1
|
Dias LM, Paul P, Pavarina AC, Siqueira WL. Salivary proteins-enhanced antimicrobial photodynamic therapy: Overcoming three distinct cultures of resistant mixed biofilms. J Dent 2025; 157:105778. [PMID: 40268113 DOI: 10.1016/j.jdent.2025.105778] [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: 02/18/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Denture stomatitis is frequently associated with biofilm formation by Candida albicans, which can coexist with Streptococcus mutans. Current treatments face several limitations, including the emergence of resistant strains and the persistent impact of biofilm formation on antimicrobial efficacy. The salivary proteins Histatin 3 (His3) and Histatin 5 (His5) have demonstrated effectiveness against C. albicans single-species biofilms. However, their efficacy against mixed-species biofilms, particularly those involving S. mutans and antifungal-resistant C. albicans strains, remains poorly understood. OBJECTIVES To investigate the efficacy of combining His3 and His5 with antimicrobial photodynamic therapy (aPDT) against mixed biofilms containing polyene-resistant (CaP+Sm), wild-type (CaW+Sm), and fluconazole-resistant (CaF+Sm) and S. mutans (Sm) on acrylic resins. METHODS 48-hour mixed biofilms (37 °C/5 % CO₂) were formed on acrylic resin disks treated with His3 and His5 (2h/37 °C/120 rpm). Biofilms were subjected to aPDT using Photodithazine (200 mg/L) followed by 30 min of red LED irradiation (660 nm, 50 J/cm²). Viability was assessed by colony-forming units (CFU), while ECM components (proteins, alkali-soluble polysaccharides (ASP), water-soluble polysaccharides (WSP), and extracellular DNA (eDNA)) were analyzed (n = 6). RESULTS Complete eradication of mixed biofilms was observed in CaW+Sm and CaF+Sm treated with His3+aPDT and His5+aPDT, while CaP+Sm showed a 98 % reduction in total microbiota. For CaP+Sm, combined His3+aPDT and His5+aPDT significantly reduced biofilm viability, achieving up to 99 % reduction in C. albicans and 80 % in S. mutans. ECM components, including proteins, ASP, WSP, and eDNA, were notably reduced, particularly in CaW+Sm and CaF+Sm cultures. CONCLUSION Combining Histatins with aPDT demonstrated superior efficacy compared to individual treatments, disrupting mixed biofilms of C. albicans and S. mutans and significantly reducing viability. CLINICAL SIGNIFICANCE Histatins with antimicrobial photodynamic therapy (aPDT) reduce biofilm viability and disrupt key components of extracellular matrix in resistant biofilm that contribute to the persistence of infections in denture stomatitis.
Collapse
Affiliation(s)
- Luana Mendonça Dias
- College of Dentistry, University of Saskatchewan (USASK), Saskatoon, SK, Canada; Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, Brazil.
| | - Promi Paul
- College of Dentistry, University of Saskatchewan (USASK), Saskatoon, SK, Canada.
| | - Ana Claudia Pavarina
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, Brazil.
| | - Walter L Siqueira
- College of Dentistry, University of Saskatchewan (USASK), Saskatoon, SK, Canada.
| |
Collapse
|
2
|
Liu C, Yang L, Wang Z, Zhu H, Luo Q, Wu D, Wang T, Hu M, Wang C, Shao J. Qi-Huang decoction alleviates DSS-induced colitis with Candida albicans dysbiosis by enhancing innate immune response through Dectin-1-associated signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156613. [PMID: 40056633 DOI: 10.1016/j.phymed.2025.156613] [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: 10/27/2024] [Revised: 01/21/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory disease of the gastrointestinal tract. Candida albicans, a common commensal fungus in the human gut, has been increasingly implicated in UC pathogenesis. Qi-Huang decoction (QHD), a traditional Chinese herbal formula known for its spleen-invigorating and purgative properties, is commonly used to restore gastrointestinal function. PURPOSE This study investigates the therapeutic potential of QHD in treating colitis exacerbated by C. albicans and explores the underlying mechanisms of action. METHODS A mouse model of colitis was induced using dextran sulfate sodium combined with gavage of C. albicans. Following QHD treatment, colitis severity was evaluated by measuring survival rate, body weight, disease activity index, colon length, and fungal burden, and through histopathological analysis using hematoxylin-eosin staining. The expression of proinflammatory genes IL-1β and TNF-α was quantified, alongside protein levels of key molecules involved in Dectin-1 signaling, including Syk, CARD-9, NLRP-3, Raf-1, and NF-κB. Barrier integrity markers, such as Occludin and Claudin-1, were also examined. To further elucidate QHD's mechanisms, Dectin-1 was inhibited using laminarin. In vitro experiments assessed QHD's antifungal activity against three Candida strains through microdilution, spot assays, and time-kill tests. RAW 264.7 macrophages were employed to study the exposure of fungal cell wall β-glucan and subsequent phagocytosis. Molecular docking simulations predicted interactions between QHD's active compounds and the Dectin-1 receptor. RESULTS QHD significantly mitigated colitis severity and reduced fungal burden in vivo. QHD enhanced β-glucan exposure on the fungal cell wall, thereby stimulating phagocytosis by RAW264.7 macrophages. QHD effectively activated Dectin-1-mediated signaling pathways and increased proinflammatory levels in RAW 264.7 cells. In colitis mice, QHD treatment markedly reduced inflammation and Dectin-1 signaling following fungal clearance. However, Dectin-1 inhibition with LAM neutralized QHD's therapeutic effects, highlighting the pathway's importance in mediating QHD's efficacy. Interestingly, QHD alone elevated Dectin-1, NF-κB, TGF-β, and IL-10 levels, whereas reduced IL-1β and TNF-α expression, suggesting a dual modulatory role in inflammation. Molecular docking confirmed a potential direct interaction between QHD's bioactive components and the Dectin-1 receptor. CONCLUSION QHD demonstrates promising therapeutic potential for managing Candida colitis by modulating immune responses and targeting Dectin-1 signaling pathways in clinical settings.
Collapse
Affiliation(s)
- Chengcheng Liu
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Liu Yang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Zixu Wang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Hanyu Zhu
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Qinai Luo
- Department of Pathology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University o Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Daqiang Wu
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Tianming Wang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Min Hu
- Department of Pathology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University o Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China.
| | - Changzhong Wang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China
| | - Jing Shao
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, Anhui, PR China.
| |
Collapse
|
3
|
de Mello Fiallos N, Zanin dos Santos IJ, Brunson DN, Kajfasz JK, Zeng L, de Aguiar Cordeiro R, Lemos JA, Abranches J. Candida albicans impacts carbohydrate metabolism of Enterococcus faecalis in interkingdom biofilms. J Oral Microbiol 2025; 17:2492194. [PMID: 40247862 PMCID: PMC12004723 DOI: 10.1080/20002297.2025.2492194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Objectives This study investigated the transcriptional and physiological responses of Enterococcus faecalis, an opportunistic pathogen linked to endodontic infections, when cultivated in dual-species biofilms with Candida albicans, a yeast pathobiont found in the oral cavity. Methods Forty-eight-hour E. faecalis OG1RF biofilms were developed in BHI medium as mono- or dual-species with C. albicans SC5314. Biofilms were assessed for biomass, colony-forming units (CFUs), and architecture using confocal microscopy. RNA sequencing was performed on an Illumina platform. Mannose-PTS activity and glycerol quantification assays were conducted to investigate changes in carbohydrate metabolism. Results Transcriptomic analysis revealed 149 E. faecalis genes differentially expressed in dual-species biofilms. Genes linked to mannose-PTS and glycerol metabolism were notably upregulated. Mannose-PTS activity was significantly higher in dual-species biofilms. Mannose, as the sole carbohydrate source, increased E. faecalis CFUs and decreased C. albicans CFUs in co-culture, while glucose had no effect. As C. albicans is a glycerol net producer, glycerol levels were always higher when C. albicans was present, likely contributing to the upregulation of glycerol metabolism genes in E. faecalis when in co-cultures. Conclusions The presence of C. albicans alters E. faecalis gene expression and metabolism, suggesting metabolic crosstalk that may influence their pathogenicity and role in oral infections.
Collapse
Affiliation(s)
- Nicole de Mello Fiallos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
- Department of Pathology and Legal Medicine, Federal University of Ceará, College of Medicine, Fortaleza, Brazil
| | - Iriana J. Zanin dos Santos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
- Department of Cariology and Operative Dentistry, Federal University of Ceará, College of Dentistry, Sobral, Brazil
| | - Debra N. Brunson
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jessica K. Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Rossana de Aguiar Cordeiro
- Department of Pathology and Legal Medicine, Federal University of Ceará, College of Medicine, Fortaleza, Brazil
| | - José A. Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jacqueline Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
4
|
Stamation R. Endogenous Ethanol Production in the Human Alimentary Tract: A Literature Review. J Gastroenterol Hepatol 2025; 40:783-790. [PMID: 39853762 PMCID: PMC11968154 DOI: 10.1111/jgh.16869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/28/2024] [Accepted: 12/22/2024] [Indexed: 01/26/2025]
Abstract
Endogenous ethanol production, or auto-brewery syndrome (ABS), is a rare condition of the human alimentary canal that results in intoxication without alcohol consumption. Despite its clinical significance, ABS remains largely undiagnosed because of a lack of awareness among clinicians. Published cases have reported extensive biopsychosocial comorbidities accompanying delayed diagnosis and incomplete management; these include social rejection and family separation, court-ordered alcohol rehabilitation and psychiatric admission, legal and employment ramifications, and deteriorating mental health and suicidality. In this mini review, we aim to educate and enlighten clinicians by discussing literature findings pertaining to the pathophysiological mechanisms of gut dysbiosis due to overgrowth of Saccharomyces cerevisiae, E. coli and Klebsiella, impaired intestinal barrier function, and dysregulation of the hypothalamic-pituitary-adrenal axis. Furthermore, we discuss recently discovered associations with sleep quality and mood disorders and explore the medical sequelae of metabolic dysfunction-associated fatty liver disease and metabolic dysfunction-associated steatohepatitis. Drawing on these data, we propose protocols for initial care in the emergency room, subsequent critical care, diagnostic testing with glucose challenge testing, and definitive microbiological testing during the acute phase of illness. We also present an empirical treatment outline while awaiting confirmation of causative organisms and sensitivities.
Collapse
Affiliation(s)
- Renee Stamation
- Department of Rural HealthUniversity of Melbourne, Echuca Clinical SchoolEchucaVictoriaAustralia
| |
Collapse
|
5
|
Hwang YJ, Kim YY, Chang JY, Kho HS. Candidacidal activities of zinc compounds based on incubation time, pH, and ionic strength - An in vitro study. J Dent Sci 2025; 20:885-894. [PMID: 40224092 PMCID: PMC11993022 DOI: 10.1016/j.jds.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/04/2024] [Indexed: 04/15/2025] Open
Abstract
Background/purpose Given the antifungal activity of zinc compounds against Candida albicans, this study aimed to investigate the effects of incubation time, pH, and ionic strength on their candidacidal activity against C. albicans. Materials and methods Two zinc compounds (zinc chloride and sulfate) and three C. albicans strains (ATCC 10231, 11006, and 18804) were used. The parameters considered include incubation times of immediate, 10, 60, and 120 min, pH of 5.5, 5.0, 4.5, and 4.0, and ionic strength of mimicking human saliva, 100, 150, and 200 mM. The candidacidal activity was determined by comparing colony numbers on the experimental and control plates. Results The candidacidal activities of the zinc compounds significantly increased with increasing incubation time, decreasing pH, and increasing ionic strength. Significant levels of candidacidal activity were detected even within 10 min of incubation. The zinc compounds provided additional candidacidal activity under each pH and ionic strength condition. However, the effects of adding zinc compounds were less than those of lowering pH or increasing ionic strength. The candidacidal activities at each incubation time, pH, and ionic strength varied depending on the zinc compound and C. albicans strain; lower activities in zinc chloride and the 18804 strain. Conclusion The candidacidal activity of the zinc compounds occurred immediately. They provided additional candidacidal activities even at low pH or high ionic strength environments. Oral healthcare products containing zinc compounds can provide antifungal activities in a changing oral environment.
Collapse
Affiliation(s)
- Yoon-Jai Hwang
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Young Kim
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Ji-Youn Chang
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hong-Seop Kho
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
- Institute on Aging, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Chang TH, Cardona Gloria Y, Hellmann MJ, Richardo T, Greve CL, Le Roy D, Roger T, Bork F, Bugl S, Jakob J, Sonnberger J, Kasper L, Hube B, Pusch S, Gow NAR, Sørlie M, Tøndervik A, Moerschbacher BM, Weber ANR. Transkingdom mechanism of MAMP generation by chitotriosidase feeds oligomeric chitin from fungal pathogens and allergens into TLR2-mediated innate immune sensing. Front Immunol 2025; 16:1497174. [PMID: 40098951 PMCID: PMC11911531 DOI: 10.3389/fimmu.2025.1497174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Chitin is a highly abundant polysaccharide in nature and is linked to immune recognition of fungal infections and asthma in humans. Ubiquitous in fungi and insects, chitin is absent inmammals and plants and, thus, represents a microbeassociatedmolecular pattern (MAMP). However, highly polymeric chitin is insoluble, which potentially hampers recognition by host immune sensors. In plants, secreted chitinases degrade polymeric chitin into diffusible oligomers, which are "fed to" innate immune receptors and co-receptors. In human and murine immune cells, a similar enzymatic activity was shown for human chitotriosidase (CHIT1), and oligomeric chitin is sensed via an innate immune receptor, Toll-like receptor (TLR) 2. However, a complete system of generating MAMPs from chitin and feeding them into a specific receptor/co-receptor-aided sensing mechanism has remained unknown in mammals. Methods The effect of the secreted chitinolytic host enzyme, CHIT1, on the TLR2 activity of polymeric chitin preparations from shrimps, house dust mites and the fungal pathogen Candida albicans was assessed in vitro using cell lines and primary immune cells. Moreover, the regulation of CHIT1 was analyzed. Results Here, we show that CHIT1 converts inert polymeric chitin into diffusible oligomers that can be sensed by TLR1/TLR2 co-receptor/receptor heterodimers, a process promoted by the lipopolysaccharide binding protein (LBP) and CD14. Furthermore, we observed that Chit1 is induced via the b-glucan receptor Dectin-1 upon direct contact of immortalized human macrophages to the fungal pathogen Candida albicans, whereas the defined fungal secreted aspartyl proteases, Sap2 and Sap6, from C. albicans were able to degrade CHIT1 in vitro. Discussion Our study shows the existence of an inducible system of MAMP generation in the human host that enables contact-independent immune activation by diffusible MAMP ligands with a striking similarity to the plant kingdom. Moreover, this study highlights CHIT1 as a potential therapeutic target for TLR2-mediated inflammatory processes that are fueled by oligomeric chitin.
Collapse
Affiliation(s)
- Tzu-Hsuan Chang
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Yamel Cardona Gloria
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Margareta J. Hellmann
- Institute for Biology and Biotechnology of Plants, University of Münster, Münster, Germany
| | - Timmy Richardo
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Carsten Leo Greve
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Francesca Bork
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Stefanie Bugl
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Johanna Jakob
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Johannes Sonnberger
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Stefan Pusch
- Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University Heidelberg and German Cancer Consortium (DKTK), Clinical Cooperation Unit (CCU) Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Neil A. R. Gow
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at The University of Exeter, University of Exeter, Exeter, United Kingdom
| | - Morten Sørlie
- Department of Chemistry, Biotechnology, and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Anne Tøndervik
- Department of Biotechnology and Nanomedicine, Stiftelsen for Industriell og Teknisk Forskning (SINTEF) Industry, Trondheim, Norway
| | - Bruno M. Moerschbacher
- Institute for Biology and Biotechnology of Plants, University of Münster, Münster, Germany
| | - Alexander N. R. Weber
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
- iFIT – Cluster of Excellence (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- CMFI – Cluster of Excellence (EXC 2124) “Controlling Microbes to Fight Infection”, University of Tübingen, Tübingen, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK; German Cancer Consortium), Partner Site Tübingen, Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Cui Z, Yang WWJ, Yang ZH, Zhang LB, Guan Y. N-acetylglucosamine sensor, Ngs1 contributes to Beauveria bassiana vegetative growth, oxidative phosphorylation, fungal development, and cell wall integrity during entomopathogen-insect interaction. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 208:106273. [PMID: 40015865 DOI: 10.1016/j.pestbp.2024.106273] [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: 10/20/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 03/01/2025]
Abstract
N-acetylglucosamine (GlcNAc), a key component of fungal cell walls and insect cuticles, is an important signal to activate fungal response during entomopathogen-insect interactions. Research on Ngs1, the only identified GlcNAc sensor and transducer, has been primarily restricted to Candida species. Although our previous work identified an Ngs1 homology in Beauveria bassiana, its physiological functions in entomopathogenic fungi remain largely unexplored. In this study, we unveiled the sub-localization of Ngs1 in the nucleolus. Further transcriptomic analysis revealed that Ngs1 plays a crucial role in vegetative growth, fungal development, and cell-wall construction by acting as a transcriptional mediator, particularly influencing carbon metabolism in response to insect cuticle stimulation. The absence of Ngs1 compromised vegetative growth across various carbon sources by downregulating expressions of key catalytic enzymes. Conversely, Ngs1 deficiency enhanced transcription levels of oxidative phosphorylation, leading to increased ATP and reactive oxygen species (ROS) production. Despite higher ATP levels, Ngs1-deletion mutants exhibited reduced asexual development and hyphal germination, primarily due to the function of Ngs1 in the central developmental pathway and Brg1/Nrg1-dependent pathway. Additionally, the downregulation of N-glycan biosynthesis in ΔNgs1 diminished cell wall components, resulting in decreased cell wall resistance to lysis and impaired fungal development. These findings advance our understanding of the regulatory role of Ngs1 in B. bassiana during host interactions and provide a theoretical foundation for engineering fungi to maintain or even enhance pesticidal activity.
Collapse
Affiliation(s)
- Zhou Cui
- College of Biological Science and Engineering, Fuzhou University, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China
| | - Wu-Wei-Jie Yang
- College of Biological Science and Engineering, Fuzhou University, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China
| | - Zhi-Hao Yang
- College of Biological Science and Engineering, Fuzhou University, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China
| | - Long-Bin Zhang
- College of Biological Science and Engineering, Fuzhou University, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China.
| | - Yi Guan
- College of Biological Science and Engineering, Fuzhou University, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China.
| |
Collapse
|
8
|
Pan Y, Shi Z, Wang Y, Chen F, Yang Y, Ma K, Li W. Baicalin promotes β-1,3-glucan exposure in Candida albicans and enhances macrophage response. Front Cell Infect Microbiol 2024; 14:1487173. [PMID: 39717547 PMCID: PMC11664218 DOI: 10.3389/fcimb.2024.1487173] [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: 08/27/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Among the diverse fungal opportunistic pathogens, Candida albicans garners significant attention due to its wide range of infections and high frequency of occurrence. The emergence of resistance and the limited number of antifungals drives the need to develop novel antifungal drugs. Although the natural product baicalin has been shown to trigger apoptosis in C. albicans in previous experiments, its influence on cell wall (CW) structure along with immune recognition remains elusive. In this work, baicalin showed a significant killing effect against C. albicans SC5314. Moreover, CW destruction, characterized by β-1,3-glucan unmasking and chitin deposition, was observed as a consequence of the treatment with baicalin. The RNA sequencing analysis revealed that treatment with baicalin resulted in eight hundred forty-two differentially expressed genes (DEGs). Sixty-five genes, such as GSC1, ENG1, CHS3, GWT1, and MKC1, were associated with CW organization or biogenesis. Baicalin-pretreated C. albicans SC5314 was phagocytosed more efficiently by RAW264.7 macrophages, accompanied by increased TNF-α and IL-1β production. Accordingly, it is hypothesized that baicalin could stimulate β-1,3-glucan unmasking by governing CW-associated gene expression in C. albicans SC5314, which contributes to macrophage recognition and clearance.
Collapse
Affiliation(s)
- Yiyuan Pan
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
| | - Zhaoling Shi
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
| | - Yadong Wang
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
| | - Feng Chen
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
| | - Yue Yang
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
| | - Kelong Ma
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
| | - Wenqian Li
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
| |
Collapse
|
9
|
Ikeda E, Yamaguchi M, Ono M, Kawabata S. In Vitro Acid Resistance of Pathogenic Candida Species in Simulated Gastric Fluid. GASTRO HEP ADVANCES 2024; 4:100591. [PMID: 39996247 PMCID: PMC11847298 DOI: 10.1016/j.gastha.2024.100591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/05/2024] [Indexed: 02/26/2025]
Abstract
Background and Aims Although species in the fungal genus Candida are often commensal residents of the gastrointestinal (GI) tract, they can also cause high-mortality systemic candidiasis. Most pathogenic Candida species are dimorphic fungi that exist predominantly in filamentous forms in the invading tissues. Candida albicans is the most prominent pathogen among Candida species, but nonalbicans Candida species have also emerged as important pathogens. The stomach is the most acidic niche in the GI tract and is maintained at pH 1-2 in healthy individuals. The aim of the present study was to determine whether Candida species can survive in gastric fluid and to observe their morphology under varied pH conditions. Methods We investigated the in vitro survival of the pathogenic Candida species C. albicans, Candida glabrata, Candida parapsilosis, and Candida tropicalis in simulated gastric fluid. Results We first described that a portion of the 4 Candida species can survive under highly acidic conditions. Moreover, dimorphic Candida species, namely, C. albicans, C. parapsilosis, and C. tropicalis, exhibited yeast-hyphal transition in simulated gastric fluid with elevated pH. Pathogenic filamentous cells had lower acid resistance than yeast cells. Conclusion These findings may illuminate the migration to the lower GI tract by commensal fungi of the oral cavity.
Collapse
Affiliation(s)
- Eri Ikeda
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan
- Bioinformatics Research Unit, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masayuki Ono
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Bioinformatics Research Unit, Graduates School of Dentistry, Osaka University, Osaka, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan
| |
Collapse
|
10
|
Alonso MF, Bain JM, Erwig LP, Brown AJP, Gow NAR. Hyphal swelling induced in the phagosome of macrophages. Fungal Biol 2024; 128:2148-2156. [PMID: 39384284 PMCID: PMC11482207 DOI: 10.1016/j.funbio.2024.08.011] [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: 04/22/2024] [Revised: 08/09/2024] [Accepted: 08/28/2024] [Indexed: 10/11/2024]
Abstract
Macrophages play critical protective roles as sentinels of the innate immune system against fungal infection. It is therefore important to understand the dynamics of the interaction between these phagocytes and their fungal prey. We show here that many of the hyphal apices formed by Candida albicans within the macrophage ceased elongating, and apical and sub-apical hyphal compartments became swollen. Swollen hyphal cell compartments assimilated less Lysotracker-Red than non-swollen compartments, suggesting they had enhanced viability. Staining with florescent dyes suggested that there were higher levels of β-glucan and chitin in internalized fungal filaments compared to non-internalized hyphae, suggesting active cell wall remodelling within macrophages. These observations suggest that the stresses imposed by macrophages upon the fungus lead to changes in cell wall composition, inhibition of polarised growth and the induction of swelling in hyphal compartments, and that this can prevent or delay loss of viability of hyphal cells within the phagocyte.
Collapse
Affiliation(s)
- María Fernanda Alonso
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Judith M Bain
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Lars P Erwig
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Alistair J P Brown
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK.
| | - Neil A R Gow
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
11
|
Aldulaijan S, Alruwili R, Almulaify R, Alhassan FA, Al-Dulaijan YA, Alshahrani FA, Mokeem L, Gad MM, Melo MAS, Balhaddad AA. Benzyldimethyldodecyl Ammonium Chloride-Doped Denture-Based Resin: Impact on Strength, Surface Properties, Antifungal Activities, and In Silico Molecular Docking Analysis. J Funct Biomater 2024; 15:310. [PMID: 39452608 PMCID: PMC11508443 DOI: 10.3390/jfb15100310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Candida albicans (C. albicans) adhering to denture-based resins (DBRs) is a known cause of denture stomatitis. A new approach to prevent denture stomatitis is to include antimicrobial substances within DBRs. Here, we examined the mechanical performance and antifungal properties of DBRs containing benzyldimethyldodecyl ammonium chloride (C12BDMA-Cl) as an antimicrobial compound. C12BDMA-Cl is a quaternary ammonium compound, and its antifungal properties have never been investigated when combined with dental acrylic resin. Therefore, we modified a commercially available heat-polymerized acrylic DBR to contain 3 and 5 wt.% of C12BDMA-Cl. Unmodified DBR was used as a control group. Specimens were prepared using the conventional heat processing method. The specimen's flexural strength, elastic modulus, microhardness, and surface roughness were evaluated. C. albicans biofilm was grown on the specimens and assessed via colony-forming units (CFUs) and scanning electron microscopy (SEM). In silico molecular docking was applied to predict the potential C12BDMA-Cl inhibition activity as an antifungal drug. The 3% C12BDMA-Cl DBR demonstrated antifungal activities without a deterioration effect on the mechanical performance. SEM images indicated fewer colonies in DBR containing C12BDMA-Cl, which can be a potential approach to managing denture stomatitis. In conclusion, C12BDMA-Cl is a promising antifungal agent for preventing and treating denture stomatitis.
Collapse
Affiliation(s)
- Sarah Aldulaijan
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Raghad Alruwili
- College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (F.A.A.)
| | - Rawan Almulaify
- College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (F.A.A.)
| | - Fatimah A. Alhassan
- College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (F.A.A.)
| | - Yousif A. Al-Dulaijan
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (M.M.G.)
| | - Faris A. Alshahrani
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (M.M.G.)
| | - Lamia Mokeem
- Department of Restorative Dentistry, College of Medicine and Dentistry, Riyadh Elm University, Riyadh 13244, Saudi Arabia
| | - Mohammed M. Gad
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (M.M.G.)
| | - Mary Anne S. Melo
- Department of Comprehensive Dentistry, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Abdulrahman A. Balhaddad
- Department of Restorative Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
12
|
Li H, Shi Y, Chen H, Liang J, Zhang S, Li B, Chen J, Li M, Peng X, Zhou X, Ren B, Cheng L. A novel pH-responsive monomer inhibits Candida albicans via a dual antifungal mode of action. J Mater Chem B 2024; 12:10367-10382. [PMID: 39290132 DOI: 10.1039/d4tb00851k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The scarcity of the antifungal drug arsenal highlights an urgent need to develop alternative treatments for candidiasis caused by Candida albicans (C. albicans). As pH is closely associated with C. albicans infection, it could be an essential target in a novel approach for designing antifungal therapy. In this study, a novel intelligent antifungal monomer, dodecylmethylaminoethyl methacrylate (DMAEM), with a pH-responsive tertiary amine group and a methacrylate-derived CC double bond group is developed. It is uncovered that the two functional groups of DMAEM contribute to a dual mode of action. Under acidic pH, the tertiary amine of DMAEM protonates into a cationic fungicide, sharing similar structural and functional characteristics with quaternary ammonium salts, which exerts fungicidal activity by targeting the CHK1 two-component system in C. albicans. At neutral pH, the methacrylate-derived CC double bond group contributes to anti-virulence activity by blocking hyphal formation. In addition, it is also identified that DMAEM suppresses filamentation by altering the extracellular vesicles of C. albicans. These findings support that the novel intelligent pH-responsive monomer could be a therapeutic candidate for treating candidiasis.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hui Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jingou Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shiyong Zhang
- National Engineering Research Center for Biomaterials, and College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Bolei Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jing Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Xian Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
13
|
Vazquez-Munoz R, Thompson A, Sobue T, Dongari-Bagtzoglou A. Lactobacillus johnsonii is a dominant Lactobacillus in the murine oral mucosa and has chitinase activity that compromises fungal cell wall integrity. mBio 2024; 15:e0241624. [PMID: 39287438 PMCID: PMC11481578 DOI: 10.1128/mbio.02416-24] [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: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
The oral microbiome is a critical determinant of health and disease, as interactions between oral microorganisms can influence their physiology and the development or severity of oral infections. Lactobacilli have a widely recognized antagonistic relationship with Candida albicans and may exhibit probiotic properties that limit oral fungal infection. We previously reported that Lactobacillus johnsonii strain MT4, an oral strain isolated from C57BL/6 mice, can induce global changes in the murine oral microbiome and has anti-Candida activity in vitro. To build on this information, we analyzed its abundance on the mouse oral mucosa, tested its impact on the severity and progression of oropharyngeal candidiasis (OPC) in a mouse model, and further explored the mechanism of antifungal activity in vitro. Our findings reveal that L. johnsonii MT4 is a dominant cultivable Lactobacillus in the oral mucosa of C57BL/6 mice. Strain MT4 has chitinase activity against C. albicans, which damages the cell wall and compromises fungal metabolic activity. Oral inoculation with strain MT4 causes a reduction in the Candida-induced rise in the abundance of oral enterococci and oral mucosal damage. This research underscores the potential of L. johnsonii strain MT4 as a novel probiotic agent in the prevention or management of OPC, and it contributes to a better understanding of the role of oral bacterial microbiota role in the pathogenesis of fungal infections. IMPORTANCE The interactions between the opportunistic pathogen Candida albicans and resident oral bacteria are particularly crucial in maintaining oral health. Emerging antifungal drug-resistant strains, slow-paced drug discovery, and the risk of side effects can compromise the effectiveness of current treatments available for oropharyngeal candidiasis. This study advances the search for alternative microbiome-targeted therapies in oral fungal infections. We report that Lactobacillus johnsonii strain MT4 prevents the Candida-induced bloom of dysbiotic oral enterococci and reduces oral mucosal lesions in an oropharyngeal candidiasis murine model. We also show that this strain directly compromises the cell wall and reduces fungal metabolic activity, partly due to its chitinase activity.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Angela Thompson
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takanori Sobue
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Anna Dongari-Bagtzoglou
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
14
|
Xu T, Liu Y, Zhang W, Li M, Zhang L, Li X, Zhang Y, Yue L, Li S, Lin Y, Zou X, Chen F. Specific cell subclusters of dental pulp stem cells respond to distinct pathogens through the ROS pathway. Front Cell Infect Microbiol 2024; 14:1452124. [PMID: 39328360 PMCID: PMC11424553 DOI: 10.3389/fcimb.2024.1452124] [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: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024] Open
Abstract
Introduction Microbial pathogens invade various human organs, including the oral cavity. Candida albicans (C.a) and Streptococcus mutans (S.m) served respectively as representative oral pathogenic fungi and bacteria to stimulate dental pulp stem cells (DPSCs) and to screen the DPSC subcluster that specifically responded to fungal infection. Methods DPSCs were obtained from the impacted third molars of six healthy subjects. Then, cells were mixed and divided into three samples, two of which were stimulated with C.a and S.m, respectively; the third sample was exposed to cell medium only (Ctrl). Single-cell mRNA sequencing analysis of treated DPSCs was performed. Results DPSCs were composed of four major clusters of which one, DPSC.7, exhibited unique changes compared to those of other subclusters. The DPSC.7 cell percentage of the C.a sample was twice those of the Ctrl and S.m samples. DPSC.7 cells expressed genes associated with the response to reactive oxygen species (ROS) response. DPSC.7 subgroup cells established characteristic aggregation under the stimulation of different pathogens in UMAP. The MAPK/ERK1/2 and NF-κB pathways were up-regulated, DUSP1/5/6 expressions were suppressed, FOS synthesis was activated, the immune-related pathway was induced, and the levels of cytokines, including IL-6 and CCL2, were up-regulated in DPSC.7 cells when stimulated with C.a. Conclusions Our study analyzed the cellular and molecular properties of DPSCs infected by oral fungi and bacteria with single-cell RNA sequencing. A subcluster of DPSCs responded specifically to infections with different pathogens, activating the MAPK and NF-κB pathways to induce immune responses via the ROS pathway. This suggests novel treatment strategies for fungal infections.
Collapse
Affiliation(s)
- Tiansong Xu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Fifth Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yangjia Liu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Wen Zhang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Stomatology, Peking University International Hospital, Beijing, China
| | - Murong Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Liqi Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xueying Li
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yifei Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lin Yue
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Sha Li
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Ye Lin
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xiaoying Zou
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Center of Stomatology, Peking University Hospital, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
15
|
Luo G, Zhang J, Wang T, Cui H, Bai Y, Luo J, Zhang J, Zhang M, Di L, Yuan Y, Xiong K, Yu X, Zhang Y, Shen C, Zhu C, Wang Y, Su C, Lu Y. A human commensal-pathogenic fungus suppresses host immunity via targeting TBK1. Cell Host Microbe 2024; 32:1536-1551.e6. [PMID: 39084229 DOI: 10.1016/j.chom.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/11/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024]
Abstract
Candida albicans stably colonizes humans but is the leading cause of hospital-acquired fungemia. Traditionally, masking immunogenic moieties has been viewed as a tactic for immune evasion. Here, we demonstrate that C. albicans blocks type I interferon (IFN-I) signaling via translocating an effector protein Cmi1 into host cells. Mechanistically, Cmi1 binds and inhibits TANK-binding kinase 1 (TBK1) to abrogate IFN-regulatory factor 3 (IRF3) phosphorylation, thereby suppressing the IFN-I cascade. Murine infection with a cmi1 mutant displays an exaggerated IFN-I response in both kidneys and bone-marrow-derived macrophages, leading to rapid fungal clearance and host survival. Remarkably, the lack of CMI1 compromises gut commensalism and increases IFN-I response in mouse colonic cells. These phenotypes of cmi1 are rescued by the depletion of IFN-I receptor. This work establishes the importance of TBK1 inhibition in fungal pathogenesis and reveals that a human commensal-pathogenic fungus significantly impacts host immunity during gut colonization and infection via delivering effector proteins into host cells.
Collapse
Affiliation(s)
- Gang Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Jingkai Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Tianxu Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Hao Cui
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Yukun Bai
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Jianchen Luo
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Jinqiu Zhang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Mao Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Linyan Di
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Yuncong Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kang Xiong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiangtai Yu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yaling Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Chao Shen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Chang Su
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yang Lu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
16
|
Schaefer S, Corrigan N, Brunke S, Lenardon MD, Boyer C. Combatting Fungal Infections: Advances in Antifungal Polymeric Nanomaterials. Biomacromolecules 2024; 25:5670-5701. [PMID: 39177507 DOI: 10.1021/acs.biomac.4c00866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Fungal pathogens cause over 6.5 million life-threatening systemic infections annually, with mortality rates ranging from 20 to 95%, even with medical intervention. The World Health Organization has recently emphasized the urgent need for new antifungal drugs. However, the range of effective antifungal agents remains limited and resistance is increasing. This Review explores the current landscape of fungal infections and antifungal drugs, focusing on synthetic polymeric nanomaterials like nanoparticles that enhance the physicochemical properties of existing drugs. Additionally, we examine intrinsically antifungal polymers that mimic naturally occurring peptides. Advances in polymer characterization and synthesis now allow precise design and screening for antifungal activity, biocompatibility, and drug interactions. These antifungal polymers represent a promising new class of drugs for combating fungal infections.
Collapse
Affiliation(s)
- Sebastian Schaefer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales 2052, Australia
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
| | - Nathaniel Corrigan
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
| | - Megan D Lenardon
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| |
Collapse
|
17
|
Jia LJ, González K, Orasch T, Schmidt F, Brakhage AA. Manipulation of host phagocytosis by fungal pathogens and therapeutic opportunities. Nat Microbiol 2024; 9:2216-2231. [PMID: 39187614 DOI: 10.1038/s41564-024-01780-0] [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: 12/20/2023] [Accepted: 07/09/2024] [Indexed: 08/28/2024]
Abstract
An important host defence mechanism against pathogens is intracellular killing, which is achieved through phagocytosis, a cellular process for engulfing and neutralizing extracellular particles. Phagocytosis results in the formation of matured phagolysosomes, which are specialized compartments that provide a hostile environment and are considered the end point of the degradative pathway. However, all fungal pathogens studied to date have developed strategies to manipulate phagosomal function directly and also indirectly by redirecting phagosomes from the degradative pathway to a non-degradative pathway with the expulsion and even transfer of pathogens between cells. Here, using the major human fungal pathogens Aspergillus fumigatus, Candida albicans, Cryptococcus neoformans and Histoplasma capsulatum as examples, we discuss the processes involved in host phagosome-fungal pathogen interactions, with a focus on fungal evasion strategies. We also discuss recent approaches to targeting intraphagosomal pathogens, including the redirection of phagosomes towards degradative pathways for fungal pathogen eradication.
Collapse
Affiliation(s)
- Lei-Jie Jia
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany.
- Junior Research Group Phagosome Biology and Engineering, Leibniz-HKI, Jena, Germany.
| | - Katherine González
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Thomas Orasch
- Transfer Group Anti-infectives, Leibniz-HKI, Jena, Germany
| | - Franziska Schmidt
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany.
- Department of Microbiology and Molecular Biology, Institute of Microbiology, Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
18
|
Zhang LB, Yang WWJ, Yang ZH, Guan Y. N-acetylglucosamine kinase (BbHxk1) has pleiotropic effects on vegetative growth, cell wall integrity, morphological transition, cuticle infection, and metabolic modulation in the biological pesticide Beauveria bassiana. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:106015. [PMID: 39084806 DOI: 10.1016/j.pestbp.2024.106015] [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: 04/28/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
Beauveria bassiana is a popular and eco-friendly biopesticide. During its pathogen-pest interaction, both N-acetylglucosamine (GlcNAc) catabolism and anabolism are crucial for nutrient supply and cell-wall construction. The initiation of GlcNAc metabolism relies on the catalysis of GlcNAc kinase, which has been extensively studied in the human pathogen Candida albicans. However, the physiological function of GlcNAc kinase remains poorly understood in entomopathogenic fungi. In the present study, a GlcNAc kinase homolog was identified and designated as BbHxk1 in B. bassiana. Deletion of BbHxk1 resulted in viable but reduced vegetative growth on various carbon sources. ΔBbHxk1 mutants displayed severe defects in cell wall integrity, making them more susceptible to cell wall stress cues. Furthermore, the absence of BbHxk1 resulted in an increase in conidial yield and blastospore production, and a faster rate of germination and filamentation, potentially attributed to higher intracellular ATP levels. BbHxk1 deficiency led to a reduction in the activities of cuticle-degrading enzymes, which might contribute to the attenuated pathogenicity specifically through cuticle penetration rather than hemocoel infection towards Galleria mellonella larvae. Being different from C. albicans Hxk1, which facultatively acts as a catalyzing enzyme and transcriptional regulator, BbHxk1 primarily acts as a catalyzing enzyme and metabolic regulator. The altered metabolomic profiling correlated with the phenotypic defects in ΔBbHxk1 mutants, further implicating a potential metabolism-dependent mechanism of BbHxk1 in mediating physiologies of B. bassiana. These findings not only unveil a novel role for GlcNAc kinase in B. bassiana, but also provide a solid theoretical basis to guide metabolic reprogramming in order to maintain or even enhance the efficiency of fungi for practical applications.
Collapse
Affiliation(s)
- Long-Bin Zhang
- College of Biological Science and Engineering, Fuzhou University, Fujian, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China.
| | - Wu-Wei-Jie Yang
- College of Biological Science and Engineering, Fuzhou University, Fujian, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China
| | - Zhi-Hao Yang
- College of Biological Science and Engineering, Fuzhou University, Fujian, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China
| | - Yi Guan
- College of Biological Science and Engineering, Fuzhou University, Fujian, China; The Key Laboratory of Marine Enzyme Engineering of Fujian Province, Fuzhou University, Fujian, China.
| |
Collapse
|
19
|
Selisana SMG, Chen X, Mahfudhoh E, Bowolaksono A, Rozaliyani A, Orihara K, Kajiwara S. Alteration of β-glucan in the emerging fungal pathogen Candida auris leads to immune evasion and increased virulence. Med Microbiol Immunol 2024; 213:13. [PMID: 38967888 PMCID: PMC11226559 DOI: 10.1007/s00430-024-00795-y] [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: 01/17/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024]
Abstract
Candida auris is an emerging pathogenic yeast that has been categorized as a global public health threat and a critical priority among fungal pathogens. Despite this, the immune response against C. auris infection is still not well understood. Hosts fight Candida infections through the immune system that recognizes pathogen-associated molecular patterns such as β-glucan, mannan, and chitin on the fungal cell wall. In this study, levels of β-glucan and mannan exposures in C. auris grown under different physiologically relevant stimuli were quantified by flow cytometry-based analysis. Lactate, hypoxia, and sublethal concentration of fluconazole trigger a decrease in surface β-glucan while low pH triggers an increase in β-glucan. There is no inverse pattern between exposure levels of β-glucan and mannan in the cell wall architecture among the three clades. To determine the effect of cell wall remodeling on the immune response, a phagocytosis assay was performed, followed by quantification of released cytokines by ELISA. Lactate-induced decrease in β-glucan leads to reduced uptake of C. auris by PMA-differentiated THP-1 and RAW 264.7 macrophages. Furthermore, reduced production of CCL3/MIP-1⍺ but not TNF-⍺ and IL-10 were observed. An in vivo infection analysis using silkworms reveals that a reduction in β-glucan triggers an increase in the virulence of C. auris. This study demonstrates that β-glucan alteration occurs in C. auris and serves as an escape mechanism from immune cells leading to increased virulence.
Collapse
Affiliation(s)
- Shiela Marie Gines Selisana
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Xinyue Chen
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Eny Mahfudhoh
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Anom Bowolaksono
- Department of Biology, Faculty of Mathematics and Science, University of Indonesia, Depok, 16424, Indonesia
| | - Anna Rozaliyani
- Faculty of Medicine, University of Indonesia, Jakarta, 10430, Indonesia
| | - Kanami Orihara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Susumu Kajiwara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan.
| |
Collapse
|
20
|
Katsipoulaki M, Stappers MHT, Malavia-Jones D, Brunke S, Hube B, Gow NAR. Candida albicans and Candida glabrata: global priority pathogens. Microbiol Mol Biol Rev 2024; 88:e0002123. [PMID: 38832801 PMCID: PMC11332356 DOI: 10.1128/mmbr.00021-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
SUMMARYA significant increase in the incidence of Candida-mediated infections has been observed in the last decade, mainly due to rising numbers of susceptible individuals. Recently, the World Health Organization published its first fungal pathogen priority list, with Candida species listed in medium, high, and critical priority categories. This review is a synthesis of information and recent advances in our understanding of two of these species-Candida albicans and Candida glabrata. Of these, C. albicans is the most common cause of candidemia around the world and is categorized as a critical priority pathogen. C. glabrata is considered a high-priority pathogen and has become an increasingly important cause of candidemia in recent years. It is now the second most common causative agent of candidemia in many geographical regions. Despite their differences and phylogenetic divergence, they are successful as pathogens and commensals of humans. Both species can cause a broad variety of infections, ranging from superficial to potentially lethal systemic infections. While they share similarities in certain infection strategies, including tissue adhesion and invasion, they differ significantly in key aspects of their biology, interaction with immune cells, host damage strategies, and metabolic adaptations. Here we provide insights on key aspects of their biology, epidemiology, commensal and pathogenic lifestyles, interactions with the immune system, and antifungal resistance.
Collapse
Affiliation(s)
- Myrto Katsipoulaki
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Mark H. T. Stappers
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Dhara Malavia-Jones
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
21
|
Ma Q, Pradhan A, Leaves I, Hickey E, Roselletti E, Dambuza I, Larcombe DE, de Assis LJ, Wilson D, Erwig LP, Netea MG, Childers DS, Brown GD, Gow NA, Brown AJ. Impact of secreted glucanases upon the cell surface and fitness of Candida albicans during colonisation and infection. Cell Surf 2024; 11:100128. [PMID: 38938582 PMCID: PMC11208952 DOI: 10.1016/j.tcsw.2024.100128] [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: 04/04/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Host recognition of the pathogen-associated molecular pattern (PAMP), β-1,3-glucan, plays a major role in antifungal immunity. β-1,3-glucan is an essential component of the inner cell wall of the opportunistic pathogen Candida albicans. Most β-1,3-glucan is shielded by the outer cell wall layer of mannan fibrils, but some can become exposed at the cell surface. In response to host signals such as lactate, C. albicans shaves the exposed β-1,3-glucan from its cell surface, thereby reducing the ability of innate immune cells to recognise and kill the fungus. We have used sets of barcoded xog1 and eng1 mutants to compare the impacts of the secreted β-glucanases Xog1 and Eng1 upon C. albicans in vitro and in vivo. Flow cytometry of Fc-dectin-1-stained strains revealed that Eng1 plays the greater role in lactate-induced β-1,3-glucan masking. Transmission electron microscopy and stress assays showed that neither Eng1 nor Xog1 are essential for cell wall maintenance, but the inactivation of either enzyme compromised fungal adhesion to gut and vaginal epithelial cells. Competitive barcode sequencing suggested that neither Eng1 nor Xog1 strongly influence C. albicans fitness during systemic infection or vaginal colonisation in mice. However, the deletion of XOG1 enhanced C. albicans fitness during gut colonisation. We conclude that both Eng1 and Xog1 exert subtle effects on the C. albicans cell surface that influence fungal adhesion to host cells and that affect fungal colonisation in certain host niches.
Collapse
Affiliation(s)
- Qinxi Ma
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Arnab Pradhan
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Ian Leaves
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Emer Hickey
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Elena Roselletti
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Ivy Dambuza
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Daniel E. Larcombe
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Leandro Jose de Assis
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Duncan Wilson
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Lars P. Erwig
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Delma S. Childers
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Gordon D. Brown
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Neil A.R. Gow
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| | - Alistair J.P. Brown
- MRC Centre for Medical Mycology, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK
| |
Collapse
|
22
|
Avelar GM, Pradhan A, Ma Q, Hickey E, Leaves I, Liddle C, Rodriguez Rondon AV, Kaune AK, Shaw S, Maufrais C, Sertour N, Bain JM, Larcombe DE, de Assis LJ, Netea MG, Munro CA, Childers DS, Erwig LP, Brown GD, Gow NAR, Bougnoux ME, d'Enfert C, Brown AJP. A CO 2 sensing module modulates β-1,3-glucan exposure in Candida albicans. mBio 2024; 15:e0189823. [PMID: 38259065 PMCID: PMC10865862 DOI: 10.1128/mbio.01898-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Microbial species capable of co-existing with healthy individuals, such as the commensal fungus Candida albicans, exploit multifarious strategies to evade our immune defenses. These strategies include the masking of immunoinflammatory pathogen-associated molecular patterns (PAMPs) at their cell surface. We reported previously that C. albicans actively reduces the exposure of the proinflammatory PAMP, β-1,3-glucan, at its cell surface in response to host-related signals such as lactate and hypoxia. Here, we show that clinical isolates of C. albicans display phenotypic variability with respect to their lactate- and hypoxia-induced β-1,3-glucan masking. We have exploited this variability to identify responsive and non-responsive clinical isolates. We then performed RNA sequencing on these isolates to reveal genes whose expression patterns suggested potential association with lactate- or hypoxia-induced β-1,3-glucan masking. The deletion of two such genes attenuated masking: PHO84 and NCE103. We examined NCE103-related signaling further because NCE103 has been shown previously to encode carbonic anhydrase, which promotes adenylyl cyclase-protein kinase A (PKA) signaling at low CO2 levels. We show that while CO2 does not trigger β-1,3-glucan masking in C. albicans, the Sch9-Rca1-Nce103 signaling module strongly influences β-1,3-glucan exposure in response to hypoxia and lactate. In addition to identifying a new regulatory module that controls PAMP exposure in C. albicans, our data imply that this module is important for PKA signaling in response to environmental inputs other than CO2.IMPORTANCEOur innate immune defenses have evolved to protect us against microbial infection in part via receptor-mediated detection of "pathogen-associated molecular patterns" (PAMPs) expressed by invading microbes, which then triggers their immune clearance. Despite this surveillance, many microbial species are able to colonize healthy, immune-competent individuals, without causing infection. To do so, these microbes must evade immunity. The commensal fungus Candida albicans exploits a variety of strategies to evade immunity, one of which involves reducing the exposure of a proinflammatory PAMP (β-1,3-glucan) at its cell surface. Most of the β-1,3-glucan is located in the inner layer of the C. albicans cell wall, hidden by an outer layer of mannan fibrils. Nevertheless, some β-1,3-glucan can become exposed at the fungal cell surface. However, in response to certain specific host signals, such as lactate or hypoxia, C. albicans activates an anticipatory protective response that decreases β-1,3-glucan exposure, thereby reducing the susceptibility of the fungus to impending innate immune attack. Here, we exploited the natural phenotypic variability of C. albicans clinical isolates to identify strains that do not display the response to β-1,3-glucan masking signals observed for the reference isolate, SC5314. Then, using genome-wide transcriptional profiling, we compared these non-responsive isolates with responsive controls to identify genes potentially involved in β-1,3-glucan masking. Mutational analysis of these genes revealed that a sensing module that was previously associated with CO2 sensing also modulates β-1,3-glucan exposure in response to hypoxia and lactate in this major fungal pathogen of humans.
Collapse
Affiliation(s)
- Gabriela M. Avelar
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Arnab Pradhan
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Qinxi Ma
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Emer Hickey
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Ian Leaves
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Corin Liddle
- Bioimaging Unit, University of Exeter, Exeter, United Kingdom
| | - Alejandra V. Rodriguez Rondon
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Ann-Kristin Kaune
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Sophie Shaw
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Corinne Maufrais
- Institut Pasteur, Université Paris Cité, INRAe USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Natacha Sertour
- Institut Pasteur, Université Paris Cité, INRAe USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Judith M. Bain
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Daniel E. Larcombe
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Leandro J. de Assis
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Carol A. Munro
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Delma S. Childers
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Lars P. Erwig
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Johnson-Johnson Innovation, EMEA Innovation Centre, London, United Kingdom
| | - Gordon D. Brown
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Neil A. R. Gow
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Marie-Elisabeth Bougnoux
- Institut Pasteur, Université Paris Cité, INRAe USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
- Unité de Parasitologie-Mycologie, Service de Microbiologie Clinique, Hôpital Necker-Enfants-Malades, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
- Université Paris Cité, Paris, France
| | - Christophe d'Enfert
- Institut Pasteur, Université Paris Cité, INRAe USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Alistair J. P. Brown
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
23
|
Schaefer S, Melodia D, Pracey C, Corrigan N, Lenardon MD, Boyer C. Mimicking Charged Host-Defense Peptides to Tune the Antifungal Activity and Biocompatibility of Amphiphilic Polymers. Biomacromolecules 2024; 25:871-889. [PMID: 38165721 DOI: 10.1021/acs.biomac.3c01038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Invasive fungal infections impose a substantial global health burden. They cause more than 1.5 million deaths annually and are insufficiently met by the currently approved antifungal drugs. Antifungal peptides are a promising alternative to existing antifungal drugs; however, they can be challenging to synthesize, and are often susceptible to proteases in vivo. Synthetic polymers which mimic the properties of natural antifungal peptides can circumvent these limitations. In this study, we developed a library of 29 amphiphilic polyacrylamides with different charged units, namely, amines, guanidinium, imidazole, and carboxylic acid groups, representative of the natural amino acids lysine, arginine, histidine, and glutamic acid. Ternary polymers incorporating primary ammonium (lysine-like) or imidazole (histidine-like) groups demonstrated superior activity against Candida albicans and biocompatibility with mammalian cells compared to the polymers containing the other charged groups. Furthermore, a combination of primary ammonium, imidazole, and guanidinium (arginine-like) within the same polymer outperformed the antifungal drug amphotericin B in terms of therapeutic index and exhibited fast C. albicans-killing activity. The most promising polymer compositions showed synergistic effects in combination with caspofungin and fluconazole against C. albicans and additionally demonstrated activity against other clinically relevant fungi. Collectively, these results indicate the strong potential of these easily producible polymers to be used as antifungals.
Collapse
Affiliation(s)
- Sebastian Schaefer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales 2052, Australia
| | - Daniele Melodia
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Christopher Pracey
- Nuclear Magnetic Resonance Facility, Mark Wainwright Analytical Centre, UNSW, Sydney, New South Wales 2052, Australia
| | - Nathaniel Corrigan
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Megan D Lenardon
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| |
Collapse
|
24
|
Wang Y, Liu Z, Yin X, Liu S, Wang K, Wan R, Chen H, Li X, Huang B. Variation in Bombyx mori immune response against fungal pathogen Beauveria bassiana with variability in cell wall β-1,3-glucan. INSECT SCIENCE 2024; 31:211-224. [PMID: 37350124 DOI: 10.1111/1744-7917.13209] [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: 01/04/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 06/24/2023]
Abstract
Entomopathogenic fungi are protected by a cell wall with dynamic structure for adapting to various environmental conditions. β-1,3-Glucan recognition proteins activate the innate immune system of insects by recognizing surface molecules of fungi. However, the associations between pathogenicity and the different components of entomopathogenic fungal cell walls remain unclear. Three Beauveria bassiana strains were selected that have significantly differing virulence against Bombyx mori. The molecular mechanisms underlying the immune response in B. mori were investigated using RNA sequencing, which revealed differences in the immune response to different B. bassiana strains at 12 h post-infection. Immunofluorescence assays revealed that β-1,3-glucan content had an opposite trend to that of fungal virulence. β-1,3-Glucan injection upregulated BmβGRP4 expression and significantly reduced the virulence of the high-virulence strain but not that of the medium-virulence or low-virulence strains. BmβGRP4 silencing in B. mori with RNA interference resulted in the opposite virulence pattern, indicating that the virulence of B. bassiana was affected by the cell walls' content of β-1,3-glucan, which could be recognized by BmβGRP4. Furthermore, interference with the gene CnA (calcineurin catalytic A subunit) involved in β-1,3-glucan synthesis eliminated differences in virulence between B. bassiana strains. These results indicate that strains of a single species of pathogenic fungi that have differing cell wall components are recognized differently by the innate immune system of B. mori.
Collapse
Affiliation(s)
- Yulong Wang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Zhen Liu
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Xuebing Yin
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Shihong Liu
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Kai Wang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Rongjie Wan
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Haoran Chen
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Xinyang Li
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| | - Bo Huang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, China
| |
Collapse
|
25
|
Sobel JD, Vempati YS. Bacterial Vaginosis and Vulvovaginal Candidiasis Pathophysiologic Interrelationship. Microorganisms 2024; 12:108. [PMID: 38257934 PMCID: PMC10820109 DOI: 10.3390/microorganisms12010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Among the infectious causes of vulvovaginal symptoms, bacterial vaginosis (BV) and vulvovaginal candidiasis (VVC) dominate. Apart from infrequent mixed infections, both are considered independent and caused by unrelated pathogenic mechanisms. Clinical experience, however, is strongly suggestive that in some populations these infections are linked with recurrent BV (RBV) serving as the dominant etiopathogenic trigger for development of recurrent VVC (RVVC) with profound clinical and therapeutic consequences. The biologic basis for this critical interrelationship is discussed and suggests that as a consequence of BV dysbiosis, and not necessarily because of antibiotics prescribed, immune defenses are compromised, neutralizing vaginal yeast tolerance. The consequent BV-induced vaginal proinflammatory environment predisposes to mixed infection or consecutive episodes of post-treatment VVC. Recurrent BV and repeated antimicrobial drug exposure also predispose to acquired fluconazole resistance in C. albicans isolates, contributing to refractory vulvovaginal candidiasis.
Collapse
Affiliation(s)
- Jack D. Sobel
- C.S. Mott Center for Growth and Human Development, 275 E. Hancock St, Detroit, MI 48201, USA
| | - Yogitha Sai Vempati
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| |
Collapse
|
26
|
Sah SK, Yadav A, Kruppa MD, Rustchenko E. Identification of 10 genes on Candida albicans chromosome 5 that control surface exposure of the immunogenic cell wall epitope β-glucan and cell wall remodeling in caspofungin-adapted mutants. Microbiol Spectr 2023; 11:e0329523. [PMID: 37966256 PMCID: PMC10714753 DOI: 10.1128/spectrum.03295-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
IMPORTANCE Candida infections are often fatal in immuno-compromised individuals, resulting in many thousands of deaths per year. Caspofungin has proven to be an excellent anti-Candida drug and is now the frontline treatment for infections. However, as expected, the number of resistant cases is increasing; therefore, new treatment modalities are needed. We are determining metabolic pathways leading to decreased drug susceptibility in order to identify mechanisms facilitating evolution of clinical resistance. This study expands the understanding of genes that modulate drug susceptibility and reveals new targets for the development of novel antifungal drugs.
Collapse
Affiliation(s)
- Sudisht K. Sah
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| | - Anshuman Yadav
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael D. Kruppa
- Department of Biomedical Sciences, Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Elena Rustchenko
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
27
|
Miranda FS, Tavares VKF, Gomes MP, Neto NFA, Chiappim W, Petraconi G, Pessoa RS, Koga-Ito CY. Physicochemical Characteristics and Antimicrobial Efficacy of Plasma-Activated Water Produced by an Air-Operated Coaxial Dielectric Barrier Discharge Plasma. WATER 2023; 15:4045. [DOI: 10.3390/w15234045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
In this study, Plasma-Activated Water (PAW) was synthesized using a coaxial Dielectric Barrier Discharge (DBD) reactor, benefiting from the elevated capacity of air-flow-assisted DBD discharges to enhance nitrogen-based species concentration. By manipulating operational parameters, including gas flow rate, activation time, and DI water volume, we achieved significant concentrations of reactive oxygen and nitrogen species (RONS). As a result, the PAW obtained displayed pronounced physicochemical attributes: a pH of 2.06, an ORP of 275 mV, conductivity of 3 mS/cm, and TDS of 1200 mg/L. A pivotal aspect of this research was the evaluation of the reactor’s efficiency, as indicated by metrics like the specific input energy and ozone efficiency yield. The antimicrobial potential of the PAW was also assessed against pathogenic microbes, with remarkable reductions in viability for both Staphylococcus aureus and Escherichia coli (99.99%) and a more moderate decrease for Candida albicans (37%). These findings underscore the capability of coaxial DBD reactors in crafting high-quality PAW with significant antimicrobial properties, necessitating further studies to validate its broad-spectrum and safe applications.
Collapse
Affiliation(s)
- F. S. Miranda
- Department of Environment Engineering, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12247-016, Brazil
- Plasmas and Processes Laboratory, Department of Physics, Aeronautics Institute of Technology, Praça Marechal Eduardo Gomes 50, São José dos Campos 12228-900, Brazil
| | - V. K. F. Tavares
- Department of Environment Engineering, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12247-016, Brazil
| | - M. P. Gomes
- Plasmas and Processes Laboratory, Department of Physics, Aeronautics Institute of Technology, Praça Marechal Eduardo Gomes 50, São José dos Campos 12228-900, Brazil
| | - N. F. Azevedo Neto
- Department of Environment Engineering, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12247-016, Brazil
| | - W. Chiappim
- Laboratory of Plasmas and Applications, Department of Physics, Faculty of Engineering and Sciences, São Paulo State University (UNESP), Guaratinguetá 12516-410, Brazil
| | - G. Petraconi
- Plasmas and Processes Laboratory, Department of Physics, Aeronautics Institute of Technology, Praça Marechal Eduardo Gomes 50, São José dos Campos 12228-900, Brazil
| | - R. S. Pessoa
- Plasmas and Processes Laboratory, Department of Physics, Aeronautics Institute of Technology, Praça Marechal Eduardo Gomes 50, São José dos Campos 12228-900, Brazil
| | - C. Y. Koga-Ito
- Department of Environment Engineering, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12247-016, Brazil
- Science Applied to Oral Health, Graduate Program of Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, Brazil
| |
Collapse
|
28
|
Fernando LD, Pérez-Llano Y, Dickwella Widanage MC, Jacob A, Martínez-Ávila L, Lipton AS, Gunde-Cimerman N, Latgé JP, Batista-García RA, Wang T. Structural adaptation of fungal cell wall in hypersaline environment. Nat Commun 2023; 14:7082. [PMID: 37925437 PMCID: PMC10625518 DOI: 10.1038/s41467-023-42693-6] [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: 04/25/2023] [Accepted: 10/18/2023] [Indexed: 11/06/2023] Open
Abstract
Halophilic fungi thrive in hypersaline habitats and face a range of extreme conditions. These fungal species have gained considerable attention due to their potential applications in harsh industrial processes, such as bioremediation and fermentation under unfavorable conditions of hypersalinity, low water activity, and extreme pH. However, the role of the cell wall in surviving these environmental conditions remains unclear. Here we employ solid-state NMR spectroscopy to compare the cell wall architecture of Aspergillus sydowii across salinity gradients. Analyses of intact cells reveal that A. sydowii cell walls contain a rigid core comprising chitin, β-glucan, and chitosan, shielded by a surface shell composed of galactomannan and galactosaminogalactan. When exposed to hypersaline conditions, A. sydowii enhances chitin biosynthesis and incorporates α-glucan to create thick, stiff, and hydrophobic cell walls. Such structural rearrangements enable the fungus to adapt to both hypersaline and salt-deprived conditions, providing a robust mechanism for withstanding external stress. These molecular principles can aid in the optimization of halophilic strains for biotechnology applications.
Collapse
Affiliation(s)
- Liyanage D Fernando
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Yordanis Pérez-Llano
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Malitha C Dickwella Widanage
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Anand Jacob
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Liliana Martínez-Ávila
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Andrew S Lipton
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Jean-Paul Latgé
- Institute of Molecular Biology and Biotechnology, University of Crete, Heraklion, Greece
- Fungal Respiratory Infections Research Unit, University of Angers, Angers, France
| | | | - Tuo Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
29
|
Lorenzo-Villegas DL, Gohil NV, Lamo P, Gurajala S, Bagiu IC, Vulcanescu DD, Horhat FG, Sorop VB, Diaconu M, Sorop MI, Oprisoni A, Horhat RM, Susan M, MohanaSundaram A. Innovative Biosensing Approaches for Swift Identification of Candida Species, Intrusive Pathogenic Organisms. Life (Basel) 2023; 13:2099. [PMID: 37895480 PMCID: PMC10608220 DOI: 10.3390/life13102099] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Candida is the largest genus of medically significant fungi. Although most of its members are commensals, residing harmlessly in human bodies, some are opportunistic and dangerously invasive. These have the ability to cause severe nosocomial candidiasis and candidemia that affect the viscera and bloodstream. A prompt diagnosis will lead to a successful treatment modality. The smart solution of biosensing technologies for rapid and precise detection of Candida species has made remarkable progress. The development of point-of-care (POC) biosensor devices involves sensor precision down to pico-/femtogram level, cost-effectiveness, portability, rapidity, and user-friendliness. However, futuristic diagnostics will depend on exploiting technologies such as multiplexing for high-throughput screening, CRISPR, artificial intelligence (AI), neural networks, the Internet of Things (IoT), and cloud computing of medical databases. This review gives an insight into different biosensor technologies designed for the detection of medically significant Candida species, especially Candida albicans and C. auris, and their applications in the medical setting.
Collapse
Affiliation(s)
| | - Namra Vinay Gohil
- Department of Internal Medicne, Medical College Baroda, Vadodara 390001, India;
- Department of Internal Medicne, SSG Hospital Vadodara, Gotri, Vadodara 390021, India
| | - Paula Lamo
- Escuela Superior de Ingeniería y Tecnología, Universidad Internacional de La Rioja, 26006 Logroño, Spain;
| | - Swathi Gurajala
- College of Applied Medical Sciences in Jubail, Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Iulia Cristina Bagiu
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (D.D.V.); (F.G.H.)
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Dan Dumitru Vulcanescu
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (D.D.V.); (F.G.H.)
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Florin George Horhat
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (D.D.V.); (F.G.H.)
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Virgiliu Bogdan Sorop
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (V.B.S.); (M.D.)
| | - Mircea Diaconu
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (V.B.S.); (M.D.)
| | - Madalina Ioana Sorop
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Andrada Oprisoni
- Department of Pediatrics, Discipline of Pediatric Oncology and Hematology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Razvan Mihai Horhat
- Department of Conservative Dentistry and Endodontics, Faculty of Dental Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Monica Susan
- Centre for Preventive Medicine, Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - ArunSundar MohanaSundaram
- School of Pharmacy, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India;
| |
Collapse
|
30
|
Zhang S, Yu M, Zhang G, He G, Ji Y, Dong J, Zheng H, Qian L. Revealing the Control Mechanisms of pH on the Solution Properties of Chitin via Single-Molecule Studies. Molecules 2023; 28:6769. [PMID: 37836611 PMCID: PMC10574145 DOI: 10.3390/molecules28196769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Chitin is one of the most common polysaccharides and is abundant in the cell walls of fungi and the shells of insects and aquatic organisms as a skeleton. The mechanism of how chitin responds to pH is essential to the precise control of brewing and the design of smart chitin materials. However, this molecular mechanism remains a mystery. Results from single-molecule studies, including single-molecule force spectroscopy (SMFS), AFM imaging, and molecular dynamic (MD) simulations, have shown that the mechanical and conformational behaviors of chitin molecules show surprising pH responsiveness. This can be compared with how, in natural aqueous solutions, chitin tends to form a more relaxed spreading conformation and show considerable elasticity under low stretching forces in acidic conditions. However, its molecular chain collapses into a rigid globule in alkaline solutions. The results show that the chain state of chitin can be regulated by the proportions of inter- and intramolecular H-bonds, which are determined via the number of water bridges on the chain under different pH values. This basic study may be helpful for understanding the cellular activities of fungi under pH stress and the design of chitin-based drug carriers.
Collapse
Affiliation(s)
- Song Zhang
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564502, China; (S.Z.); (G.Z.); (G.H.); (Y.J.); (J.D.)
| | - Miao Yu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China;
| | - Guoqiang Zhang
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564502, China; (S.Z.); (G.Z.); (G.H.); (Y.J.); (J.D.)
| | - Guanmei He
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564502, China; (S.Z.); (G.Z.); (G.H.); (Y.J.); (J.D.)
| | - Yunxu Ji
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564502, China; (S.Z.); (G.Z.); (G.H.); (Y.J.); (J.D.)
| | - Juan Dong
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564502, China; (S.Z.); (G.Z.); (G.H.); (Y.J.); (J.D.)
| | - Huayan Zheng
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564502, China; (S.Z.); (G.Z.); (G.H.); (Y.J.); (J.D.)
| | - Lu Qian
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
31
|
Anaya EU, Amin AE, Wester MJ, Danielson ME, Michel KS, Neumann AK. Dectin-1 multimerization and signaling depends on fungal β-glucan structure and exposure. Biophys J 2023; 122:3749-3767. [PMID: 37515324 PMCID: PMC10541497 DOI: 10.1016/j.bpj.2023.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023] Open
Abstract
Dectin-1A is a C-type lectin innate immunoreceptor that recognizes β-(1,3;1,6)-glucan, a structural component of Candida species cell walls. β-Glucans can adopt solution structures ranging from random coil to insoluble fiber due to tertiary (helical) and quaternary structure. Fungal β-glucans of medium and high molecular weight are highly structured, but low molecular weight glucan is much less structured. Despite similar affinity for Dectin-1, the ability of glucans to induce Dectin-1A-mediated signaling correlates with degree of structure. Glucan denaturation experiments showed that glucan structure determines agonistic potential, but not receptor binding affinity. We explored the impact of glucan structure on molecular aggregation of Dectin-1A. Stimulation with glucan signaling decreased Dectin-1A diffusion coefficient. Fluorescence measurements provided direct evidence of ligation-induced Dectin-1A aggregation, which positively correlated with increasing glucan structure content. In contrast, Dectin-1A is predominantly in a low aggregation state in resting cells. Molecular aggregates formed during interaction with highly structured, agonistic glucans did not exceed relatively small (<15 nm) clusters of a few engaged receptors. Finally, we observed increased molecular aggregation of Dectin-1A at fungal particle contact sites in a manner that positively correlated with the degree of exposed glucan on the particle surface. These results indicate that Dectin-1A senses the solution conformation of β-glucans through their varying ability to drive receptor dimer/oligomer formation and activation of membrane proximal signaling events.
Collapse
Affiliation(s)
- Eduardo U Anaya
- Department of Pathology, University of New Mexico, School of Medicine, Albuquerque, New Mexico
| | - Akram Etemadi Amin
- Department of Pathology, University of New Mexico, School of Medicine, Albuquerque, New Mexico; Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico
| | - Michael J Wester
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, New Mexico
| | | | | | - Aaron K Neumann
- Department of Pathology, University of New Mexico, School of Medicine, Albuquerque, New Mexico.
| |
Collapse
|
32
|
Choy HL, Gaylord EA, Doering TL. Ergosterol distribution controls surface structure formation and fungal pathogenicity. mBio 2023; 14:e0135323. [PMID: 37409809 PMCID: PMC10470819 DOI: 10.1128/mbio.01353-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 07/07/2023] Open
Abstract
Ergosterol, the major sterol in fungal membranes, is critical for defining membrane fluidity and regulating cellular processes. Although ergosterol synthesis has been well defined in model yeast, little is known about sterol organization in the context of fungal pathogenesis. We identified a retrograde sterol transporter, Ysp2, in the opportunistic fungal pathogen Cryptococcus neoformans. We found that the lack of Ysp2 under host-mimicking conditions leads to abnormal accumulation of ergosterol at the plasma membrane, invagination of the plasma membrane, and malformation of the cell wall, which can be functionally rescued by inhibiting ergosterol synthesis with the antifungal drug fluconazole. We also observed that cells lacking Ysp2 mislocalize the cell surface protein Pma1 and have abnormally thin and permeable capsules. As a result of perturbed ergosterol distribution and its consequences, ysp2∆ cells cannot survive in physiologically relevant environments such as host phagocytes and are dramatically attenuated in virulence. These findings expand our knowledge of cryptococcal biology and underscore the importance of sterol homeostasis in fungal pathogenesis. IMPORTANCE Cryptococcus neoformans is an opportunistic fungal pathogen that kills over 100,000 people worldwide each year. Only three drugs are available to treat cryptococcosis, and these are variously limited by toxicity, availability, cost, and resistance. Ergosterol is the most abundant sterol in fungi and a key component in modulating membrane behavior. Two of the drugs used for cryptococcal infection, amphotericin B and fluconazole, target this lipid and its synthesis, highlighting its importance as a therapeutic target. We discovered a cryptococcal ergosterol transporter, Ysp2, and demonstrated its key roles in multiple aspects of cryptococcal biology and pathogenesis. These studies demonstrate the role of ergosterol homeostasis in C. neoformans virulence, deepen our understanding of a pathway with proven therapeutic importance, and open a new area of study.
Collapse
Affiliation(s)
- Hau Lam Choy
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth A. Gaylord
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Wagner AS, Lumsdaine SW, Mangrum MM, Reynolds TB. Caspofungin-induced β(1,3)-glucan exposure in Candida albicans is driven by increased chitin levels. mBio 2023; 14:e0007423. [PMID: 37377417 PMCID: PMC10470516 DOI: 10.1128/mbio.00074-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/04/2023] [Indexed: 06/29/2023] Open
Abstract
To successfully induce disease, Candida albicans must effectively evade the host immune system. One mechanism used by C. albicans to achieve this is to mask immunogenic β(1,3)-glucan epitopes within its cell wall under an outer layer of mannosylated glycoproteins. Consequently, induction of β(1,3)-glucan exposure (unmasking) via genetic or chemical manipulation increases fungal recognition by host immune cells in vitro and attenuates disease during systemic infection in mice. Treatment with the echinocandin caspofungin is one of the most potent drivers of β(1,3)-glucan exposure. Several reports using murine infection models suggest a role for the immune system, and specifically host β(1,3)-glucan receptors, in mediating the efficacy of echinocandin treatment in vivo. However, the mechanism by which caspofungin-induced unmasking occurs is not well understood. In this report, we show that foci of unmasking co-localize with areas of increased chitin within the yeast cell wall in response to caspofungin, and that inhibition of chitin synthesis via nikkomycin Z attenuates caspofungin-induced β(1,3)-glucan exposure. Furthermore, we find that both the calcineurin and Mkc1 mitogen-activated protein kinase pathways work synergistically to regulate β(1,3)-glucan exposure and chitin synthesis in response to drug treatment. When either of these pathways are interrupted, it results in a bimodal population of cells containing either high or low chitin content. Importantly, increased unmasking correlates with increased chitin content within these cells. Microscopy further indicates that caspofungin-induced unmasking correlates with actively growing cells. Collectively, our work presents a model in which chitin synthesis induces unmasking within the cell wall in response to caspofungin in growing cells. IMPORTANCE Systemic candidiasis has reported mortality rates ranging from 20% to 40%. The echinocandins, including caspofungin, are first-line antifungals used to treat systemic candidiasis. However, studies in mice have shown that echinocandin efficacy relies on both its cidal impacts on Candida albicans, as well as a functional immune system to successfully clear invading fungi. In addition to direct C. albicans killing, caspofungin increases exposure (unmasking) of immunogenic β(1,3)-glucan moieties. To evade immune detection, β(1,3)-glucan is normally masked within the C. albicans cell wall. Consequently, unmasked β(1,3)-glucan renders these cells more visible to the host immune system and attenuates disease progression. Therefore, discovery of how caspofungin-induced unmasking occurs is needed to elucidate how the drug facilitates host immune system-mediated clearance in vivo. We report a strong and consistent correlation between chitin deposition and unmasking in response to caspofungin and propose a model in which altered chitin synthesis drives increased unmasking during drug exposure.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | | | - Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
34
|
Larcombe DE, Bohovych IM, Pradhan A, Ma Q, Hickey E, Leaves I, Cameron G, Avelar GM, de Assis LJ, Childers DS, Bain JM, Lagree K, Mitchell AP, Netea MG, Erwig LP, Gow NAR, Brown AJP. Glucose-enhanced oxidative stress resistance-A protective anticipatory response that enhances the fitness of Candida albicans during systemic infection. PLoS Pathog 2023; 19:e1011505. [PMID: 37428810 PMCID: PMC10358912 DOI: 10.1371/journal.ppat.1011505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/20/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023] Open
Abstract
Most microbes have developed responses that protect them against stresses relevant to their niches. Some that inhabit reasonably predictable environments have evolved anticipatory responses that protect against impending stresses that are likely to be encountered in their niches-termed "adaptive prediction". Unlike yeasts such as Saccharomyces cerevisiae, Kluyveromyces lactis and Yarrowia lipolytica and other pathogenic Candida species we examined, the major fungal pathogen of humans, Candida albicans, activates an oxidative stress response following exposure to physiological glucose levels before an oxidative stress is even encountered. Why? Using competition assays with isogenic barcoded strains, we show that "glucose-enhanced oxidative stress resistance" phenotype enhances the fitness of C. albicans during neutrophil attack and during systemic infection in mice. This anticipatory response is dependent on glucose signalling rather than glucose metabolism. Our analysis of C. albicans signalling mutants reveals that the phenotype is not dependent on the sugar receptor repressor pathway, but is modulated by the glucose repression pathway and down-regulated by the cyclic AMP-protein kinase A pathway. Changes in catalase or glutathione levels do not correlate with the phenotype, but resistance to hydrogen peroxide is dependent on glucose-enhanced trehalose accumulation. The data suggest that the evolution of this anticipatory response has involved the recruitment of conserved signalling pathways and downstream cellular responses, and that this phenotype protects C. albicans from innate immune killing, thereby promoting the fitness of C. albicans in host niches.
Collapse
Affiliation(s)
- Daniel E. Larcombe
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Iryna M. Bohovych
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Arnab Pradhan
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Qinxi Ma
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Emer Hickey
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Ian Leaves
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Gary Cameron
- Rowett Institute, School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Gabriela M. Avelar
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Leandro J. de Assis
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Delma S. Childers
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Judith M. Bain
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Katherine Lagree
- Department of Microbiology, Biosciences Building, University of Georgia, Athens, Georgia, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, Biosciences Building, University of Georgia, Athens, Georgia, United States of America
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Lars P. Erwig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Johnson-Johnson Innovation, EMEA Innovation Centre, One Chapel Place, London, United Kingdom
| | - Neil A. R. Gow
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| | - Alistair J. P. Brown
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Medical Research Council Centre for Medical Mycology, University of Exeter, School of Biosciences, Geoffrey Pope Building, Exeter, United Kingdom
| |
Collapse
|
35
|
Wu H, Wang L, Wang W, Shao Z, Jia X, Xiao H, Chen J. The secretory Candida effector Sce1 licenses fungal virulence by masking the immunogenic β-1,3-glucan and promoting apoptosis of the host cells. MLIFE 2023; 2:159-177. [PMID: 38817625 PMCID: PMC10989805 DOI: 10.1002/mlf2.12066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 06/01/2024]
Abstract
Candida albicans deploys a variety of mechanisms such as morphological switch and elicitor release to promote virulence. However, the intricate interactions between the fungus and the host remain poorly understood, and a comprehensive inventory of fungal virulence factors has yet to be established. In this study, we identified a C. albicans secretory effector protein Sce1, whose induction and secretion are associated with vagina-simulative conditions and chlamydospore formation. Sequence alignment showed that Sce1 belongs to a Pir family in C. albicans, which is conserved across several fungi and primarily characterized as a β-glucan binding protein in the Saccharomyces cerevisiae. Mechanically, Sce1 is primarily localized to the cell wall in a cleaved form as an alkali-labile β-1,3-glucan binding protein and plays a role in masking β-glucan in acidic environments and chlamydospores, a feature that might underline C. albicans' ability to evade host immunity. Further, a cleaved short form of Sce1 protein could be released into extracellular compartments and presented in bone marrow-derived macrophages infected with chlamydospores. This cleaved short form of Sce1 also demonstrated a unique ability to trigger the caspases-8/9-dependent apoptosis in various host cells. Correspondingly, genetic deletion of SCE1 led to dampened vaginal colonization of C. albicans and diminished fungal virulence during systemic infection. The discovery of Sce1 as a versatile virulence effector that executes at various compartments sheds light on the fungus-host interactions and C. albicans pathogenesis.
Collapse
Affiliation(s)
- Hongyu Wu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| | - Li Wang
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Wenjuan Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| | - Zhugui Shao
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
- Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Biomedical SciencesShandong UniversityJinanChina
| | - Xin‐Ming Jia
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Hui Xiao
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| | - Jiangye Chen
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
36
|
Upadhya R, Lam WC, Hole CR, Vasselli JG, Lodge JK. Cell wall composition in Cryptococcus neoformans is media dependent and alters host response, inducing protective immunity. FRONTIERS IN FUNGAL BIOLOGY 2023; 4:1183291. [PMID: 37538303 PMCID: PMC10399910 DOI: 10.3389/ffunb.2023.1183291] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Introduction Cryptococcus neoformans is a basidiomycete fungus that can cause meningoencephalitis, especially in immunocompromised patients. Cryptococcus grows in many different media, although little attention has been paid to the role of growth conditions on the cryptococcal cell wall or on virulence. Objective The purpose of this study was to determine how different media influenced the amount of chitin and chitosan in the cell wall, which in turn impacted the cell wall architecture and host response. Methods Yeast extract, peptone, and dextrose (YPD) and yeast nitrogen base (YNB) are two commonly used media for growing Cryptococcus before use in in vitro or in vivo experiments. As a result, C. neoformans was grown in either YPD or YNB, which were either left unbuffered or buffered to pH 7 with MOPS. These cells were then labeled with cell wall-specific fluorescent probes to determine the amounts of various cell wall components. In addition, these cells were employed in animal virulence studies using the murine inhalation model of infection. Results We observed that the growth of wild-type C. neoformans KN99 significantly changes the pH of unbuffered media during growth. It raises the pH to 8.0 when grown in unbuffered YPD but lowers the pH to 2.0 when grown in unbuffered YNB (YNB-U). Importantly, the composition of the cell wall was substantially impacted by growth in different media. Cells grown in YNB-U exhibited a 90% reduction in chitosan, the deacetylated form of chitin, compared with cells grown in YPD. The decrease in pH and chitosan in the YNB-U-grown cells was associated with a significant increase in some pathogen-associated molecular patterns on the surface of cells compared with cells grown in YPD or YNB, pH 7. This altered cell wall architecture resulted in a significant reduction in virulence when tested using a murine model of infection. Furthermore, when heat-killed cells were used as the inoculum, KN99 cells grown in YNB-U caused an aberrant hyper-inflammatory response in the lungs, resulting in rapid animal death. In contrast, heat-killed KN99 cells grown in YNB, pH 7, caused little to no inflammatory response in the host lung, but, when used as a vaccine, they conferred a robust protective response against a subsequent challenge infection with the virulent KN99 cells. Conclusion These findings emphasize the importance of culture media and pH during growth in shaping the content and organization of the C. neoformans cell wall, as well as their impact on fungal virulence and the host response.
Collapse
Affiliation(s)
- Rajendra Upadhya
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Woei C. Lam
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Camaron R. Hole
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Joseph G. Vasselli
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Jennifer K. Lodge
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
37
|
Becker M, Sobel R. Vulvovaginal Candidiasis in Postmenopausal Women. Curr Infect Dis Rep 2023. [DOI: 10.1007/s11908-023-00801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
38
|
Derkacz D, Krasowska A. Alterations in the Level of Ergosterol in Candida albicans' Plasma Membrane Correspond with Changes in Virulence and Result in Triggering Diversed Inflammatory Response. Int J Mol Sci 2023; 24:ijms24043966. [PMID: 36835379 PMCID: PMC9964392 DOI: 10.3390/ijms24043966] [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: 01/28/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Opportunistic pathogen Candida albicans possesses multiple virulence factors which enable colonization and infection of host tissues. Candida-related infections frequently occur in immunocompromised patients, which is related to an insufficient inflammatory response. Furthermore, immunosuppression and multidrug resistance of C. albicans clinical isolates make the treatment of candidiasis a challenge for modern medicine. The most common resistance mechanism of C. albicans to antifungals includes point mutations in the ERG11 gene, which encodes target protein for azoles. We investigated whether the mutations or deletion of the ERG11 gene influence the pathogen-host interactions. We prove that both C. albicans erg11∆/∆ and ERG11K143R/K143R exhibit increased cell surface hydrophobicity. Additionally, C. albicans KS058 has an impaired ability of biofilm and hyphae formation. Analysis of the inflammatory response of human dermal fibroblasts and vaginal epithelial cell lines revealed that altered morphology of C. albicans erg11∆/∆ results in a significantly weaker immune response. C. albicans ERG11K143R/K143R triggered stronger production of pro-inflammatory response. Analysis of genes encoding adhesins confirmed differences in the expression pattern of key adhesins for both erg11∆/∆ and ERG11K143R/K143R strains. Obtained data indicate that alterations in Erg11p consequence in resistance to azoles and affect the key virulence factors and inflammatory response of host cells.
Collapse
|
39
|
Brown AJP. Fungal resilience and host-pathogen interactions: Future perspectives and opportunities. Parasite Immunol 2023; 45:e12946. [PMID: 35962618 PMCID: PMC10078341 DOI: 10.1111/pim.12946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/31/2023]
Abstract
We are constantly exposed to the threat of fungal infection. The outcome-clearance, commensalism or infection-depends largely on the ability of our innate immune defences to clear infecting fungal cells versus the success of the fungus in mounting compensatory adaptive responses. As each seeks to gain advantage during these skirmishes, the interactions between host and fungal pathogen are complex and dynamic. Nevertheless, simply compromising the physiological robustness of fungal pathogens reduces their ability to evade antifungal immunity, their virulence, and their tolerance against antifungal therapy. In this article I argue that this physiological robustness is based on a 'Resilience Network' which mechanistically links and controls fungal growth, metabolism, stress resistance and drug tolerance. The elasticity of this network probably underlies the phenotypic variability of fungal isolates and the heterogeneity of individual cells within clonal populations. Consequently, I suggest that the definition of the fungal Resilience Network represents an important goal for the future which offers the clear potential to reveal drug targets that compromise drug tolerance and synergise with current antifungal therapies.
Collapse
Affiliation(s)
- Alistair J P Brown
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, UK
| |
Collapse
|
40
|
de Assis LJ, Bain JM, Liddle C, Leaves I, Hacker C, Peres da Silva R, Yuecel R, Bebes A, Stead D, Childers DS, Pradhan A, Mackenzie K, Lagree K, Larcombe DE, Ma Q, Avelar GM, Netea MG, Erwig LP, Mitchell AP, Brown GD, Gow NAR, Brown AJP. Nature of β-1,3-Glucan-Exposing Features on Candida albicans Cell Wall and Their Modulation. mBio 2022; 13:e0260522. [PMID: 36218369 PMCID: PMC9765427 DOI: 10.1128/mbio.02605-22] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 01/15/2023] Open
Abstract
Candida albicans exists as a commensal of mucosal surfaces and the gastrointestinal tract without causing pathology. However, this fungus is also a common cause of mucosal and systemic infections when antifungal immune defenses become compromised. The activation of antifungal host defenses depends on the recognition of fungal pathogen-associated molecular patterns (PAMPs), such as β-1,3-glucan. In C. albicans, most β-1,3-glucan is present in the inner cell wall, concealed by the outer mannan layer, but some β-1,3-glucan becomes exposed at the cell surface. In response to host signals, such as lactate, C. albicans induces the Xog1 exoglucanase, which shaves exposed β-1,3-glucan from the cell surface, thereby reducing phagocytic recognition. We show here that β-1,3-glucan is exposed at bud scars and punctate foci on the lateral wall of yeast cells, that this exposed β-1,3-glucan is targeted during phagocytic attack, and that lactate-induced masking reduces β-1,3-glucan exposure at bud scars and at punctate foci. β-1,3-Glucan masking depends upon protein kinase A (PKA) signaling. We reveal that inactivating PKA, or its conserved downstream effectors, Sin3 and Mig1/Mig2, affects the amounts of the Xog1 and Eng1 glucanases in the C. albicans secretome and modulates β-1,3-glucan exposure. Furthermore, perturbing PKA, Sin3, or Mig1/Mig2 attenuates the virulence of lactate-exposed C. albicans cells in Galleria. Taken together, the data are consistent with the idea that β-1,3-glucan masking contributes to Candida pathogenicity. IMPORTANCE Microbes that coexist with humans have evolved ways of avoiding or evading our immunological defenses. These include the masking by these microbes of their "pathogen-associated molecular patterns" (PAMPs), which are recognized as "foreign" and used to activate protective immunity. The commensal fungus Candida albicans masks the proinflammatory PAMP β-1,3-glucan, which is an essential component of its cell wall. Most of this β-1,3-glucan is hidden beneath an outer layer of the cell wall on these microbes, but some can become exposed at the fungal cell surface. Using high-resolution confocal microscopy, we examine the nature of the exposed β-1,3-glucan at C. albicans bud scars and at punctate foci on the lateral cell wall, and we show that these features are targeted by innate immune cells. We also reveal that downstream effectors of protein kinase A (Mig1/Mig2, Sin3) regulate the secretion of major glucanases, modulate the levels of β-1,3-glucan exposure, and influence the virulence of C. albicans in an invertebrate model of systemic infection. Our data support the view that β-1,3-glucan masking contributes to immune evasion and the virulence of a major fungal pathogen of humans.
Collapse
Affiliation(s)
- Leandro José de Assis
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Judith M. Bain
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Corin Liddle
- Bioimaging Unit, University of Exeter, Exeter, United Kingdom
| | - Ian Leaves
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | | | - Roberta Peres da Silva
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Raif Yuecel
- Exeter Centre for Cytomics, University of Exeter, Exeter, United Kingdom
| | - Attila Bebes
- Exeter Centre for Cytomics, University of Exeter, Exeter, United Kingdom
| | - David Stead
- Aberdeen Proteomics Facility, Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Delma S. Childers
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Arnab Pradhan
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Kevin Mackenzie
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Katherine Lagree
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Daniel E. Larcombe
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Qinxi Ma
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Gabriela Mol Avelar
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Lars P. Erwig
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Johnson-Johnson Innovation, EMEA Innovation Centre, London, United Kingdom
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Gordon D. Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Neil A. R. Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Alistair J. P. Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
41
|
Hall RA, Wallace EW. Post-transcriptional control of fungal cell wall synthesis. Cell Surf 2022; 8:100074. [PMID: 35097244 PMCID: PMC8783092 DOI: 10.1016/j.tcsw.2022.100074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
Pathogenic fungi hide from their hosts by camouflage, obscuring immunogenic cell wall components such as beta-glucan with innocuous coverings such as mannoproteins and alpha-glucan that are less readily recognised by the host. Attempts to understand how such processes are regulated have met with varying success. Typically studies focus on understanding the transcriptional response of fungi to either their reservoir environment or the host. However, such approaches do not fully address this research question, due to the layers of post-transcriptional and post-translational regulation that occur within a cell. Although in animals the impact of post-transcriptional and post-translational regulation has been well characterised, our knowledge of these processes in the fungal kingdom is more limited. Mutations in RNA-binding proteins, like Ssd1 and Candida albicans Slr1, affect cell wall composition and fungal virulence indicating that post-transcriptional regulation plays a key role in these processes. Here, we review the current state of knowledge of fungal post-transcriptional regulation, and link this to potential mechanisms of immune evasion by drawing on studies from model yeast and plant pathogenic fungi. We highlight several RNA-binding proteins that regulate cell wall synthesis and could be involved in local translation of cell wall components. Expanding our knowledge on post-transcriptional regulation in human fungal pathogens is essential to fully comprehend fungal virulence strategies and for the design of novel antifungal therapies.
Collapse
Affiliation(s)
- Rebecca A. Hall
- Kent Fungal Group, Division of Natural Sciences, School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom
| | - Edward W.J. Wallace
- Institute for Cell Biology and SynthSys, School of Biological Sciences, University of Edinburgh, EH9 3FF, United Kingdom
| |
Collapse
|
42
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
43
|
Farooq H, Monowar T, Latt SS, Chinni SV, Zainol NH, Sabesan GS. A Review of Studies on Candida Species in Peninsular Malaysia. Adv Biomed Res 2022; 11:98. [PMID: 36660752 PMCID: PMC9843594 DOI: 10.4103/abr.abr_3_22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/09/2022] [Accepted: 06/16/2022] [Indexed: 01/21/2023] Open
Abstract
Data on the epidemiology and the antifungal susceptibility of Candida species infections in Malaysia is still limited. The study aimed to review and compare studies reporting the prevalence of Candida species and antifungal susceptibility of Candida infections in Peninsular Malaysia. Data from 22 studies published between 2009 and 2018. Data was collected using National Center for Biotechnology Information and Google Scholar using the keywords "Candida and Malaysia." Around 19 Candida species were identified in a total of 35,608 Candida isolates analyzed in these studies. In most studies examined, C. albicans (66.3%) was the predominant species, followed by C. glabrata (11.7%), C. parapsilosis (10.7%), C. tropicalis (9.5%), and C. krusei (1.19%). Vaginal swabs yielded the most isolates, followed by the respiratory system, urine, blood, oral, pus, and other locations. The demographic, racial, and gender data were recorded only in two studies. Totally, eight studies examined 396 isolates for antifungal susceptibility to common antifungal medications. The average antifungal susceptibility of isolates and efficacy of drugs in these studies ranged between 45 and 99% for most common antifungal drugs. Caspofungin had the highest susceptibility at 99%, whereas itraconazole had the lowest at only 45%. Overall, this review provided a comprehensive summary of all the current research on predominant Candida species in Peninsular Malaysia.
Collapse
Affiliation(s)
- Humaira Farooq
- Faculty of Medicine, AIMST University, Malaysia,Address for correspondence: Miss. Humaira Farooq, AIMST University, Faculty of Medicine, Department of Medical Microbiology, Batu 3 1/2, Bukit Air Nasi, Jalan Bedong—Semeling, 08100 Bedong, Kedah, Malaysia. E-mail:
| | | | - Swe Swe Latt
- Faculty of Medicine, RCSI and UCD Malaysia Campus, Penang, Malaysia
| | - Suresh V. Chinni
- Faculty of Medicine, Bioscience and Nursing MAHASA University, Selangor, Malaysia
| | | | | |
Collapse
|
44
|
Valand N, Brunt E, Gazioglu O, Yesilkaya H, Mitchell D, Horley N, Arroo R, Kishore U, Wallis R, Girija UV. Inactivation of the Complement Lectin Pathway by Candida tropicalis Secreted Aspartyl Protease-1. Immunobiology 2022; 227:152263. [DOI: 10.1016/j.imbio.2022.152263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/16/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022]
|
45
|
Avelar GM, Dambuza IM, Ricci L, Yuecel R, Mackenzie K, Childers DS, Bain JM, Pradhan A, Larcombe DE, Netea MG, Erwig LP, Brown GD, Duncan SH, Gow NA, Walker AW, Brown AJ. Impact of changes at the Candida albicans cell surface upon immunogenicity and colonisation in the gastrointestinal tract. CELL SURFACE (AMSTERDAM, NETHERLANDS) 2022; 8:100084. [PMID: 36299406 PMCID: PMC9589014 DOI: 10.1016/j.tcsw.2022.100084] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022]
Abstract
The immunogenicity of Candida albicans cells is influenced by changes in the exposure of microbe-associated molecular patterns (MAMPs) on the fungal cell surface. Previously, the degree of exposure on the C. albicans cell surface of the immunoinflammatory MAMP β-(1,3)-glucan was shown to correlate inversely with colonisation levels in the gastrointestinal (GI) tract. This is important because life-threatening systemic candidiasis in critically ill patients often arises from translocation of C. albicans strains present in the patient's GI tract. Therefore, using a murine model, we have examined the impact of gut-related factors upon β-glucan exposure and colonisation levels in the GI tract. The degree of β-glucan exposure was examined by imaging flow cytometry of C. albicans cells taken directly from GI compartments, and compared with colonisation levels. Fungal β-glucan exposure was lower in the cecum than the small intestine, and fungal burdens were correspondingly higher in the cecum. This inverse correlation did not hold for the large intestine. The gut fermentation acid, lactate, triggers β-glucan masking in vitro, leading to attenuated anti-Candida immune responses. Additional fermentation acids are present in the GI tract, including acetate, propionate, and butyrate. We show that these acids also influence β-glucan exposure on C. albicans cells in vitro and, like lactate, they influence β-glucan exposure via Gpr1/Gpa2-mediated signalling. Significantly, C. albicans gpr1Δ gpa2Δ cells displayed elevated β-glucan exposure in the large intestine and a corresponding decrease in fungal burden, consistent with the idea that Gpr1/Gpa2-mediated β-glucan masking influences colonisation of this GI compartment. Finally, extracts from the murine gut and culture supernatants from the mannan grazing gut anaerobe Bacteroides thetaiotaomicron promote β-glucan exposure at the C. albicans cell surface. Therefore, the local microbiota influences β-glucan exposure levels directly (via mannan grazing) and indirectly (via fermentation acids), whilst β-glucan masking appears to promote C. albicans colonisation of the murine large intestine.
Collapse
Affiliation(s)
- Gabriela M. Avelar
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Ivy M. Dambuza
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Liviana Ricci
- Microbiome, Food Innovation and Food Security Research Theme, Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Raif Yuecel
- Iain Fraser Cytometry Centre, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Kevin Mackenzie
- Microscopy & Histology Facility, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Delma S. Childers
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Judith M. Bain
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Arnab Pradhan
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Daniel E. Larcombe
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Lars P. Erwig
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Johnson-Johnson Innovation, EMEA Innovation Centre, One Chapel Place, London W1G 0BG, UK
| | - Gordon D. Brown
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Sylvia H. Duncan
- Microbiome, Food Innovation and Food Security Research Theme, Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Neil A.R. Gow
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Alan W. Walker
- Microbiome, Food Innovation and Food Security Research Theme, Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J.P. Brown
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
- Corresponding author at: Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK.
| |
Collapse
|
46
|
Lam PPY, Zhou N, Wong HM, Yiu CKY. Oral Health Status of Children and Adolescents Living with HIV Undergoing Antiretroviral Therapy: A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12864. [PMID: 36232165 PMCID: PMC9564723 DOI: 10.3390/ijerph191912864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
Antiretroviral therapy (ART) increases the survival of HIV-infected children, but might also bring in oral health-related side effects and increase their risks of oral diseases. The review compared the oral health status of children living with HIV (CLWH) undergoing ART with healthy controls. Dual independent screening and study selection from four electronic databases and manual searches, data extraction, risk of bias assessment, and quality-of-evidence evaluation with Grading of Recommendations Assessment Development and Evaluation were performed. Twelve studies were included in qualitative and quantitative analysis. CLWH taking ART had a significantly higher prevalence of periodontal diseases (OR = 3.11, 95% CI 1.62-5.97), mucosal hyperpigmentation (OR = 20.35, 95% CI 3.86-107.39), and orofacial-related opportunistic infections than healthy controls. No significant differences regarding caries prevalence and tooth development were identified. Those with CD4+ T-cell counts below 250 cells/mm3 were more likely to manifest opportunistic infections, while medication duration had minimal influence on the prevalence of orofacial opportunistic infections. The current findings did not identify HIV and antiretroviral status as predisposing factors to dental caries, but affirmed the associated increased risk of periodontal diseases, mucosal hyperpigmentation and candidiasis.
Collapse
Affiliation(s)
- Phoebe Pui Ying Lam
- Paediatric Dentistry, Faculty of Dentistry, the University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ni Zhou
- School of Stomatology, Kunming Medical University, Kunming 650032, China
| | - Hai Ming Wong
- Paediatric Dentistry, Faculty of Dentistry, the University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry, Faculty of Dentistry, the University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
47
|
da Silva Neto JX, Dias LP, Lopes de Souza LA, Silva da Costa HP, Vasconcelos IM, Pereira ML, de Oliveira JTA, Cardozo CJP, Gonçalves Moura LFW, de Sousa JS, Carneiro RF, Lopes TDP, Bezerra de Sousa DDO. Insights into the structure and mechanism of action of the anti-candidal lectin Mo-CBP2 and evaluation of its synergistic effect and antibiofilm activity. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Wagner AS, Lumsdaine SW, Mangrum MM, King AE, Hancock TJ, Sparer TE, Reynolds TB. Cek1 regulates ß(1,3)-glucan exposure through calcineurin effectors in Candida albicans. PLoS Genet 2022; 18:e1010405. [PMID: 36121853 PMCID: PMC9521907 DOI: 10.1371/journal.pgen.1010405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
In order to successfully induce disease, the fungal pathogen Candida albicans regulates exposure of antigens like the cell wall polysaccharide ß(1,3)-glucan to the host immune system. C. albicans covers (masks) ß(1,3)-glucan with a layer of mannosylated glycoproteins, which aids in immune system evasion by acting as a barrier to recognition by host pattern recognition receptors. Consequently, enhanced ß(1,3)-glucan exposure (unmasking) makes fungal cells more visible to host immune cells and facilitates more robust fungal clearance. However, an understanding of how C. albicans regulates its exposure levels of ß(1,3)-glucan is needed to leverage this phenotype. Signal transduction pathways and their corresponding effector genes mediating these changes are only beginning to be defined. Here, we report that the phosphatase calcineurin mediates unmasking of ß(1,3)-glucan in response to inputs from the Cek1 MAPK pathway and in response to caspofungin exposure. In contrast, calcineurin reduces ß-glucan exposure in response to high levels of extracellular calcium. Thus, depending on the input, calcineurin acts as a switchboard to regulate ß(1,3)-glucan exposure levels. By leveraging these differential ß(1,3)-glucan exposure phenotypes, we identified two novel effector genes in the calcineurin regulon, FGR41 and C1_11990W_A, that encode putative cell wall proteins and mediate masking/unmasking. Loss of either effector caused unmasking and attenuated virulence during systemic infection in mice. Furthermore, immunosuppression restored the colonization decrease seen in mice infected with the fgr41Δ/Δ mutant to wild-type levels, demonstrating a reliance on the host immune system for virulence attenuation. Thus, calcineurin and its downstream regulon are general regulators of unmasking.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Stephen W. Lumsdaine
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Ainsley E. King
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Trevor J. Hancock
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Timothy E. Sparer
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| |
Collapse
|
49
|
Shao TY, Haslam DB, Bennett RJ, Way SS. Friendly fungi: symbiosis with commensal Candida albicans. Trends Immunol 2022; 43:706-717. [PMID: 35961916 PMCID: PMC10027380 DOI: 10.1016/j.it.2022.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 12/22/2022]
Abstract
Mucosal tissues are constitutively colonized by a wide assortment of host-adapted microbes. This includes the polymorphic fungus Candida albicans which is a primary target of human adaptive responses. Immunogenicity is replicated after intestinal colonization in preclinical models with a surprising array of protective benefits for most hosts, but harmful consequences for a few. The interaction between fungus and host is complex, and traditionally, the masking of antigenic fungal ligands has been viewed as a tactic for fungal immune evasion during invasive infection. However, we propose that dynamic expression of cell wall moieties, host cell lysins, and other antigenic C. albicans determinants is necessary during the more ubiquitous context of intestinal colonization to prime immunogenicity and optimize mammalian host symbiosis.
Collapse
Affiliation(s)
- Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Immunobiology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - David B Haslam
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Richard J Bennett
- Molecular Microbiology and Immunology Department, Brown University, Providence, RI 02912, USA.
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
50
|
Jenull S, Shivarathri R, Tsymala I, Penninger P, Trinh PC, Nogueira F, Chauhan M, Singh A, Petryshyn A, Stoiber A, Chowdhary A, Chauhan N, Kuchler K. Transcriptomics and Phenotyping Define Genetic Signatures Associated with Echinocandin Resistance in Candida auris. mBio 2022; 13:e0079922. [PMID: 35968956 PMCID: PMC9426441 DOI: 10.1128/mbio.00799-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Candida auris emerged as a human fungal pathogen only during the past decade. Remarkably, C. auris displays high degrees of genomic diversity and phenotypic plasticity, with four major clades causing hospital outbreaks with high mortality and morbidity rates. C. auris can show clinical resistance to all classes of antifungal drugs, including echinocandins that are usually recommended as first-line therapies for invasive candidiasis. Here, we exploit transcriptomics coupled with phenotypic profiling to characterize a set of clinical C. auris isolates displaying pronounced echinocandin resistance (ECN-R). A hot spot mutation in the echinocandin FKS1 target gene is present in all resistant isolates. Moreover, ECN-R strains share a core signature set of 362 genes differentially expressed in ECN-R isolates. Among others, mitochondrial gene expression and genes affecting cell wall function appear to be the most prominent, with the latter correlating well with enhanced adhesive traits, increased cell wall mannan content, and altered sensitivity to cell wall stress of ECN-R isolates. Moreover, ECN-R phenotypic signatures were also linked to pathogen recognition and interaction with immune cells. Hence, transcriptomics paired with phenotyping is a suitable tool to predict resistance and fitness traits as well as treatment outcomes in pathogen populations with complex phenotypic diversity. IMPORTANCE The surge in antimicrobial drug resistance in some bacterial and fungal pathogens constitutes a significant challenge to health care facilities. The emerging human fungal pathogen Candida auris has been particularly concerning, as isolates can display pan-antifungal resistance traits against all drugs, including echinocandins. However, the mechanisms underlying this phenotypic diversity remain poorly understood. We identify transcriptomic signatures in C. auris isolates resistant to otherwise fungicidal echinocandins. We identify a set of differentially expressed genes shared by resistant strains compared to unrelated susceptible isolates. Moreover, phenotyping demonstrates that resistant strains show distinct behaviors, with implications for host-pathogen interactions. Hence, this work provides a solid basis to identify the mechanistic links between antifungal multidrug resistance and fitness costs that affect the interaction of C. auris with host immune defenses.
Collapse
Affiliation(s)
- Sabrina Jenull
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Raju Shivarathri
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Irina Tsymala
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Philipp Penninger
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Phan-Canh Trinh
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Filomena Nogueira
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
- CCRI-St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Manju Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Ashutosh Singh
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Andriy Petryshyn
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anton Stoiber
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anuradha Chowdhary
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Neeraj Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Karl Kuchler
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| |
Collapse
|