PMID: 39589783 DOI: 10.1158/0008-5472.can-24-1609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/13/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
Fungal dysbiosis is increasingly recognized as a key factor in cancer, influencing tumor initiation, progression, and treatment outcomes. This review explores the role of fungi in carcinogenesis, with a focus on mechanisms such as immunomodulation, inflammation induction, tumor microenvironment remodeling, and interkingdom interactions. Fungal metabolites are involved in oncogenesis, and antifungals can interact with anticancer drugs, including eliciting potential adverse effects and influencing immune responses. Furthermore, mycobiota profiles have potential as diagnostic and prognostic biomarkers, emphasizing their clinical relevance. The interplay between fungi and cancer therapies can affect drug resistance, therapeutic efficacy, and risk of invasive fungal infections associated with targeted therapies. Finally, emerging strategies for modulating mycobiota in cancer care are promising approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Hazrat Bilal
- Jiangxi Key Laboratory of Oncology, JXHC Key Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Muhammad Nadeen Khan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Sabir Khan
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Muhammad Shafiq
- Department of Pharmacology, Research Institute of Clinical Pharmacy, Shantou University Medical College, Shantou, China
| | - Wenjie Fang
- Department of Dermatology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yuebin Zeng
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yangzhong Guo
- Jiangxi Key Laboratory of Oncology, JXHC Key Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Xiaohui Li
- Jiangxi Key Laboratory of Oncology, JXHC Key Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Bing Zhao
- Jiangxi Key Laboratory of Oncology, JXHC Key Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Qiao-Li Lv
- Jiangxi Key Laboratory of Oncology, JXHC Key Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Bin Xu
- Jiangxi Key Laboratory of Oncology, JXHC Key Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| |
Collapse
|
28
|
Li Z, Wu H, Li C, Wang Y, Shao J, Wu D, Wang T, Wang C. n-butanol extract of Pulsatilla decoction alleviates vulvovaginal candidiasis via the regulation of mitochondria-associated Type I interferon signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119292. [PMID: 39732299 DOI: 10.1016/j.jep.2024.119292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vulvovaginal candidiasis (VVC) is a relatively common fungal infectious disease in the female reproductive tract. The pathogenesis of VVC not only involves Candida albicans (C. albicans) infection, but also the improper immune response of the vaginal mucosal immune system to the fungus. As a classical formula, Pulsatilla decoction has been proven to exert protective effect in both clinical and experimental research in VVC. However, the specific mechanism of Pulsatilla decoction in VVC remains elusive. This study investigated the mechanism of n-butanol extract of Pulsatilla decoction (BEPD) in treating VVC from the perspective of type I interferon signaling and related mitochondrial function. MATERIALS AND METHODS A VVC-rat model was developed using an estrogen-based method to evaluate the effectiveness of BEPD in treating VVC, and the therapeutic efficacy of BEPD against VVC was comprehensively evaluated by fungal morphology and burden, neutrophil numbers, histopathology, pro-inflammatory and anti-inflammatory cytokines production, and LDH level. Immunohistochemistry (IHC), immune fluorescence (IF), Western Blot (WB) and RT-qPCR assays were conducted to assess type I interferon signaling and mitochondria functions. Candidalysin-induced vaginal epithelial inflammation in vitro, as cellular models, was employed to detect the changes in type I interferon signaling and mitochondria function before and after BEPD-containing serum intervention. RESULTS BEPD could significantly improve the inflammation, reduce fungal loads and inhibit fungal growth, balance pro-inflammatory and anti-inflammatory cytokine levels in VVC model rats. The findings from IHC, IF, WB and RT-qPCR revealed that BEPD could promote type I interferon signaling and alleviate mitochondrial functional damage in VVC model rats, and BEPD-containing serum could play the same role in vitro. CONCLUSION The study findings generally demonstrated that BEPD could improve the inflammation and correct the immune imbalance in VVC through regulation of type I interferon signaling and mitochondrial function.
Collapse
Affiliation(s)
- Ziyi Li
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hui Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Can Li
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yemei Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Shao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tianming Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Changzhong Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.
| |
Collapse
|
29
|
Praetorius JP, Hitzler SUJ, Gresnigt MS, Figge MT. Image-based quantification of Candida albicans filamentation and hyphal length using the open-source visual programming language JIPipe. FEMS Yeast Res 2025; 25:foaf011. [PMID: 40082735 PMCID: PMC11963753 DOI: 10.1093/femsyr/foaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/16/2025] Open
Abstract
The formation of hyphae is one of the most crucial virulence traits the human pathogenic fungus Candida albicans possesses. The assessment of hyphal length in response to various stimuli, such as exposure to human serum, provides valuable insights into the adaptation strategies of C. albicans to the host environment. Despite the increasing high-throughput capacity live-cell imaging and data generation, the accurate analysis of hyphal growth has remained a laborious, error-prone, and subjective manual process. We developed an analysis pipeline utilizing the open-source visual programming language Java Image Processing Pipeline (JIPipe) to overcome the limitations associated with manual analysis of hyphal growth. By comparing our automated approach with manual analysis, we refined the strategies to achieve accurate differentiation between yeast cells and hyphae. The automated method enables length measurements of individual hyphae, facilitating a time-efficient, high-throughput, and user-friendly analysis. By utilizing this JIPipe analysis approach, we obtained insights into the filamentation behavior of two C. albicans strains when exposed to human serum albumin (HSA), the most abundant protein in human serum. Our findings indicate that despite the known role of HSA in stimulating fungal growth, it reduces filamentous growth. The implementation of our automated JIPipe analysis approach for hyphal growth represents a long-awaited and time-efficient solution to meet the demand of high-throughput data generation. This tool can benefit different research areas investigating the virulence aspects of C. albicans.
Collapse
Affiliation(s)
- Jan-Philipp Praetorius
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), 07745 Jena, Germany
| | - Sophia U J Hitzler
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), 07745 Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, 07745 Jena, Germany
| |
Collapse
|
30
|
Znaidi S. When HSFs bring the heat-mapping the transcriptional circuitries of HSF-type regulators in Candida albicans. mSphere 2025; 10:e0064423. [PMID: 39704513 PMCID: PMC11774045 DOI: 10.1128/msphere.00644-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Heat shock factor (HSF)-type regulators are stress-responsive transcription factors widely distributed among eukaryotes, including fungi. They carry a four-stranded winged helix-turn-helix DNA-binding domain considered as the signature domain for HSFs. The genome of the opportunistic yeast Candida albicans encodes four HSF members, namely, Sfl1, Sfl2, Skn7, and the essential regulator, Hsf1. C. albicans HSFs do not only respond to heat shock and/or temperature variation but also to CO2 levels, oxidative stress, and quorum sensing, acting this way as central decision makers. In this minireview, I follow on the heels of my mSphere of Influence commentary (2020) to provide an overview of the repertoire of HSF regulators in Saccharomyces cerevisiae and C. albicans and describe how their genetic perturbation in C. albicans, coupled with genome-wide expression and location analyses, allow to map their transcriptional circuitry. I highlight how they can regulate, in common, a crucial developmental program: filamentous growth.
Collapse
Affiliation(s)
- Sadri Znaidi
- Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique, Tunis, Tunisia
- Institut Pasteur, INRA, Département Mycologie, Unité Biologie et Pathogénicité Fongiques, Paris, France
| |
Collapse
|
31
|
Liao B, Zhang C, Shen J, Chen D, Wang J, Chen X, Zhou Y, Wei Y, Shi Y, Gou L, Guo Q, Zhou X, Xie H, Zhao L, Liao G, Zhu Z, Cheng L, Zhou X, Li Y, Ren B. Aloin remodels the cell wall of Candida albicans to reduce its hyphal virulence against oral candidiasis. Appl Microbiol Biotechnol 2025; 109:21. [PMID: 39853490 PMCID: PMC11761986 DOI: 10.1007/s00253-025-13411-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Aloe vera (L.) Burm.f. is a traditional Chinese medicine known for treating various ailments, including fungal infections. Aloin is one of the major components from A. vera, but its antifungal mechanism and therapeutic potential against oral candidiasis are not clear. This study aimed to examine the mechanism of aloin against Candida albicans and its inhibitory activity against oral candidiasis. In this study, we for the first time found that aloin could induce the formation of abnormal hyphae with smaller hyphal diameters and fewer branching points in C. albicans including 11 clinical isolates without growth inhibition. The transcriptome and further cell wall contents analysis indicated that aloin remodeled the cell wall to increase the contents of β-1,3-glucan and furtherly showed an antagonistic effect with micafungin. Aloin also significantly inhibited the cell damage of oral epithelial cells and oral candidiasis in mice infected by C. albicans due to its inhibitory actions on the hyphal development and expressions of virulence factors, including candidalysin (coded by ECE1). Our results suggest that aloin is a promising antifungal agent for controlling candidiasis and targeting hyphal development and pathogenesis represents a practical strategy for developing new antifungal drugs. KEY POINTS: • Aloin remodels the C. albicans cell wall to form avirulent hyphae. • Aloin inhibits C. albicans infections in oral epithelial cells and mouse mucosa without toxicity. • Aloin is a promising antifungal agent with therapeutic potential against C. albicans infections.
Collapse
Affiliation(s)
- Binyou Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chuanli Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Wei
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hongyu Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lin Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Information Management & Department of Stomatology Informatics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
32
|
Cosio T, Romeo A, Pistoia ES, Pica F, Freni C, Iacovelli F, Orlandi A, Falconi M, Campione E, Gaziano R. Retinoids as Alternative Antifungal Agents Against Candida albicans: In Vitro and In Silico Evidence. Microorganisms 2025; 13:237. [PMID: 40005604 PMCID: PMC11857849 DOI: 10.3390/microorganisms13020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Candida albicans (C. albicans) is the most common pathogen responsible for a wide spectrum of human infections ranging from superficial mucocutaneous mycoses to systemic life-threatening diseases. Its main virulence factors are the morphological transition between yeast and hyphal forms and the ability to produce biofilm. Novel antifungal strategies are required given the severity of systemic candidiasis, especially in immunocompromised patients, and the lack of effective anti-biofilm treatments. We previously demonstrated that all-trans retinoic acid (ATRA), an active metabolite of vitamin A, exerted an inhibitory effect on Candida growth, yeast-hyphal transition and biofilm formation. Here, we further investigated the possible anti-Candida potential of trifarotene and tazarotene, which are the other two molecules belonging to the retinoid family, compared to ATRA. The results indicate that both drugs were able to suppress Candida growth, germination and biofilm production, although trifarotene was proven to be more effective than tazarotene, showing effectiveness comparable to ATRA. In silico studies suggest that all three retinoids may exert antifungal activity through their molecular interactions with the heat shock protein (Hsp) 90 and 14α-demethylase of C. albicans. Moreover, interactions between retinoids and ergosterol have been observed, suggesting that those compounds have great potential against C. albicans infections.
Collapse
Affiliation(s)
- Terenzio Cosio
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Alice Romeo
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Enrico Salvatore Pistoia
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
| | - Francesca Pica
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
| | - Claudia Freni
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Augusto Orlandi
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Mattia Falconi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Elena Campione
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
| |
Collapse
|
33
|
Veerapandian R, Paudyal A, Schneider SM, Lee STM, Vediyappan G. A mouse model of immunosuppression facilitates oral Candida albicans biofilms, bacterial dysbiosis and dissemination of infection. Front Cell Infect Microbiol 2025; 14:1467896. [PMID: 39902181 PMCID: PMC11788080 DOI: 10.3389/fcimb.2024.1467896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/26/2024] [Indexed: 02/05/2025] Open
Abstract
Opportunistic pathogens are a major threat to people, especially those with impaired immune systems. Two of the most important microbes in this category are the fungus Candida albicans and Gram-positive bacteria of the genus Enterococcus, which share overlapping niches in the oral cavity, gastrointestinal and urogenital tracts. The clinical importance of oral C. albicans biofilm and its interaction with the host under immunosuppressive conditions remains largely understudied. Here, we used a mouse model of oropharyngeal candidiasis (OPC) with cortisone acetate injection on alternate days and a continuous supply of C. albicans in drinking water for three days, resulting in immunosuppression. Results showed abundant growth of resident oral bacteria and a strong C. albicans biofilm on the tongue consisting of hyphae which damaged papillae, the epidermal layer, and invaded tongue tissue with the accumulation of inflammatory cells as demonstrated by Grocott's methenamine silver and hematoxylin and eosin staining, respectively. The dispersed microbes from the oral biofilm colonized the gastrointestinal (GI) tract and damaged its integrity, disseminating microbes to other organs. Although no visible damage was observed in the kidney and liver, except increased lipid vacuoles in the liver cells, C. albicans was found in the liver homogenate. Intriguingly, we found co-occurrence of Enterococcus faecalis in the tongue, liver, and stool of immunosuppressed control and C. albicans infected organs. Targeted 16S rRNA and ITS2 amplicon sequencing of microbes from the fecal samples of mice confirmed the above results in the stool samples and revealed an inverse correlation of beneficial microbes in the dysbiosis condition. Our study shows that mucosal-oral infection of C. albicans under immunosuppressed conditions causes tissue damage and invasion in local and distant organs; the invasion may be aided by the overgrowth of the resident endogenous Enterobacteriaceae and other members, including the opportunistic pathogen Enterococcus faecalis.
Collapse
Affiliation(s)
- Raja Veerapandian
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Anuja Paudyal
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sarah M. Schneider
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Sonny T. M. Lee
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Govindsamy Vediyappan
- Division of Biology, Kansas State University, Manhattan, KS, United States
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
34
|
Ding T, Liu C, Li Z. The mycobiome in human cancer: analytical challenges, molecular mechanisms, and therapeutic implications. Mol Cancer 2025; 24:18. [PMID: 39815314 PMCID: PMC11734361 DOI: 10.1186/s12943-025-02227-8] [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: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
The polymorphic microbiome is considered a new hallmark of cancer. Advances in High-Throughput Sequencing have fostered rapid developments in microbiome research. The interaction between cancer cells, immune cells, and microbiota is defined as the immuno-oncology microbiome (IOM) axis. Fungal microbes (the mycobiome), although representing only ∼ 0.1-1% of the microbiome, are a critical immunologically active component of the tumor microbiome. Accumulating evidence suggests a possible involvement of commensal and pathogenic fungi in cancer initiation, progression, and treatment responsiveness. The tumor-associated mycobiome mainly consists of the gut mycobiome, the oral mycobiome, and the intratumoral mycobiome. However, the role of fungi in cancer remains poorly understood, and the diversity and complexity of analytical methods make it challenging to access this field. This review aims to elucidate the causal and complicit roles of mycobiome in cancer development and progression while highlighting the issues that need to be addressed in executing such research. We systematically summarize the advantages and limitations of current fungal detection and analysis methods. We enumerate and integrate these recent findings into our current understanding of the tumor mycobiome, accompanied by the prospect of novel and exhilarating clinical implications.
Collapse
Affiliation(s)
- Ting Ding
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Zhengyu Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Kulig K, Wronowska E, Juszczak M, Zawrotniak M, Karkowska-Kuleta J, Rapala-Kozik M. Host cell responses to Candida albicans biofilm-derived extracellular vesicles. Front Cell Infect Microbiol 2025; 14:1499461. [PMID: 39877654 PMCID: PMC11772320 DOI: 10.3389/fcimb.2024.1499461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025] Open
Abstract
Candida albicans is a prevalent fungal pathogen responsible for infections in humans. As described recently, nanometer-sized extracellular vesicles (EVs) produced by C. albicans play a crucial role in the pathogenesis of infection by facilitating host inflammatory responses and intercellular communication. This study investigates the functional properties of EVs released by biofilms formed by two C. albicans strains-3147 (ATCC 10231) and SC5314-in eliciting host responses. We demonstrate the capability of C. albicans EVs to trigger reactions in human epithelial and immune cells. The involvement of EVs in pathogenesis was evidenced from the initial stages of infection, specifically in adherence to epithelial cells. We further established the capacity of these EVs to induce cytokine production in the epithelial A549 cell line, THP-1 macrophage-like cells, and blood-derived monocytes differentiated into macrophages. Internalization of EVs by THP-1 macrophage-like cells was confirmed, identifying macropinocytosis and phagocytosis as the most probable mechanisms, as demonstrated using various inhibitors that target potential vesicle uptake pathways in human cells. Additionally, C. albicans EVs and their cargo were identified as chemoattractants for blood-derived neutrophils. After verification of the in vivo effect of biofilm-derived EVs on the host, using Galleria mellonella larvae as an alternative model, it was demonstrated that vesicles from C. albicans SC5314 increased mortality in the injected larvae. In conclusion, for both types of EVs a predominantly pro-inflammatory effect on host was observed, highlighting their significant role in the inflammatory response during C. albicans infection.
Collapse
Affiliation(s)
- Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
36
|
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
|
37
|
Janeczko M, Skrzypek T. Relationships Between Candida auris and the Rest of the Candida World-Analysis of Dual-Species Biofilms and Infections. Pathogens 2025; 14:40. [PMID: 39861001 PMCID: PMC11768094 DOI: 10.3390/pathogens14010040] [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: 11/05/2024] [Revised: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
In this study, we investigated the interactions between Candida auris and C. albicans, C. tropicalis, C. glabrata, and C. krusei in mixed infections. Initially, these interactions were studied qualitatively and quantitatively in dual-species biofilms formed in vitro. The MTT assays, determination of the total CFU/mL, and SEM analysis showed that C. auris interacted differentially with the other Candida spp. during the dual-species biofilm formation. Depending on the stage of the biofilm development, C. auris was found to be a particularly dominant species during its interaction with the C. krusei biofilms but significantly submissive in the C. auris-C. albicans biofilms. These studies were then extended to in vivo host models of experimental candidiasis. G. mellonella larvae were inoculated with monotypic and heterotypic suspensions of Candida. The survival rates and quantification of fungal cells in the hemolymph showed that the highest mortality was exhibited by larvae in the C. auris-C. albicans co-infection (100% mortality after 36 h). The CFU/mL values of C. auris from the larval hemolymph were lower in the interactive groups compared to the mono-species group. As a newly emerging species, C. auris persists in environments in the presence of other Candida species and is involved in both competitive and noncompetitive interactions with other Candida species during biofilm formation and development of experimental candidiasis.
Collapse
Affiliation(s)
- Monika Janeczko
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland
| | - Tomasz Skrzypek
- Department of Biomedicine and Environmental Research, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1j, 20-708 Lublin, Poland;
| |
Collapse
|
38
|
He J, Cheng L. The Oral Microbiome: A Key Determinant of Oral Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:133-149. [PMID: 40111690 DOI: 10.1007/978-3-031-79146-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
As the second largest reservoir of human microbes, the oral cavity is colonized by millions of tiny creatures collectively named as oral microbiome. Species detected in human mouth are diverse, including bacteria, fungi, viruses, and protozoa. Active bidirectional interaction exists between the oral microbiome and the host. Stresses from hosts shape the composition, distribution pattern, and the community behaviors of the oral microbiome, while any changes occurring on the oral microbiome may disrupt its symbiosis relationship with the host and ultimately lead to oral and systemic diseases that jeopardize the host's health. In this chapter, the latest understanding about the role of oral microbiome in common oral diseases, including dental caries, periodontal disease, oral candidiasis, and hyposalivation, is discussed.
Collapse
Affiliation(s)
- Jinzhi He
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
39
|
Patnaik S, Durairajan SSK, Singh AK, Krishnamoorthi S, Iyaswamy A, Mandavi SP, Jeewon R, Williams LL. Role of Candida species in pathogenesis, immune regulation, and prognostic tools for managing ulcerative colitis and Crohn's disease. World J Gastroenterol 2024; 30:5212-5220. [PMID: 39735273 PMCID: PMC11612695 DOI: 10.3748/wjg.v30.i48.5212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
The gut microbiome plays a key role in the pathogenesis and disease activity of inflammatory bowel disease (IBD). While research has focused on the bacterial microbiome, recent studies have shifted towards host genetics and host-fungal interactions. The mycobiota is a vital component of the gastrointestinal microbial community and plays a significant role in immune regulation. Among fungi, Candida species, particularly Candida albicans (C. albicans), have been extensively studied due to their dual role as gut commensals and invasive pathogens. Recent findings indicate that various strains of C. albicans exhibit considerable differences in virulence factors, impacting IBD's pathophysiology. Intestinal fungal dysbiosis and antifungal mucosal immunity may be associated to IBD, especially Crohn's disease (CD). This article discusses intestinal fungal dysbiosis and antifungal immunity in healthy individuals and CD patients. It discusses factors influencing the mycobiome's role in IBD pathogenesis and highlights significant contributions from the scientific community aimed at enhancing understanding of the mycobiome and encouraging further research and targeted intervention studies on specific fungal populations. Our article also provided insights into a recent study by Wu et al in the World Journal of Gastroenterology regarding the role of the gut microbiota in the pathogenesis of CD.
Collapse
Affiliation(s)
- Supriti Patnaik
- Molecular Mycology and Neurodegenerative Disease Research Laboratory, Department of Microbiology, Central University of Tamil Nadu, Thiruvarur 610005, India
| | - Siva Sundara Kumar Durairajan
- Molecular Mycology and Neurodegenerative Disease Research Laboratory, Department of Microbiology, Central University of Tamil Nadu, Thiruvarur 610005, India
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Abhay Kumar Singh
- Molecular Mycology and Neurodegenerative Disease Research Laboratory, Department of Microbiology, Central University of Tamil Nadu, Thiruvarur 610005, India
| | - Senthilkumar Krishnamoorthi
- Mr. & Mrs Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ashok Iyaswamy
- Mr. & Mrs Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Shiva Prasad Mandavi
- Department of Chemistry, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Rajesh Jeewon
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Reduit 80837, Mauritius
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Leonard L Williams
- Center for Excellence in Post Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, Kannapolis, NC 28081, United States
| |
Collapse
|
40
|
Penninger P, Brezovec H, Tsymala I, Teufl M, Phan-Canh T, Bitencourt T, Brinkmann M, Glaser W, Ellmeier W, Bonelli M, Kuchler K. HDAC1 fine-tunes Th17 polarization in vivo to restrain tissue damage in fungal infections. Cell Rep 2024; 43:114993. [PMID: 39580799 DOI: 10.1016/j.celrep.2024.114993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Histone deacetylases (HDACs) contribute to shaping many aspects of T cell lineage functions in anti-infective surveillance; however, their role in fungus-specific immune responses remains poorly understood. Using a T cell-specific deletion of HDAC1, we uncover its critical role in limiting polarization toward Th17 by restricting expression of the cytokine receptors gp130 and transforming growth factor β receptor 2 (TGF-βRII) in a fungus-specific manner, thus limiting Stat3 and Smad2/3 signaling. Controlled release of interleukin-17A (IL-17A) and granulocyte-macrophage colony-stimulating factor (GM-CSF) is vital to minimize apoptotic processes in renal tubular epithelial cells in vitro and in vivo. Consequently, animals harboring excess Th17-polarized HDCA1-deficient CD4+ T cells develop increased kidney pathology upon invasive Candida albicans infection. Importantly, pharmacological inhibition of class I HDACs similarly increased IL-17A release by both mouse and human CD4+ T cells. Collectively, this work shows that HDAC1 controls T cell polarization, thus playing a critical role in the antifungal immune defense and infection outcomes.
Collapse
Affiliation(s)
- Philipp Penninger
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Helena Brezovec
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Irina Tsymala
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Magdalena Teufl
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Trinh Phan-Canh
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Tamires Bitencourt
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; CCRI - St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Marie Brinkmann
- Medical University of Vienna, Division of Rheumatology, Department of Internal Medicine III, 1090 Vienna, Austria
| | - Walter Glaser
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, 1090 Vienna, Austria
| | - Michael Bonelli
- Medical University of Vienna, Division of Rheumatology, Department of Internal Medicine III, 1090 Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria.
| |
Collapse
|
41
|
Jaeger M, Dietschmann A, Austermeier S, Dinçer S, Porschitz P, Vornholz L, Maas RJ, Sprenkeler EG, Ruland J, Wirtz S, Azam T, Joosten LA, Hube B, Netea MG, Dinarello CA, Gresnigt MS. Alpha1-antitrypsin impacts innate host-pathogen interactions with Candida albicans by stimulating fungal filamentation. Virulence 2024; 15:2333367. [PMID: 38515333 PMCID: PMC11008552 DOI: 10.1080/21505594.2024.2333367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
Our immune system possesses sophisticated mechanisms to cope with invading microorganisms, while pathogens evolve strategies to deal with threats imposed by host immunity. Human plasma protein α1-antitrypsin (AAT) exhibits pleiotropic immune-modulating properties by both preventing immunopathology and improving antimicrobial host defence. Genetic associations suggested a role for AAT in candidemia, the most frequent fungal blood stream infection in intensive care units, yet little is known about how AAT influences interactions between Candida albicans and the immune system. Here, we show that AAT differentially impacts fungal killing by innate phagocytes. We observed that AAT induces fungal transcriptional reprogramming, associated with cell wall remodelling and downregulation of filamentation repressors. At low concentrations, the cell-wall remodelling induced by AAT increased immunogenic β-glucan exposure and consequently improved fungal clearance by monocytes. Contrastingly, higher AAT concentrations led to excessive C. albicans filamentation and thus promoted fungal immune escape from monocytes and macrophages. This underscores that fungal adaptations to the host protein AAT can differentially define the outcome of encounters with innate immune cells, either contributing to improved immune recognition or fungal immune escape.
Collapse
Affiliation(s)
- Martin Jaeger
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Sude Dinçer
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Pauline Porschitz
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Larsen Vornholz
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Ralph J.A. Maas
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evelien G.G. Sprenkeler
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, USA
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Mark S. Gresnigt
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| |
Collapse
|
42
|
Dickenson RE, Pellon A, Ponde NO, Hepworth O, Daniels Gatward LF, Naglik JR, Moyes DL. EGR1 regulates oral epithelial cell responses to Candida albicans via the EGFR- ERK1/2 pathway. Virulence 2024; 15:2435374. [PMID: 39635778 PMCID: PMC11622614 DOI: 10.1080/21505594.2024.2435374] [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: 01/15/2024] [Revised: 07/11/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Abstract
Candida albicans is a fungal pathobiont colonizing mucosal surfaces of the human body, including the oral cavity. Under certain predisposing conditions, C. albicans invades mucosal tissues activating EGFR-MAPK signalling pathways in epithelial cells via the action of its peptide toxin candidalysin. However, our knowledge of the epithelial mechanisms involved during C. albicans colonization is rudimentary. Here, we describe the role of the transcription factor early growth response protein 1 (EGR1) in human oral epithelial cells (OECs) in response to C. albicans. EGR1 expression increases in OECs when exposed to C. albicans independently of fungal viability, morphology, or candidalysin release, suggesting EGR1 is involved in the fundamental recognition of C. albicans, rather than in response to invasion or "pathogenesis." Upregulation of EGR1 is mediated by EGFR via Raf1, ERK1/2, and NF-κB signalling but not PI3K/mTOR signalling. Notably, EGR1 mRNA silencing impacts on anti-C. albicans immunity, reducing GM-CSF, IL-1α and IL-1β release, and increasing IL-6 and IL-8 production. These findings identify an important role for EGR1 in priming epithelial cells to respond to subsequent invasive infection by C. albicans and elucidate the regulation circuit of this transcription factor after contact.
Collapse
Affiliation(s)
- Ruth E. Dickenson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- Department of Infectious Diseases, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Aize Pellon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Nicole O. Ponde
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Olivia Hepworth
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Lydia F. Daniels Gatward
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - David L. Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| |
Collapse
|
43
|
Jørgensen MR. Pathophysiological microenvironments in oral candidiasis. APMIS 2024; 132:956-973. [PMID: 38571459 DOI: 10.1111/apm.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024]
Abstract
Oral candidiasis (OC), a prevalent opportunistic infection of the oral mucosa, presents a considerable health challenge, particularly in individuals with compromised immune responses, advanced age, and local predisposing conditions. A considerable part of the population carries Candida in the oral cavity, but only few develop OC. Therefore, the pathogenesis of OC may depend on factors other than the attributes of the fungus, such as host factors and other predisposing factors. Mucosal trauma and inflammation compromise epithelial integrity, fostering a conducive environment for fungal invasion. Molecular insights into the immunocompromised state reveal dysregulation in innate and adaptive immunity, creating a permissive environment for Candida proliferation. Detailed examination of Candida species (spp.) and their virulence factors uncovers a nuanced understanding beyond traditional C. albicans focus, which embrace diverse Candida spp. and their strategies, influencing adhesion, invasion, immune evasion, and biofilm formation. Understanding the pathophysiological microenvironments in OC is crucial for the development of targeted therapeutic interventions. This review aims to unravel the diverse pathophysiological microenvironments influencing OC development focusing on microbial, host, and predisposing factors, and considers Candida resistance to antifungal therapy. The comprehensive approach offers a refined perspective on OC, seeking briefly to identify potential therapeutic targets for future effective management.
Collapse
Affiliation(s)
- Mette Rose Jørgensen
- Section of Oral Pathology and Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
Sabbatini S, Zatini L, Narducci E, Rosati L, Ardizzoni A, Mencacci A, Rende M, Pericolini E, Galli F, Bartolini D, Monari C. Modulation of C. albicans-Induced Immune Response in Vaginal Epithelial Cells by Garcinoic Acid. Microorganisms 2024; 12:2455. [PMID: 39770658 PMCID: PMC11678841 DOI: 10.3390/microorganisms12122455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Vulvovaginal candidiasis (VVC) is a prevalent women's infection characterized by excessive inflammation and damage of the vaginal epithelium that, in its recurrent form (RVVC), causes more than three symptomatic episodes per year, impacting nearly 8% of women globally. Current antifungal treatments alleviate symptoms but often fail to restore the inflammatory homeostasis of mucosal tissue and prevent recurrences. α-Tocopherol (α-TOH) and garcinoic acid (GA), a vitamin E metabolite, with immunomodulatory properties, were investigated for the first time in vaginal epithelial cells exposed to C. albicans infection to assess their effects on inflammatory signaling parameters important to restore cellular homeostasis. For this purpose, the protein kinases MKK3/6, p38 stress kinase (SAPK), and ERK1/2 were studied together with c-Fos transcription factor and IL-6, IL-1α, and IL-1β secretion in A-431 vaginal epithelial cells pre-treated with GA or with α-TOH and then infected with C. albicans. GA, differently from α-TOH, significantly reduced the C. albicans-induced activation of p38-SAPK while increasing pro-survival MAPK ERK1/2 activity. This resulted in a significant reduction in the secretion levels of the inflammatory cytokines IL-6 and IL-1α, as well as IL-1β. Overall, our data indicate that GA holds potential for restoring the immuno-metabolic properties of the vaginal epithelium exposed to C. albicans infection, which may help to treat inflammatory symptoms in VVC/RVVC.
Collapse
Affiliation(s)
- Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| | - Linda Zatini
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy; (L.Z.); (F.G.)
| | - Eleonora Narducci
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| | - Lucrezia Rosati
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy;
| | - Andrea Ardizzoni
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.P.)
| | - Antonella Mencacci
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy;
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.P.)
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy; (L.Z.); (F.G.)
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy; (L.Z.); (F.G.)
| | - Claudia Monari
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| |
Collapse
|
45
|
Zhang D, Wang Q, Li D, Chen C, Lv Y, Huang S, Zeng F, Huang X, Mao F, Bai F. Different fungal signatures in ALD and MAFLD. Front Microbiol 2024; 15:1510507. [PMID: 39669777 PMCID: PMC11636606 DOI: 10.3389/fmicb.2024.1510507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
Objective This study investigates the differential impact of fecal fungal microbiota on the pathogenesis of alcohol-associated liver disease (ALD) and metabolic-associated fatty liver disease (MAFLD). We aim to delineate distinct microbial patterns across various stages of each disease. Methods We conducted fungal internal transcribed spacer 2 (ITS2) sequencing analysis on fecal samples from 48 ALD patients, 55 MAFLD patients, and 64 healthy controls (HCs). Results Distinct fungal microbiota profiles were significantly identified between the ALD and MAFLD patients. In the ALD group, genera such as Trichosporon, Davidiella and Asterotremella along with species like Trichosporon unclassified and Davidiella unclassified were elevated compared to those in the MAFLD group. Conversely, Fungi unclassified, Rhizopus, Periconia, and Candida albicans were more prevalent in MAFLD patients. A specific fungal signature comprising Asterotremella_pseudolonga, Malassezia_restricta and Malassezia, was notably effective in differentiating ALD from MAFLD, achieving an area under the curve (AUC) of 0.94. Periconia and Periconia byssoides were more abundant in non-obese MAFLD patients compared to obese MAFLD and HCs. Rhizopus microsporus var. chinensis and var. rhizopodiformis, along with Pleosporales unclassified, were predominantly found in MAFLD patients with moderate to severe hepatic steatosis (HS). The genera Pleosporales_unclassified and the species Candida_albicans were markedly elevated in ALC patients when contrasted with AFL or HCs. Conclusion This investigation introduces a novel fungal signature that successfully differentiates between ALD and MAFLD, underscoring Pleosporales unclassified, as biomarkers for disease progression in ALD and MAFLD. The findings also suggest a significant role for Periconia in the progression of non-obese MAFLD.
Collapse
Affiliation(s)
- Daya Zhang
- Graduate School, Hainan Medical University, Haikou, China
| | - Qi Wang
- Graduate School, Hainan Medical University, Haikou, China
| | - Da Li
- Graduate School, Hainan Medical University, Haikou, China
| | - Chen Chen
- Graduate School, Hainan Medical University, Haikou, China
| | - Yanting Lv
- Graduate School, Hainan Medical University, Haikou, China
| | - Shimei Huang
- Graduate School, Hainan Medical University, Haikou, China
| | - Fan Zeng
- Graduate School, Hainan Medical University, Haikou, China
| | - Xianfeng Huang
- Graduate School, Hainan Medical University, Haikou, China
| | - Fengjiao Mao
- Graduate School, Hainan Medical University, Haikou, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- The Gastroenterology Clinical Medical Center of Hainan Province, Haikou, China
| |
Collapse
|
46
|
Tesfamariam M, Vij R, Trümper V, Hube B, Brunke S. Shining a light on Candida-induced epithelial damage with a luciferase reporter. mSphere 2024; 9:e0050924. [PMID: 39412273 PMCID: PMC11580449 DOI: 10.1128/msphere.00509-24] [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: 06/13/2024] [Accepted: 09/17/2024] [Indexed: 11/22/2024] Open
Abstract
Host cell damage is a key parameter for research in infection biology, drug testing, and substance safety screening. In this study, we introduce a luciferase reporter system as a new and reliable assay to measure cell damage and validate it with the pathogenic yeast, Candida albicans, as a test case. We transduced human epithelial cell lines with a lentiviral vector to stably express an optimized luciferase enzyme, Nanoluc. Upon cell damage, the release of cytoplasmic luciferase into the extracellular space can be easily detected by a luminometer. We used the luciferase reporter system to investigate the damage caused by C. albicans to different newly generated epithelial reporter cell lines. We found that fungus-induced cell damage, as determined by established methods, correlated tightly with the release of the luciferase. The new luciferase reporter system is a simple, sensitive, robust, and inexpensive method for measuring host cell damage and has a sensitivity comparable to the standard assay, release of lactate dehydrogenase. It is suitable for high-throughput studies of pathogenesis mechanisms of any microbe, for antimicrobial drug screening, and many other applications.IMPORTANCEWe present a quick, easy, inexpensive, and reliable assay to measure damage to mammalian cells. To this end, we created reporter cell lines which artificially express luciferase, an enzyme that can be easily detected in the supernatant when these cells are damaged. We used infections with the well-investigated fungal pathogen of humans, Candida albicans, as a test case of our system. Using our reporter, we were able to recapitulate the known effects of strain variability, gene deletions, and antifungal treatments on host cell damage. This easily adaptable reporter system can be used to screen for damage in infection models with different microbial species, assay cell-damaging potential of substances, discover new non-toxic antibiotics, and many other damage-based applications.
Collapse
Affiliation(s)
- Millen Tesfamariam
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| | - Raghav Vij
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| | - Verena Trümper
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Sascha Brunke
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| |
Collapse
|
47
|
Ye X, Liu Y, Chen D, Liao B, Wang J, Shen J, Gou L, Zhou Y, Zhou X, Liao G, Zhou X, Zou J, Ren B. Moxidectin elevates Candida albicans ergosterol levels to synergize with polyenes against oral candidiasis. Appl Microbiol Biotechnol 2024; 108:509. [PMID: 39527144 PMCID: PMC11554702 DOI: 10.1007/s00253-024-13343-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Candida albicans, the most common opportunistic pathogenic fungus, is also the main pathogenic organism for oral candidiasis. This condition is particularly prevalent among the elderly, children, and individuals undergoing radiotherapy or suffering from HIV. The lack of new antifungal drugs, and drug resistance coupled with the side effects of current antifungal agents have increased the challenges of clinical antifungal therapies. Polyenes, including amphotericin B and nystatin, are clinical fungicidal drugs, however, their side effects and low solubility have limited their clinical applications. Here, we identified that moxidectin, a novel approved antiparasitic agent, could synergize with both amphotericin B and nystatin to inhibit the growth and biofilm formation of Candida albicans including 60 clinical isolates. The transcriptome and RT-PCR analysis indicated that moxidectin activated the biosynthesis pathway of ergosterol, the direct target of polyenes, further being verified by the loss of the synergistic activities with polyenes against ergosterol pathway mutants, including Δ/Δerg3, Δ/Δerg11 and Δ/Δerg3 Δ/Δerg11. Moxidectin was then confirmed to elevate the ergosterol biosynthesis levels of C. albicans and enhance the binding between cells and polyenes. In a mouse oral candidiasis model, moxidectin combined with low dosages of polyenes to significantly reduce the infection area, colonization of C. albicans and the inflammatory degree of tongue mucosa. Our study originally demonstrated that moxidectin could activate the ergosterol biosynthesis then elevate the ergosterol contents to enhance the antifungal effects of polyenes against C. albicans and its infections. Moxidectin can serve as the candidate potentiator of polyenes for further clinical practice. KEY POINTS: • Moxidectin synergized with polyenes against Candida albicans. • Moxidectin activated the ergosterol biosynthesis of Candida albicans. • Moxidectin combined with polyenes to effectively combat oral candidiasis in mice.
Collapse
Affiliation(s)
- Xingchen Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ga Liao
- Department of Information Management & Department of Stomatology Informatics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jing Zou
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China School of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
48
|
Ji Y, Chen D, Shao M, Liu Z, Li M, Yu Q. The P-type calcium pump Spf1 regulates immune response by maintenance of the endoplasmic reticulum-plasma membrane contacts during
Candida albicans
systemic infection. Mycology 2024:1-20. [DOI: 10.1080/21501203.2024.2409299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/20/2024] [Indexed: 01/03/2025] Open
Affiliation(s)
- Yuchao Ji
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin, China
| | - Dou Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin, China
| | - Menglin Shao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhuo Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin, China
| | - Mingchun Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin, China
| | - Qilin Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
49
|
Zhao N, Guo W, Li J, Wang H, Guo X. Rapid and accurate identification of yeast subspecies by MALDI-MS combined with a cell membrane disruption reagent. Food Chem 2024; 457:140102. [PMID: 38905823 DOI: 10.1016/j.foodchem.2024.140102] [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: 02/03/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) has been widely used for microbial analysis. However, due to the impenetrable shell of fungi the direct identification of fungi remains challenges. Targeting on this problem, the yeast Saccharomyces cerevisiae (S. cerevisiae) was selected as a model fungus, and a new fungal cell membrane disruption reagent C18-G1 was used before MALDI-MS detection. As a result, much more intensive peaks which distributed in wider m/z range of S. cerevisiae have been identified in comparison with the use of traditional fungal pretreatment methods. Furthermore, a differential peak at m/z 4993 between two subspecies of S. cerevisiae has been identified. The corresponding protein with exclusive sequence of the specific peak was obtained based on MS/MS fragments and database searching. In addition, the method was successfully applied for the discrimination of four commercial yeasts.
Collapse
Affiliation(s)
- Nan Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wei Guo
- Department of Nuclear Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jiarui Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Hao Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Xinhua Guo
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun 130012, China.
| |
Collapse
|
50
|
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
|