1
|
Jia K, Zhang Y, Jiang M, Cui M, Wang J, Zhang J, Wang H, Zhao H, Li M, Zou Q, Zeng H. Dual-antigen fusion protein vaccination induces protective immunity against Candida albicans infection in mice. Hum Vaccin Immunother 2024; 20:2406065. [PMID: 39327639 PMCID: PMC11441037 DOI: 10.1080/21645515.2024.2406065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Candida albicans Is a leading cause of nosocomial bloodstream infections, particularly in immunocompromised patients. Current therapeutic strategies are insufficient, highlighting the need for effective vaccines. This study aimed to evaluate the efficacy of a dual-antigen fusion protein vaccine (AH) targeting the Als3 and Hyr1 proteins of C. albicans, using AlPO4 as an adjuvant. The AH vaccine was constructed by fusing Als317-432 and Hyr125-350 proteins, and its immunogenicity was tested in BALB/c mice and New Zealand white rabbits. Mice received three intramuscular doses of the vaccine combined with AlPO4, followed by a lethal challenge with C. albicans SC5314. Survival rates, antibody responses, cytokine production, fungal burdens, and organ pathology were assessed. The vaccine's efficacy was also validated using rabbit serum. Mice vaccinated with the AH-AlPO4 combination exhibited significantly higher antibody titers, particularly IgG and its subclasses, compared to controls (p < .001). The survival rate of vaccinated mice was 80% post-infection, significantly higher than the control group (p < .01). Vaccinated mice showed reduced fungal loads in the blood, kidneys, spleen, and liver (p < .05). Increased levels of interferon gamma and interleukin (IL)-17A were observed, indicating robust T helper (Th) 1 and Th17 cell responses. Vaccination mitigated organ damage, with kidney and liver pathology scores significantly lower than those of unvaccinated mice (p < .05). Rabbit serum with polyclonal antibodies demonstrated effective antifungal activity, confirming vaccine efficacy across species. The AH-AlPO4 vaccine effectively induced strong immune responses, reduced fungal burden, and protected against organ pathology in C. albicans infections. These findings support further development of dual-antigen vaccine strategies.
Collapse
Affiliation(s)
- Keran Jia
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, China
| | - Yanhao Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, China
| | - Mengyu Jiang
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Mengge Cui
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Jia Wang
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Jiajia Zhang
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Hua Wang
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Huihai Zhao
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Mengyan Li
- Department of Clinical Laboratory, The 980th Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Pang LM, Zeng G, Chow EWL, Xu X, Li N, Kok YJ, Chong SC, Bi X, Gao J, Seneviratne CJ, Wang Y. Sdd3 regulates the biofilm formation of Candida albicans via the Rho1-PKC-MAPK pathway. mBio 2024:e0328324. [PMID: 39688394 DOI: 10.1128/mbio.03283-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Candida albicans, the most frequently isolated fungal pathogen in humans, forms biofilms that enhance resistance to antifungal drugs and host immunity, leading to frequent treatment failure. Understanding the molecular mechanisms governing biofilm formation is crucial for developing anti-biofilm therapies. In this study, we conducted a genetic screen to identify novel genes that regulate biofilm formation in C. albicans. One identified gene is ORF19.6693, a homolog of the Saccharomyces cerevisiae SDD3 gene. The sdd3∆/∆ mutant exhibited severe defects in biofilm formation and significantly reduced chitin content in the cell wall. Overexpression of the constitutively active version of the Rho1 GTPase Rho1G18V, an upstream activator of the protein kinase C (PKC)-mitogen-activated protein kinase (MAPK) cell-wall integrity pathway, rescued these defects. Affinity purification, mass spectrometry, and co-immunoprecipitation revealed Sdd3's physical interaction with Bem2, the GTPase-activating protein of Rho1. Deletion of SDD3 significantly reduced the amount of the active GTP-bound form of Rho1, thereby diminishing PKC-MAPK signaling and downregulating chitin synthase genes CHS2 and CHS8. Taken together, our studies identify a new biofilm regulator, Sdd3, in C. albicans that modulates Rho1 activity through its inhibitory interaction with Bem2, thereby regulating the PKC-MAPK pathway to control chitin biosynthesis, which is critical for biofilm formation. As an upstream component of the pathway and lacking a homolog in mammals, Sdd3 has the potential to serve as an antifungal target for biofilm infections.IMPORTANCEThe human fungal pathogen Candida albicans is categorized as a critical priority pathogen on the World Health Organization's Fungal Priority Pathogens List. A key virulence attribute of this pathogen is its ability to form biofilms on the surfaces of indwelling medical devices. Fungal cells in biofilms are highly resistant to antifungal drugs and host immunity, leading to treatment failure. This study conducted a genetic screen to discover novel genes that regulate biofilm formation. We found that deletion of the SDD3 gene caused severe biofilm defects. Sdd3 negatively regulates the Rho1 GTPase, an upstream activator of the protein kinase C-mitogen-activated protein kinase pathway, through direct interaction with Bem2, the GTPase-activating protein of Rho1, resulting in a significant decrease in chitin content in the fungal cell wall. This chitin synthesis defect leads to biofilm formation failure. Given its essential role in biofilm formation, Sdd3 could serve as an antifungal target for biofilm infections.
Collapse
Affiliation(s)
- Li Mei Pang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center Singapore, Singapore, Singapore
| | - Guisheng Zeng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Eve Wai Ling Chow
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xiaoli Xu
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ning Li
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Shu Chen Chong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Jiaxin Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Beijing, China
| | - Chaminda Jayampath Seneviratne
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center Singapore, Singapore, Singapore
- Oral Health ACP, Duke NUS Medical School, Singapore, Singapore
- School of Dentistry, The University of Queensland, St Lucia, Australia
| | - Yue Wang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Bekirian C, Valsecchi I, Bachellier-Bassi S, Scandola C, Guijarro JI, Chauvel M, Mourer T, Gow NAR, Aimanianda VK, d'Enfert C, Fontaine T. β-1,6-Glucan plays a central role in the structure and remodeling of the bilaminate fungal cell wall. eLife 2024; 13:RP100569. [PMID: 39636210 PMCID: PMC11620752 DOI: 10.7554/elife.100569] [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: 12/07/2024] Open
Abstract
The cell wall of human fungal pathogens plays critical roles as an architectural scaffold and as a target and modulator of the host immune response. Although the cell wall of the pathogenic yeast Candida albicans is intensively studied, one of the major fibrillar components in its cell wall, β-1,6-glucan, has been largely neglected. Here, we show that β-1,6-glucan is essential for bilayered cell wall organization, cell wall integrity, and filamentous growth. For the first time, we show that β-1,6-glucan production compensates the defect in mannan elongation in the outer layer of the cell wall. In addition, β-1,6-glucan dynamics are also coordinated by host environmental stimuli and stresses with wall remodeling, where the regulation of β-1,6-glucan structure and chain length is a crucial process. As we point out that β-1,6-glucan is exposed at the yeast surface and modulate immune response, β-1,6-glucan must be considered a key factor in host-pathogen interactions.
Collapse
Affiliation(s)
- Clara Bekirian
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie et Pathogénicité FongiquesParisFrance
| | - Isabel Valsecchi
- EA DYNAMYC 7380, Faculté de Santé, Université Paris-Est Créteil (UPEC), École Nationale Vétérinaire d'Alfort (EnvA), USC AnsesCréteilFrance
| | - Sophie Bachellier-Bassi
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie et Pathogénicité FongiquesParisFrance
| | - Cyril Scandola
- Institut Pasteur, Université Paris Cité, Ultrastructural Bioimaging UnitParisFrance
| | - J Inaki Guijarro
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Biological NMR and HDX-MS Technological PlatformParisFrance
| | - Murielle Chauvel
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie et Pathogénicité FongiquesParisFrance
| | - Thierry Mourer
- Institut Pasteur, Advanced Molecular Virology GroupParisFrance
| | - Neil AR Gow
- Medical Research Council Centre for Medical Mycology, University of ExeterExeterUnited Kingdom
| | | | - Christophe d'Enfert
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie et Pathogénicité FongiquesParisFrance
| | - Thierry Fontaine
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie et Pathogénicité FongiquesParisFrance
| |
Collapse
|
4
|
Jaeger M, Dietschmann A, Austermeier S, Dinçer S, Porschitz P, Vornholz L, Maas RJ, Sprenkeler EG, Ruland J, Wirtz S, Azam T, Joosten LA, Hube B, Netea MG, Dinarello CA, Gresnigt MS. Alpha1-antitrypsin impacts innate host-pathogen interactions with Candida albicans by stimulating fungal filamentation. Virulence 2024; 15:2333367. [PMID: 38515333 PMCID: PMC11008552 DOI: 10.1080/21505594.2024.2333367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
Our immune system possesses sophisticated mechanisms to cope with invading microorganisms, while pathogens evolve strategies to deal with threats imposed by host immunity. Human plasma protein α1-antitrypsin (AAT) exhibits pleiotropic immune-modulating properties by both preventing immunopathology and improving antimicrobial host defence. Genetic associations suggested a role for AAT in candidemia, the most frequent fungal blood stream infection in intensive care units, yet little is known about how AAT influences interactions between Candida albicans and the immune system. Here, we show that AAT differentially impacts fungal killing by innate phagocytes. We observed that AAT induces fungal transcriptional reprogramming, associated with cell wall remodelling and downregulation of filamentation repressors. At low concentrations, the cell-wall remodelling induced by AAT increased immunogenic β-glucan exposure and consequently improved fungal clearance by monocytes. Contrastingly, higher AAT concentrations led to excessive C. albicans filamentation and thus promoted fungal immune escape from monocytes and macrophages. This underscores that fungal adaptations to the host protein AAT can differentially define the outcome of encounters with innate immune cells, either contributing to improved immune recognition or fungal immune escape.
Collapse
Affiliation(s)
- Martin Jaeger
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Sude Dinçer
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Pauline Porschitz
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Larsen Vornholz
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Ralph J.A. Maas
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evelien G.G. Sprenkeler
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, USA
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Department of Internal Medicine, Radboud University Medical Center and Radboud Center for Infectious diseases (RCI), Nijmegen, the Netherlands
| | - Mark S. Gresnigt
- Department of Medicine, University of Colorado Denver, Aurora, USA
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| |
Collapse
|
5
|
Almeida NLM, Peralta LCF, Pontes FML, Rinaldo D, Porto VC, Lara VS. Anti-Candida activity and biocompatibility of silver nanoparticles associated with denture glaze: a new approach to the management of denture stomatitis. Folia Microbiol (Praha) 2024; 69:1229-1246. [PMID: 38652435 DOI: 10.1007/s12223-024-01161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
The association of silver nanoparticles (AgNps) to sealant agent Palaseal® can be a promising alternative for complete denture wearers who may develop denture stomatitis (DS). The study aimed to evaluate the anti-Candida and biocompatible potential of silver nanoparticles synthesized by three routes associated with denture glaze to prevent and/or treat oral candidiasis. Surface acrylic resin specimens were treated with different associations of glaze with AgNps (VER+AgUV, VER+AgTurk and VER+AgGm). As controls, specimens were treated with glaze+nystatin (VER+Nyst), glaze only (VER) or submerged in PBS (PBS). Afterwards, Candida albicans biofilm was developed for 24 h, 15 d and 30 d. Subsequently, the biofilm was quantified by CFU/mL, XTT assay and confocal laser scanning microscopy. Fibroblasts were submitted to conditioned medium with the same associations for 24, 48 and 72 h and LIVE/DEAD® viability test was carried out. Regardless of the period, there was a significant reduction (p < 0.01) of viable fungal cells load, as well as inhibition of fungal metabolic activity, in specimens treated with glaze+AgNps associations, compared to VER and PBS. The anti-Candida effects of the associations were similar to the VER+Nyst group, with emphasis on VER+AgGm, which showed the highest percentage values of non-viable fungal cells maintained over time. The associations did not prove toxicity to fibroblasts. The AgNps exerted antimicrobial activity against C. albicans biofilms and are biocompatible. The most effective results were achieved with the association of glaze+silver nanoparticles synthesized by the green chemistry method (AgGm), proving to be an innovative alternative in the management of DS.
Collapse
Affiliation(s)
- Nara Lígia Martins Almeida
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, 17012-901, Brazil
| | - Laura Catalí Ferreira Peralta
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru 17012-901, Brazil
| | | | - Daniel Rinaldo
- Department of Chemistry, São Paulo State University (UNESP), Bauru 17033-360, Brazil
| | - Vinicius Carvalho Porto
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru 17012-901, Brazil
| | - Vanessa Soares Lara
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, 17012-901, Brazil.
| |
Collapse
|
6
|
Park J, Park S, Kim J, Cho YJ, Lee JS. Ctr9 promotes virulence of Candida albicans by regulating methionine metabolism. Virulence 2024; 15:2405616. [PMID: 39316797 PMCID: PMC11423685 DOI: 10.1080/21505594.2024.2405616] [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: 01/16/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
Candida albicans, a part of normal flora, is an opportunistic fungal pathogen and causes severe health issues in immunocompromised patients. Its pathogenicity is intricately linked to the transcriptional regulation of its metabolic pathways. Paf1 complex (Paf1C) is a crucial transcriptional regulator that is highly conserved in eukaryotes. The objective of this study was to explore the role of Paf1C in the metabolic pathways and how it influences the pathogenicity of C. albicans. Paf1C knockout mutant strains of C. albicans (ctr9Δ/Δ, leo1Δ/Δ, and cdc73Δ/Δ) were generated using the CRISPR-Cas9 system. To investigate the effect of Paf1C on pathogenicity, macrophage interaction assays and mouse survival tests were conducted. The growth patterns of the Paf1C knockout mutants were analyzed through spotting assays and growth curve measurements. Transcriptome analysis was conducted under yeast conditions (30°C without serum) and hyphal conditions (37°C with 10% FBS), to further elucidate the role of Paf1C in the pathogenicity of C. albicans. CTR9 deletion resulted in the attenuation of C. albicans virulence, in macrophage and mouse models. Furthermore, we confirmed that the reduced virulence of the ctr9Δ/Δ mutant can be attributed to a decrease in C. albicans cell abundance. Moreover, transcriptome analysis revealed that metabolic processes required for cell proliferation are impaired in ctr9Δ/Δ mutant. Notably, CTR9 deletion led to the downregulation of methionine biosynthetic genes and the cAMP-PKA signaling pathway-related hypha essential genes, which are pivotal for virulence. Our results suggest that Ctr9-regulated methionine metabolism is a crucial factor for determining C. albicans pathogenicity.
Collapse
Affiliation(s)
- Jiyeon Park
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Shinae Park
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Jueun Kim
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Yong-Joon Cho
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon, Republic of Korea
| | - Jung-Shin Lee
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
7
|
Li L, Cai F, Guo C, Liu Z, Qin J, Huang J. Gut microbiome and NAFLD: impact and therapeutic potential. Front Microbiol 2024; 15:1500453. [PMID: 39664063 PMCID: PMC11632136 DOI: 10.3389/fmicb.2024.1500453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 12/13/2024] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) affects approximately 32.4% of the global population and poses a significant health concern. Emerging evidence underscores the pivotal role of the gut microbiota-including bacteria, viruses, fungi, and parasites-in the development and progression of NAFLD. Dysbiosis among gut bacteria alters key biological pathways that contribute to liver fat accumulation and inflammation. The gut virome, comprising bacteriophages and eukaryotic viruses, significantly shapes microbial community dynamics and impacts host metabolism through complex interactions. Similarly, gut fungi maintain a symbiotic relationship with bacteria; the relationship between gut fungi and bacteria is crucial for overall host health, with certain fungal species such as Candida in NAFLD patients showing detrimental associations with metabolic markers and liver function. Additionally, the "hygiene hypothesis" suggests that reduced exposure to gut parasites may affect immune regulation and metabolic processes, potentially influencing conditions like obesity and insulin resistance. This review synthesizes current knowledge on the intricate interactions within the gut microbiota and their associations with NAFLD. We highlight the therapeutic potential of targeting these microbial communities through interventions such as probiotics, prebiotics, and fecal microbiota transplantation. Addressing the complexities of NAFLD requires comprehensive strategies that consider the multifaceted roles of gut microorganisms in disease pathology.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
8
|
Wang J, Pu X, Gu Z. Clotrimazole-induced shifts in vaginal bacteriome and lipid metabolism: insights into recovery mechanisms in vulvovaginal candidiasis. J Appl Microbiol 2024; 135:lxae269. [PMID: 39419780 DOI: 10.1093/jambio/lxae269] [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: 09/15/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
AIMS Vulvovaginal candidiasis (VVC) is a prevalent condition affecting a significant proportion of women worldwide, with recurrent episodes leading to detrimental effects on quality of life. While treatment with clotrimazole is common, the specific alterations it evokes in the vaginal bacteriome and metabolome were previously underexplored. METHODS AND RESULTS In this prospective study, we enrolled reproductive-age women diagnosed with single VVC and conducted comprehensive analyses of vaginal fungi, bacteriome, and metabolome before and after local clotrimazole treatment. We observed a significant reduction in Candida albicans and notable improvements in vaginal cleanliness. Advanced sequencing revealed substantial shifts in the vaginal bacteriome, with an increase in Lactobacillus-dominant communities post-treatment. Our findings identified 17 differentially abundant bacterial species, including notable decreases in pathogenic anaerobes such as Gardnerella vaginalis, Dialister micraerophilus, and Aerococcus christensenii, suggesting a restoration of a healthier microbial balance. Furthermore, metabolomic analysis revealed significant changes in 230 metabolites, particularly within lipid metabolism pathways, with marked downregulation of lipid-related compounds linked to inflammation. Correlation studies indicated a strong interplay between lipid metabolites and specific bacterial species, emphasizing the influence of clotrimazole treatment on microbial and metabolic interactions. Importantly, predictive models using microbiota and metabolite signatures demonstrated high accuracy in distinguishing pre- and post-treatment states. CONCLUSIONS This research highlights clotrimazole's dual role in effectively clearing Candida infection and promoting a healthier vaginal microenvironment, paving the way for novel microbial and metabolomic-based diagnostic approaches to enhance VVC management and understand its underlying mechanisms.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gaoke Road, Shanghai 201204, China
| | - Xiaowen Pu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gaoke Road, Shanghai 201204, China
| | - Zhengrong Gu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gaoke Road, Shanghai 201204, China
| |
Collapse
|
9
|
Pan H, Xu J, Wang R, Cheng M, Wang Y, Song B. Development of ROS-sensitive capofungin hydrogel by crosslinking chitosan with four-arm polyethylene glycol derivative for treatment of vulvovaginal candidiasis. Int J Biol Macromol 2024; 279:135157. [PMID: 39214224 DOI: 10.1016/j.ijbiomac.2024.135157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Both exogenous and endogenous reactive oxygen species (ROS) in vulvovaginal candidiasis (VVC) play pivotal roles in promoting the hyphal formation of Candida albicans (CA), which suggests that clearing ROS could inhibit CA hyphae formation. A ROS-sensitive hydrogel (CAS@4Arm-PB/CS) was formulated by using a novel four-arm polyethylene glycol (4Arm-PEG) derivative (4Arm-PB) as a crosslinking agent, chitosan (CS) as the hydrogel matrix, and caspofungin (CAS) as the antifungal drug against CA. The ROS-sensitivity, disintegration mechanism, crosslinking action, swelling degree, microstructure, modulus, and rheological properties of 4Arm-PB were characterized. According to the results, 5.0 % 4Arm-PB could quickly and efficiently cross-link 0.5 mg/mL of CS. The ROS-sensitivity of 4Arm-PB was 10-50 μM, indicating a strong ROS sensitivity. The in vitro and in vivo anti-CA results indicated that CAS@4Arm-PB/CS not only cleared endogenous and exogenous ROS and inhibited the formation of CA hyphae and biofilm but also contributed beneficially to the treatment of VVC mice caused by CA infection, implying a certain safety aspect and an in vivo applicability. This research introduces a novel functional crosslinking agent for CS hydrogel formulation, presenting a new avenue for hydrogel-based drug delivery systems and therapeutic strategies for VVC treatment.
Collapse
Affiliation(s)
- Hui Pan
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Junjing Xu
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Ruizhe Wang
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Min Cheng
- Basic Medical School, Shandong Second Medical University, Weifang 261053, China
| | - Yuzhen Wang
- Medical Imaging Specialty, Shandong Second Medical University, Weifang 261053, China.
| | - Bo Song
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
10
|
Lee YH, Jung J, Hong JY. Oral Microbial Changes in Oral Squamous Cell Carcinoma: Focus on Treponema denticola, Lactobacillus casei, and Candida albicans. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1753. [PMID: 39596938 PMCID: PMC11596053 DOI: 10.3390/medicina60111753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/24/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: In this study, we aimed to explore the oral bacteria and fungi that can help discern oral squamous cell carcinoma (OSCC) and investigate the correlations between multiple key pathogens. Materials and Methods: Twelve participants (8 females and 4 males; mean age, 54.33 ± 20.65 years) were prospectively recruited into three groups: Group 1: healthy control, Group 2: patients with stomatitis, and Group 3: patients with OSCC, with 4 individuals in each group. Unstimulated whole saliva samples from these participants were analyzed using real-time PCR to assess the presence and abundance of 14 major oral bacterial species and Candida albicans. Results: The analysis revealed significant differences for certain microorganisms, namely, Treponema denticola (T. denticola), Lactobacillus casei (L. casei), and Candida albicans. T. denticola was most abundant in the OSCC group (5,358,692.95 ± 3,540,767.33), compared to the stomatitis (123,355.54 ± 197,490.86) and healthy control (9999.21 ± 11,998.40) groups. L. casei was undetectable in the healthy control group but was significantly more abundant in the stomatitis group (1653.94 ± 2981.98) and even higher in the OSCC group (21,336.95 ± 9258.79) (p = 0.001). A similar trend was observed for C. albicans, with DNA copy numbers rising from the healthy control (464.29 ± 716.76) to the stomatitis (1861.30 ± 1206.15) to the OSCC group (9347.98 ± 5128.54) (p = 0.006). The amount of T. denticola was positively correlated with L. casei (r = 0.890, p < 0.001) and C. albicans (r = 0.724, p = 0.008). L. casei's DNA copy number was strongly correlated with C. albicans (r = 0.931, p < 0.001). These three oral microbes exhibited strong positive correlations with each other and had various direct or indirect relationships with other species. Conclusions: In the OSCC group, T. denticola, L. casei, and C. albicans exhibited strong positive correlations with one another, further emphasizing the need for a deeper understanding of the complex microbial interactions in the OSCC environment.
Collapse
Affiliation(s)
- Yeon-Hee Lee
- Department of Orofacial Pain and Oral Medicine, Kyung Hee University Dental Hospital, #613 Hoegi-dong, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Junho Jung
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea;
| | - Ji-Youn Hong
- Department of Periodontology, Periodontal-Implant Clinical Research Institute, School of Dentistry, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea;
| |
Collapse
|
11
|
Alshahrani MM. Antifungal potential of marine bacterial compounds in inhibiting Candida albicans Yck2 to overcome echinocandin resistance: a molecular dynamics study. Front Pharmacol 2024; 15:1459964. [PMID: 39484169 PMCID: PMC11525067 DOI: 10.3389/fphar.2024.1459964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/17/2024] [Indexed: 11/03/2024] Open
Abstract
Candida albicans (C. albicans), a common fungal pathogen, poses a significant threat to immunocompromised individuals, particularly due to the emergence of resistance against echinocandins, a primary class of antifungal agents. Yck2 protein, a key regulator of cell wall integrity and signaling pathways in C. albicans, was targeted to overcome this resistance. A virtual screening was used to identify Yck2 inhibitors from marine bacterial compounds. Further re-docking, molecular dynamics simulations, and various analyses such as root mean square deviation (RMSD), root mean square fluctuation (RMSF), hydrogen bonding, free binding energy calculations, and RG-RMSD-based free energy landscape were conducted to evaluate the efficacy and stability of the identified compounds. Among the compounds screened, CMNPD27166 and CMNPD27283 emerged as the most promising candidates, demonstrating superior binding affinities, enhanced stability, and favorable interaction dynamics with Yck2, surpassing both the control and other compounds in efficacy. In contrast, CMNPD19660 and CMNPD24402, while effective, showed lesser potential. These findings highlight the utility of computational drug discovery techniques in identifying and optimizing potential therapeutic agents and suggest that marine-derived molecules could significantly impact the development of novel antifungal therapies. Further experimental validation of the leading candidates, CMNPD27166 and CMNPD27283, is recommended to confirm their potential as effective antifungal agents against echinocandin-resistant C. albicans infections.
Collapse
|
12
|
Hill JH, Round JL. Intestinal fungal-host interactions in promoting and maintaining health. Cell Host Microbe 2024; 32:1668-1680. [PMID: 39389031 DOI: 10.1016/j.chom.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 10/12/2024]
Abstract
The resident microbiota are a key component of a healthy organism. The vast majority of microbiome studies have focused on bacterial members, which constitute a significant portion of resident microbial biomass. Recent studies have demonstrated how the fungal component of the microbiota, or the mycobiome, influences mammalian biology despite its low abundance compared to other microbes. Fungi are known for their pathogenic potential, yet fungi are also prominent colonizers in healthy states, highlighting their duality. We summarize the characteristics that define the gut mycobiome across life, the factors that can impact its composition, and studies that identify mechanisms of how fungi confer health benefits. The goal of this review is to synthesize our knowledge regarding the composition and function of a healthy mycobiome with a view to inspiring future therapeutic advances.
Collapse
Affiliation(s)
- Jennifer H Hill
- University of Colorado Boulder, BioFrontiers Institute, Department of Molecular, Cellular & Developmental Biology, Boulder, CO 80303, USA.
| | - June L Round
- University of Utah, School of Medicine, Department of Pathology, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA.
| |
Collapse
|
13
|
Holzknecht J, Marx F. Navigating the fungal battlefield: cysteine-rich antifungal proteins and peptides from Eurotiales. FRONTIERS IN FUNGAL BIOLOGY 2024; 5:1451455. [PMID: 39323611 PMCID: PMC11423270 DOI: 10.3389/ffunb.2024.1451455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024]
Abstract
Fungi are ubiquitous in the environment and play a key role in the decomposition and recycling of nutrients. On the one hand, their special properties are a great asset for the agricultural and industrial sector, as they are used as source of nutrients, producers of enzymes, pigments, flavorings, and biocontrol agents, and in food processing, bio-remediation and plant growth promotion. On the other hand, they pose a serious challenge to our lives and the environment, as they are responsible for fungal infections in plants, animals and humans. Although host immunity opposes invading pathogens, certain factors favor the manifestation of fungal diseases. The prevalence of fungal infections is on the rise, and there is an alarming increase in the resistance of fungal pathogens to approved drugs. The limited number of antimycotics, the obstacles encountered in the development of new drugs due to the poor tolerability of antifungal agents in patients, the limited number of unique antifungal targets, and the low species specificity contribute to the gradual depletion of the antifungal pipeline and newly discovered antifungal drugs are rare. Promising candidates as next-generation therapeutics are antimicrobial proteins and peptides (AMPs) produced by numerous prokaryotic and eukaryotic organisms belonging to all kingdom classes. Importantly, filamentous fungi from the order Eurotiales have been shown to be a rich source of AMPs with specific antifungal activity. A growing number of published studies reflects the efforts made in the search for new antifungal proteins and peptides (AFPs), their efficacy, species specificity and applicability. In this review, we discuss important aspects related to fungi, their impact on our life and issues involved in treating fungal infections in plants, animals and humans. We specifically highlight the potential of AFPs from Eurotiales as promising alternative antifungal therapeutics. This article provides insight into the structural features, mode of action, and progress made toward their potential application in a clinical and agricultural setting. It also identifies the challenges that must be overcome in order to develop AFPs into therapeutics.
Collapse
Affiliation(s)
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Innsbruck Medical University,
Innsbruck, Austria
| |
Collapse
|
14
|
Huma ZE, Saleem S, Imran M, Raza SM, Jabeen K, Arshad F. Role of ERG11 and MDR1 genes in cycloheximide and multidrug resistance in Candida species. Braz J Microbiol 2024; 55:2569-2579. [PMID: 38980650 PMCID: PMC11405649 DOI: 10.1007/s42770-024-01436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Candida species are amongst the commensals of the mucosal surfaces of the human body which include the oral cavity, vagina, and intestinal mucosa. Fungal infections are on the rise worldwide. The overall burden of infections due to fungi is difficult to estimate because the majority of them remain undiagnosed. The present study aims to determine the burden of antifungal resistance in low socioeconomic country, Pakistan and the frequency of ERG11 and MDR1 genes involved. A total of 636 Candida isolates were obtained from various tertiary care institutions in Lahore in the form of culture on various culture plates. Sabouraud agar culture plates were used to culture the Candida spp. Antifungal resistance was determined against Fluconazole, Itraconazole, Ketoconazole, and Nystatin via disk diffusion technique. Most resistance was observed against Fluconazole followed by Itraconazole, Ketoconazole, and Nystatin. The Candida isolates recovering from CVP tip and tissue have a high resistance profile. Candida species resistant to at least two antifungals were chosen for further ERG11 and MDR1 detection through real-time PCR. Among 255 Candida isolates, 240 contained ERG11 gene while MDR1 gene is present in 149 Candida isolates. The isolates carrying both genes were tested by the broth microdilution technique for the susceptibility against cycloheximide, all of them were able to grow in cycloheximide. The genetic determinants of antifungal resistance such as ERG11 and MDR1 are as important in the multidrug resistance against a variety of compounds and antifungal drugs.
Collapse
Affiliation(s)
- Zill-E- Huma
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan.
| | - Sidrah Saleem
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| | - Muhammad Imran
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| | - Syed Mohsin Raza
- Department of Allied Health Sciences and Medical Education, University of Health Sciences, Lahore, Pakistan
| | - Kokab Jabeen
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| | - Faiqa Arshad
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
15
|
Xia X, Wei Q, Wu H, Chen X, Xiao C, Ye Y, Liu C, Yu H, Guo Y, Sun W, Liu W. Bacillus species are core microbiota of resistant maize cultivars that induce host metabolic defense against corn stalk rot. MICROBIOME 2024; 12:156. [PMID: 39180084 PMCID: PMC11342587 DOI: 10.1186/s40168-024-01887-w] [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: 11/20/2023] [Accepted: 07/27/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Microbes colonizing each compartment of terrestrial plants are indispensable for maintaining crop health. Although corn stalk rot (CSR) is a severe disease affecting maize (Zea mays) worldwide, the mechanisms underlying host-microbe interactions across vertical compartments in maize plants, which exhibit heterogeneous CSR-resistance, remain largely uncharacterized. RESULTS Here, we investigated the microbial communities associated with CSR-resistant and CSR-susceptible maize cultivars using multi-omics analysis coupled with experimental verification. Maize cultivars resistant to CSR reshaped the microbiota and recruited Bacillus species with three phenotypes against Fusarium graminearum including niche pre-emption, potential secretion of antimicrobial compounds, and no inhibition to alleviate pathogen stress. By inducing the expression of Tyrosine decarboxylase 1 (TYDC1), encoding an enzyme that catalyzes the production of tyramine and dopamine, Bacillus isolates that do not directly suppress pathogen infection induced the synthesis of berberine, an isoquinoline alkaloid that inhibits pathogen growth. These beneficial bacteria were recruited from the rhizosphere and transferred to the stems but not grains of the CSR-resistant plants. CONCLUSIONS The current study offers insight into how maize plants respond to and interact with their microbiome and lays the foundation for preventing and treating soil-borne pathogens. Video Abstract.
Collapse
Affiliation(s)
- Xinyao Xia
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Department of Plant Pathology, Ministry of Agriculture Key Laboratory of Pest Monitoring and Green Management and Joint International Research Laboratory of Crop Molecular Breeding, Ministry of Education, China Agricultural University, Beijing, 100193, China
- Key Laboratory for Safety Assessment (Environment) of Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Qiuhe Wei
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hanxiang Wu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xinyu Chen
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Chunxia Xiao
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yiping Ye
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Chaotian Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Haiyue Yu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yuanwen Guo
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Wenxian Sun
- Department of Plant Pathology, Ministry of Agriculture Key Laboratory of Pest Monitoring and Green Management and Joint International Research Laboratory of Crop Molecular Breeding, Ministry of Education, China Agricultural University, Beijing, 100193, China
| | - Wende Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Integrated Pest Management in Crops, Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
16
|
Marsaux B, Moens F, Vandevijver G, Marzorati M, van de Wiele T. Candida species-specific colonization in the healthy and impaired human gastrointestinal tract as simulated using the Mucosal Ileum-SHIME® model. FEMS Microbiol Ecol 2024; 100:fiae113. [PMID: 39169462 DOI: 10.1093/femsec/fiae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/14/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024] Open
Abstract
Candida species primarily exist as harmless commensals in the gastrointestinal tract of warm-blooded animals. However, they can also cause life-threatening infections, which are often associated with gut microbial dysbiosis. Identifying the microbial actors that restrict Candida to commensalism remains a significant challenge. In vitro models could enable a mechanistic study of the interactions between Candida and simulated colon microbiomes. Therefore, this study aimed to elucidate the spatial and temporal colonization kinetics of specific Candida, including C. albicans, C. tropicalis, and C. parapsilosis, and their relative Nakaseomyces glabratus, by using an adapted SHIME® model, simulating the ileum, and proximal and distal colons. We monitored fungal and bacterial colonization kinetics under conditions of eubiosis (commensal lifestyle) and antibiotic-induced dysbiosis (pathogenic lifestyle). Our findings highlighted the variability in the colonization potential of Candida species across different intestinal regions. The ileum compartment proved to be the most favourable environment for C. albicans and C. parapsilosis under conditions of eubiosis. Antibiotic-induced dysbiosis resulted in resurgence of opportunistic Candida species, especially C. tropicalis and C. albicans. Future research should focus on identifying specific bacterial species influencing Candida colonization resistance and explore the long-term effects of antibiotics on the mycobiome and bacteriome.
Collapse
Affiliation(s)
- Benoît Marsaux
- ProDigest B.V., 9052 Ghent, Belgium
- CMET, Ghent University, 9000 Ghent, Belgium
| | | | | | - Massimo Marzorati
- ProDigest B.V., 9052 Ghent, Belgium
- CMET, Ghent University, 9000 Ghent, Belgium
| | - Tom van de Wiele
- ProDigest B.V., 9052 Ghent, Belgium
- CMET, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
17
|
Jaswal K, Todd OA, Flores Audelo RC, Santus W, Paul S, Singh M, Miao J, Underhill DM, Peters BM, Behnsen J. Commensal Yeast Promotes Salmonella Typhimurium Virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606421. [PMID: 39211098 PMCID: PMC11360897 DOI: 10.1101/2024.08.08.606421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Enteric pathogens engage in complex interactions with the host and the resident microbiota to establish gut colonization. Although mechanistic interactions between enteric pathogens and bacterial commensals have been extensively studied, whether and how commensal fungi affect pathogenesis of enteric infections remains largely unknown. Here we show that colonization with the common human gut commensal fungus Candida albicans worsened infections with the enteric pathogen Salmonella enterica serovar Typhimurium. Presence of C. albicans in the mouse gut increased Salmonella cecum colonization and systemic dissemination. We investigated the underlying mechanism and found that Salmonella binds to C. albicans via Type 1 fimbriae and uses its Type 3 Secretion System (T3SS) to deliver effector proteins into C. albicans . A specific effector, SopB, was sufficient to manipulate C. albicans metabolism, triggering increased arginine biosynthesis in C. albicans and the release of millimolar amounts of arginine into the extracellular environment. The released arginine, in turn, induced T3SS expression in Salmonella , increasing its invasion of epithelial cells. C. albicans deficient in arginine production was unable to increase Salmonella virulence in vitro or in vivo . In addition to modulating pathogen invasion, arginine also directly influenced the host response to infection. Arginine-producing C. albicans dampened the inflammatory response during Salmonella infection, whereas C. albicans deficient in arginine production did not. Arginine supplementation in the absence of C. albicans increased the systemic spread of Salmonella and decreased the inflammatory response, phenocopying the presence of C. albicans . In summary, we identified C. albicans colonization as a susceptibility factor for disseminated Salmonella infection, and arginine as a central metabolite in the cross-kingdom interaction between fungi, bacteria, and host.
Collapse
|
18
|
Šovljanski O, Aćimović M, Tomić A, Lončar B, Miljković A, Čabarkapa I, Pezo L. Antibacterial and Antifungal Potential of Helichrysum italicum (Roth) G. Don Essential Oil. Antibiotics (Basel) 2024; 13:722. [PMID: 39200022 PMCID: PMC11350649 DOI: 10.3390/antibiotics13080722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
Helichrysum italicum (Roth) G. Don is a typical Mediterranean plant, with limited distribution on the islands of Sardinia, Corsica, and the Iberian Peninsula, as well as the islands of the Adriatic Sea and the Balkan Peninsula. In these regions, H. italicum is mainly collected from spontaneous nature, while in recent years, there has been a pronounced cultivation trend due to increased demand and market requirements for constant quality of raw materials. Bearing in mind that biological activity is linked with chemical composition, this review aimed to collect data from different scientific databases (Scopus, PubMed, Web of Science, and Google Scholar) on the antimicrobial activity of essential oil and its chemical composition. A total of 20 papers investigating the antibacterial, antibiofilm, and antifungal activities of H. italicum essential oil were found. Furthermore, in these samples, several compounds occurred as dominant: neryl acetate, α-pinene, and γ-curcumene. These compounds are known for their antimicrobial properties, which likely contribute to the essential oil's efficacy against various microbial strains.
Collapse
Affiliation(s)
- Olja Šovljanski
- Faculty of Technology Novi Sad, University of Novi Sad, Bulevar Cara Lazara 1, 21000 Novi Sad, Serbia; (O.Š.); (A.T.); (B.L.)
| | - Milica Aćimović
- Institute of Field and Vegetable Crops Novi Sad, Maksima Gorkog 30, 21000 Novi Sad, Serbia;
| | - Ana Tomić
- Faculty of Technology Novi Sad, University of Novi Sad, Bulevar Cara Lazara 1, 21000 Novi Sad, Serbia; (O.Š.); (A.T.); (B.L.)
| | - Biljana Lončar
- Faculty of Technology Novi Sad, University of Novi Sad, Bulevar Cara Lazara 1, 21000 Novi Sad, Serbia; (O.Š.); (A.T.); (B.L.)
| | - Ana Miljković
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
| | - Ivana Čabarkapa
- Institute of Food Technology, University of Novi Sad, Bulevar Cara Lazara 1, 21000 Novi Sad, Serbia;
| | - Lato Pezo
- Institute of General and Physical Chemistry, University of Belgrade, Studentski trg 12, 11000 Belgrade, Serbia
| |
Collapse
|
19
|
Lindemann-Perez E, Perez JC. Candida albicans natural diversity: a resource to dissect fungal commensalism and pathogenesis. Curr Opin Microbiol 2024; 80:102493. [PMID: 38833793 DOI: 10.1016/j.mib.2024.102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Candida albicans is a ubiquitous fungus of humans. It is not only a component of the oral and intestinal microbiota of most healthy adults but also a major cause of mucosal disorders and life-threatening disseminated infections. Until recently, research on the biology and pathogenesis of the fungus was largely based on a single clinical isolate. We review investigations that have started to dissect a diverse set of C. albicans strains. Using different approaches to leverage the species' phenotypic and/or genetic diversity, these studies illuminate the wide range of interactions between fungus and host. While connecting genetic variants to phenotypes of interest remains challenging, research on C. albicans' natural diversity is central to understand fungal commensalism and pathogenesis.
Collapse
Affiliation(s)
- Elena Lindemann-Perez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | - J Christian Perez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA.
| |
Collapse
|
20
|
Owotade FJ, Gulube Z, Patel M. Oral Candida albicans strain diversity and maintenance in HIV positive women in South Africa. Arch Oral Biol 2024; 164:106007. [PMID: 38795522 DOI: 10.1016/j.archoralbio.2024.106007] [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: 01/11/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVE This study investigated C. albicans strain diversity and maintenance in the oral cavity of HIV positive women over a 6 month period. STUDY DESIGN C. albicans strains were isolated from 17 HIV positive women at Charlotte Maxeke Academic Hospital, Johannesburg at 3 intervals over a 6 month period. Strains were genotyped using ABC and Multilocus Sequence Typing (MLST) techniques. In the MLST technique, for each strain, a Diploid Sequence Type (DST) number was obtained. Using cluster analysis, an Unweighted Pair Group Method with Arithmetic Mean (UPGMA) dendrogram and a matrix of strain similarities were generated. Strains were also compared to the previous South African isolates documented in the MLST database. RESULTS Ninety four percent of women carried the same ABC genotype for 6 months. MLST technique, showed that ten women (58.8%) carried the same DST at 2 visits, while seven (41.2%) carried different DST at all visits. Further analysis showed that 64.7% of women were recolonised with different strains and 35.3% carried the same strains of C. albicans with heterozygosity. A total of 40 diploid sequence types were identified of which 27 DSTs were unique to this study group that were added to the MLST database. Most of the strains were closely related to previously isolated strains from South Africa. CONCLUSION Recolonization of the oral cavity with different strains and microevolution of the original strains of C. albicans can occur, which can be a potential problem for HIV patients, in whom highly virulent and drug resistant strains can emerge.
Collapse
Affiliation(s)
- F J Owotade
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, South Africa; Faculty of Dentistry, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Z Gulube
- Department of Oral Biological Sciences School of Oral Health Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| | - M Patel
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, South Africa.
| |
Collapse
|
21
|
Elnagar RM. Cross interaction between bacterial and fungal microbiota and their relevance to human health and disease: mechanistic pathways and prospective therapy. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:309-320. [PMID: 39364131 PMCID: PMC11444862 DOI: 10.12938/bmfh.2024-031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/27/2024] [Indexed: 10/05/2024]
Abstract
Diverse bacterial and fungal microbiota communities inhabit the human body, and their presence is essential for maintaining host homeostasis. The oral cavity, lung, gut, and vagina are just a few of the bodily cavities where these microorganisms communicate with one another, either directly or indirectly. The effects of this interaction can be either useful or detrimental to the host. When the healthy microbial diversity is disturbed, for instance, as a result of prolonged treatment with broad spectrum antibiotics, this allows the growth of specific microbes at the expense of others and alters their pathogenicity, causing a switch of commensal germs into pathogenic germs, which could promote tissue invasion and damage, as occurs in immunocompromised patients. Consequently, antimicrobials that specifically target pathogens may help in minimizing secondary issues that result from the disruption of useful bacterial/fungal interactions (BFIs). The interface between Candida albicans and Aspergillus fumigatus with bacteria at various body sites is emphasized in the majority of the medically important BFIs that have been reported thus far. This interface either supports or inhibits growth, or it enhances or blocks the generation of virulence factors. The aim of this review is to draw attention to the link between the bacterial and fungal microbiota and how they contribute to both normal homeostasis and disease development. Additionally, recent research that has studied microbiota as novel antimicrobials is summarized.
Collapse
Affiliation(s)
- Rasha Mokhtar Elnagar
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
22
|
Vasconcelos PGS, Lee KM, Abuna GF, Costa EMMB, Murata RM. Monoterpene antifungal activities: evaluating geraniol, citronellal, and linalool on Candida biofilm, host inflammatory responses, and structure-activity relationships. Front Pharmacol 2024; 15:1394053. [PMID: 39101130 PMCID: PMC11294919 DOI: 10.3389/fphar.2024.1394053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/06/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction: Despite the rising concern with fungal resistance, a myriad of molecules has yet to be explored. Geraniol, linalool, and citronellal are monoterpenes with the same molecular formula (C10H18O), however, neither the effect of these compounds on inflammatory axis induced by Candida spp. nor the antibiofilm Structure-Activity Relationship (SAR) have been well-investigated. Herein we analyzed geraniol, linalool and citronellal antifungal activity, cytotoxicity, and distinctive antibiofilm SAR, also the influence of geraniol on Candida spp induced dysregulated inflammatory axis, and in vivo toxicity. Methods: Minimal inhibitory (MIC) and fungicidal (MFC) concentrations against Candida spp were defined, followed by antibiofilm activity (CFU-colony forming unit/mL/g of dry weight). Cytotoxic activity was assessed using human monocytes (THP-1) and oral squamous cell (TR146). Geraniol was selected for further analysis based on antifungal, antibiofilm and cytotoxic results. Geraniol was tested using a dual-chamber co-culture model with TR146 cells infected with C. albicans, and THP-1 cells, used to mimic oral epithelium upon fungal infection. Expression of Candida enzymes (phospholipase-PLB and aspartyl proteases-SAP) and host inflammatory cytokines (interleukins: IL-1β, IL-6, IL-17, IL-18, IL-10, and Tumor necrosis factor-TNF) were analyzed. Lastly, geraniol in vivo toxicity was assessed using Galleria mellonella. Results: MIC values obtained were 1.25-5 mM/mL for geraniol, 25-100 mM/mL for linalool, and 100-200 mM/mL for citronellal. Geraniol 5 and 50 mM/mL reduced yeast viability during biofilm analysis, only 500 mM/mL of linalool was effective against a 72 h biofilm and no biofilm activity was seen for citronellal. LD50 for TR146 and THP-1 were, respectively: geraniol 5.883 and 8.027 mM/mL; linalool 1.432 and 1.709 mM/mL; and citronellal 0.3006 and 0.1825 mM/mL. Geraniol was able to downregulate expression of fungal enzymes and host pro-inflammatory cytokines IL-1β, IL-6, and IL-18. Finally, safety in vivo parameters were observed up to 20 mM/Kg. Discussion: Despite chemical similarities, geraniol presented better antifungal, antibiofilm activity, and lower cytotoxicity when compared to the other monoterpenes. It also showed low in vivo toxicity and capacity to downregulate the expression of fungal enzymes and host pro-inflammatory cytokines. Thus, it can be highlighted as a viable option for oral candidiasis treatment.
Collapse
Affiliation(s)
| | - Kyu Min Lee
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Gabriel Flores Abuna
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Edja Maria Melo Brito Costa
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Paraíba, Brazil
| | - Ramiro Mendonça Murata
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
23
|
Velleuer E, Carlberg C. A Nutrigenomic View on the Premature-Aging Disease Fanconi Anemia. Nutrients 2024; 16:2271. [PMID: 39064714 PMCID: PMC11280142 DOI: 10.3390/nu16142271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Fanconi anemia, a rare disorder with an incidence of 1 in 300,000, is caused by mutations in FANC genes, which affect the repair of DNA interstrand crosslinks. The disease is characterized by congenital malformations, bone marrow failure within the first decade of life, and recurrent squamous cell carcinomas of the oral cavity, esophagus, and anogenital regions starting around age 20. In this review, we propose that Fanconi anemia should be considered a premature-aging syndrome. Interestingly, the onset and severity of the life-limiting clinical features of Fanconi anemia can be influenced by lifestyle choices, such as a healthy diet and physical activity. These factors shape the epigenetic status of at-risk cell types and enhance the competence of the immune system through nutritional signaling. Fanconi anemia may serve as a model for understanding the aging process in the general population, addressing research gaps in its clinical presentation and suggesting prevention strategies. Additionally, we will discuss how the balance of genetic and environmental risk factors-affecting both cancer onset and the speed of aging-is interlinked with signal transduction by dietary molecules. The underlying nutrigenomic principles will offer guidance for healthy aging in individuals with Fanconi anemia as well as for the general population.
Collapse
Affiliation(s)
- Eunike Velleuer
- Department for Cytopathology, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany;
- Department for Pediatric Hemato-Oncology, Helios Children’s Hospital, D-47805 Krefeld, Germany
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, PL-10-748 Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
24
|
Maia CMDA, Vasconcelos PGS, Pasetto S, Godwin WC, Silva JPRE, Tavares JF, Pardi V, Costa EMMDB, Murata RM. Anadenanthera colubrina regulated LPS-induced inflammation by suppressing NF-κB and p38-MAPK signaling pathways. Sci Rep 2024; 14:16028. [PMID: 38992070 PMCID: PMC11239917 DOI: 10.1038/s41598-024-66590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
We aimed to determine the chemical profile and unveil Anadenanthera colubrina (Vell.) Brenan standardized extract effects on inflammatory cytokines expression and key proteins from immunoregulating signaling pathways on LPS-induced THP-1 monocyte. Using the RT-PCR and Luminex Assays, we planned to show the gene expression and the levels of IL-8, IL-1β, and IL-10 inflammatory cytokines. Key proteins of NF-κB and MAPK transduction signaling pathways (NF-κB, p-38, p-NF-κB, and p-p38) were detected by Simple Western. Using HPLC-ESI-MSn (High-Performance Liquid-Chromatography) and HPLC-HRESIMS, we showed the profile of the extract that includes an opus of flavonoids, including the catechins, quercetin, kaempferol, and the proanthocyanidins. Cell viability was unaffected up to 250 µg/mL of the extract (LD50 = 978.7 µg/mL). Thereafter, the extract's impact on the cytokine became clear. Upon LPS stimuli, in the presence of the extract, gene expression of IL-1β and IL-10 were downregulated and the cytokines expression of IL-1β and IL-10 were down an upregulated respectively. The extract is involved in TLR-4-related NF-κB/MAPK pathways; it ignited phosphorylation of p38 and NF-κB, orchestrating a reduced signal intensity. Therefore, Anadenanthera colubrina's showed low cytotoxicity and profound influence as a protector against the inflammation, modulating IL-1β and IL-10 inflammatory cytokines gene expression and secretion by regulating intracellular NF-κB and p38-MAPK signaling pathways.
Collapse
Affiliation(s)
- Carolina Medeiros de Almeida Maia
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Paraiba, Brazil
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA
| | | | - Silvana Pasetto
- Department of Biology, East Carolina University, Greenville, NC, USA
| | - Walton Colby Godwin
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA
| | - Joanda Paolla Raimundo E Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, Paraiba, Brazil
| | - Josean Fechine Tavares
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, Paraiba, Brazil
| | - Vanessa Pardi
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA
| | - Edja Maria Melo de Brito Costa
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Paraiba, Brazil.
| | - Ramiro Mendonça Murata
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
25
|
Alfaifi AA, Holm JB, Wang TW, Lim J, Meiller TF, Rock P, Sultan AS, Jabra-Rizk MA. Oral Microbiota Alterations in Subjects with SARS-CoV-2 Displaying Prevalence of the Opportunistic Fungal Pathogen Candida albicans. Microorganisms 2024; 12:1356. [PMID: 39065125 PMCID: PMC11278750 DOI: 10.3390/microorganisms12071356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/19/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The oral cavity remains an underappreciated site for SARS-CoV-2 infection despite the myriad of oral conditions in COVID-19 patients. Recently, SARS-CoV-2 was shown to replicate in the salivary gland cells causing tissue inflammation. Given the established association between inflammation and microbiome disruption, we comparatively profiled oral microbial differences at a metagenomic level in a cohort of hospitalized COVID-19 patients and matched healthy controls. Specifically, we aimed to evaluate colonization by the opportunistic fungal pathogen Candida albicans, the etiologic agent of oral candidiasis. Comprehensive shotgun metagenomic analysis indicated that, overall, COVID-19 patients exhibited significantly reduced bacterial and viral diversity/richness; we identified 12 differentially abundant bacterial species to be negatively associated with COVID-19, and the functional pathways of certain bacteria to be highly associated with COVID-19 status. Strikingly, C. albicans was recovered from approximately half of the COVID-19 subjects but not from any of the healthy controls. The prevalence of Candida is likely linked to immune hypo-dysregulation caused by COVID-19 favoring Candida proliferation, warranting investigations into the interplay between Candida and SARS-CoV2 and potential therapeutic approaches directed toward oral candidiasis. Collectively, our findings prompt a reassessment of oral opportunistic infection risks during COVID-19 disease and their potential long-term impacts on oral health.
Collapse
Affiliation(s)
- Areej A. Alfaifi
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD 21201, USA; (A.A.A.); (T.W.W.); (T.F.M.); (A.S.S.)
- Department of Restorative and Prosthetic Dental Sciences, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
| | - Johanna B. Holm
- Institute for Genome Sciences, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (J.B.H.); (J.L.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Tristan W. Wang
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD 21201, USA; (A.A.A.); (T.W.W.); (T.F.M.); (A.S.S.)
| | - Jonathan Lim
- Institute for Genome Sciences, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (J.B.H.); (J.L.)
| | - Timothy F. Meiller
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD 21201, USA; (A.A.A.); (T.W.W.); (T.F.M.); (A.S.S.)
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Peter Rock
- Department of Anesthesia, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| | - Ahmed S. Sultan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD 21201, USA; (A.A.A.); (T.W.W.); (T.F.M.); (A.S.S.)
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD 21201, USA; (A.A.A.); (T.W.W.); (T.F.M.); (A.S.S.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
26
|
Cinicola BL, Uva A, Duse M, Zicari AM, Buonsenso D. Mucocutaneous Candidiasis: Insights Into the Diagnosis and Treatment. Pediatr Infect Dis J 2024; 43:694-703. [PMID: 38502882 PMCID: PMC11191067 DOI: 10.1097/inf.0000000000004321] [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] [Accepted: 02/08/2024] [Indexed: 03/21/2024]
Abstract
Recent progress in the methods of genetic diagnosis of inborn errors of immunity has contributed to a better understanding of the pathogenesis of chronic mucocutaneous candidiasis (CMC) and potential therapeutic options. This review describes the latest advances in the understanding of the pathophysiology, diagnostic strategies, and management of chronic mucocutaneous candidiasis.
Collapse
Affiliation(s)
- Bianca Laura Cinicola
- From the Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Uva
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Marzia Duse
- From the Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Anna Maria Zicari
- From the Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Center for Global Health Research and Studies, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
27
|
Bhumitrakul J, Lam-Ubol A, Matangkasombut O. Oral Candida in post-radiotherapy patients with xerostomia/hyposalivation: A narrative review. Oral Dis 2024. [PMID: 38946209 DOI: 10.1111/odi.15060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 07/02/2024]
Abstract
OBJECTIVE Head and Neck Cancer (HNC) patients receiving radiotherapy (RT) often suffer from xerostomia and/or hyposalivation. As saliva plays an important antimicrobial and cleansing roles, these patients are at higher risks of opportunistic infections. This narrative review aims to provide an overview of current evidence on oral Candida colonisation and infection in these patients. METHODS A literature review of clinical studies on oral Candida colonisation and candidiasis in HNC patients receiving radiotherapy/chemoradiotherapy was conducted. RESULTS Many clinical studies found high levels of Candida colonisation and a substantial proportion of post-RT HNC patients suffering from oropharyngeal candidiasis (OPC). Importantly, oral Candida could be a reservoir for life-threatening systemic infection in immunocompromised patients. The rising prevalence of non-albicans Candida species and drug-resistant infections has made identification of Candida species and antifungal susceptibility more important. Recent advances in oral microbiome and its interactions with Candida are discussed. This review also offers perspectives on limitations of current evidence and suggestions for future research. CONCLUSION Further research to better understand Candida carriage, microbiome, OPC, and xerostomia/hyposalivation post-RT would aid in devising a more comprehensive long-term management plan and novel therapeutic approaches for HNC patients to achieve the full benefits of RT while minimising side effects.
Collapse
Affiliation(s)
- Jom Bhumitrakul
- King's College London GKT School of Medical Education, King's College London, London, UK
| | - Aroonwan Lam-Ubol
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Oranart Matangkasombut
- Department of Microbiology and Center of Excellence on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Research Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| |
Collapse
|
28
|
Barbosa PF, Gonçalves DS, Ramos LS, Mello TP, Braga-Silva LA, Pinto MR, Taborda CP, Branquinha MH, Santos ALS. Saps1-3 Antigens in Candida albicans: Differential Modulation Following Exposure to Soluble Proteins, Mammalian Cells, and Infection in Mice. Infect Dis Rep 2024; 16:572-586. [PMID: 39051243 PMCID: PMC11270244 DOI: 10.3390/idr16040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The secreted aspartic peptidases (Saps) of Candida albicans play crucial roles in various steps of fungal-host interactions. Using a flow cytometry approach, this study investigated the expression of Saps1-3 antigens after (i) incubation with soluble proteins, (ii) interaction with mammalian cells, and (iii) infection in immunosuppressed BALB/c mice. Supplementation strategies involving increasing concentrations of bovine serum albumin (BSA) added to yeast carbon base (YCB) medium as the sole nitrogenous source revealed a positive and significant correlation between BSA concentration and both the growth rate and the percentage of fluorescent cells (%FC) labeled with anti-Saps1-3 antibodies. Supplementing the YCB medium with various soluble proteins significantly modulated the expression of Saps1-3 antigens in C. albicans. Specifically, immunoglobulin G, gelatin, and total bovine/human sera significantly reduced the %FC, while laminin, human serum albumin, fibrinogen, hemoglobin, and mucin considerably increased the %FC compared to BSA. Furthermore, co-cultivating C. albicans yeasts with either live epithelial or macrophage cells induced the expression of Saps1-3 antigens in 78% (mean fluorescence intensity [MFI] = 152.1) and 82.7% (MFI = 178.2) of the yeast cells, respectively, compared to BSA, which resulted in 29.3% fluorescent cells (MFI = 50.9). Lastly, the yeasts recovered from the kidneys of infected immunosuppressed mice demonstrated a 4.8-fold increase in the production of Saps1-3 antigens (MFI = 246.6) compared to BSA, with 95.5% of yeasts labeled with anti-Saps1-3 antibodies. Altogether, these results demonstrated the positive modulation of Saps' expression in C. albicans by various key host proteinaceous components, as well as by in vitro and in vivo host challenges.
Collapse
Affiliation(s)
- Pedro F. Barbosa
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Diego S. Gonçalves
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Thaís P. Mello
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Lys A. Braga-Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Marcia R. Pinto
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense (UFF), Niterói 24210-130, Brazil;
| | - Carlos P. Taborda
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo 05508-060, Brazil;
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-901, Brazil
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-901, Brazil
| |
Collapse
|
29
|
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
|
30
|
Li S, Xv Y, Sun Y, Shen Z, Hao R, Yan J, Liu M, Liu Z, Jing T, Li X, Zhang X. Macrophage-derived CD36 + exosome subpopulations as novel biomarkers of Candida albicans infection. Sci Rep 2024; 14:14723. [PMID: 38926392 PMCID: PMC11208550 DOI: 10.1038/s41598-024-60032-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/18/2024] [Indexed: 06/28/2024] Open
Abstract
Invasive candidiasis (IC) is a notable healthcare-associated fungal infection, characterized by high morbidity, mortality, and substantial treatment costs. Candida albicans emerges as a principal pathogen in this context. Recent academic advancements have shed light on the critical role of exosomes in key biological processes, such as immune responses and antigen presentation. This burgeoning body of research underscores the potential of exosomes in the realm of medical diagnostics and therapeutics, particularly in relation to fungal infections like IC. The exploration of exosomal functions in the pathophysiology of IC not only enhances our understanding of the disease but also opens new avenues for innovative therapeutic interventions. In this investigation, we focus on exosomes (Exos) secreted by macrophages, both uninfected and those infected with C. albicans. Our objective is to extract and analyze these exosomes, delving into the nuances of their protein compositions and subgroups. To achieve this, we employ an innovative technique known as Proximity Barcoding Assay (PBA). This methodology is pivotal in our quest to identify novel biological targets, which could significantly enhance the diagnostic and therapeutic approaches for C. albicans infection. The comparative analysis of exosomal contents from these two distinct cellular states promises to yield insightful data, potentially leading to breakthroughs in understanding and treating this invasive fungal infection. In our study, we analyzed differentially expressed proteins in exosomes from macrophages and C. albicans -infected macrophages, focusing on proteins such as ACE2, CD36, CAV1, LAMP2, CD27, and MPO. We also examined exosome subpopulations, finding a dominant expression of MPO in the most prevalent subgroup, and a distinct expression of CD36 in cluster14. These findings are crucial for understanding the host response to C. albicans and may inform targeted diagnostic and therapeutic approaches. Our study leads us to infer that MPO and CD36 proteins may play roles in the immune escape mechanisms of C. albicans. Additionally, the CD36 exosome subpopulations, identified through our analysis, could serve as potential biomarkers and therapeutic targets for C. albicans infection. This insight opens new avenues for understanding the infection's pathology and developing targeted treatments.
Collapse
Affiliation(s)
- Shuo Li
- Clinical Medical College of Hebei University of Engineering, Handan, 056000, China
| | - Yanyan Xv
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China
| | - Yuanyuan Sun
- Hebei Medical University, Shijiazhuang, 050000, China
| | - Ziyi Shen
- The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ruiying Hao
- Clinical Medical College of Hebei University of Engineering, Handan, 056000, China
| | - Jingjing Yan
- Clinical Medical College of Hebei University of Engineering, Handan, 056000, China
| | - Mengru Liu
- Clinical Medical College of Hebei University of Engineering, Handan, 056000, China
| | - Zhao Liu
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China
| | - Tingting Jing
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China
| | - Xiaojing Li
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China.
| | - Xiujuan Zhang
- Department of Laboratory, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China.
| |
Collapse
|
31
|
Liang YX, Wang YK, Meng WJ, Wang Q, Li JX, Huang WH, Xie M. Microfluidic Electrochemical Integrated Sensor for Efficient and Sensitive Detection of Candida albicans. Anal Chem 2024; 96:10013-10020. [PMID: 38836548 DOI: 10.1021/acs.analchem.4c01419] [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: 06/06/2024]
Abstract
Traditional methods for the detection of pathogenic bacteria are time-consuming, less efficient, and sensitive, which affects infection control and bungles illness. Therefore, developing a method to remedy these problems is very important in the clinic to diagnose the pathogenic diseases and guide the rational use of antibiotics. Here, microfluidic electrochemical integrated sensor (MEIS) has been investigated, functionally for rapid, efficient separation and sensitive detection of pathogenic bacteria. Three-dimensional macroporous PDMS and Au nanotube-based electrode are successfully assembled into the modeling microchip, playing the functions of "3D chaotic flow separator" and "electrochemical detector," respectively. The 3D chaotic flow separator enhances the turbulence of the fluid, achieving an excellent bacteria capture efficiency. Meanwhile, the electrochemical detector provides a quantitative signal through enzyme-linked immunoelectrochemistry with improved sensitivity. The microfluidic electrochemical integrated sensor could successfully isolate Candida albicans (C. albicans) in the range of 30-3,000,000 CFU in the saliva matrix with over 95% capture efficiency and sensitively detect C. albicans in 1 h in oral saliva samples. The integrated device demonstrates great potential in the diagnosis of oral candidiasis and is also applicable in the detection of other pathogenic bacteria.
Collapse
Affiliation(s)
- Ying-Xue Liang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yi-Ke Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Jie Meng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Qian Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jia-Xin Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Min Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
32
|
Chand P, Narula K, Vs R, Sharma S, Kumari S, Mondal N, Singh SP, Mishra P, Prasad T. Mechanistic Insights into Cellular and Molecular Targets of Zinc Oxide Quantum Dots (ZnO QDs) in Fungal Pathogen, Candida albicans: One Drug Multi-Targeted Therapeutic Approach. ACS Infect Dis 2024; 10:1914-1934. [PMID: 38831663 DOI: 10.1021/acsinfecdis.3c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Rationally designed multitargeted drugs, known as network therapeutics/multimodal drugs, have emerged as versatile therapeutic solutions to combat drug-resistant microbes. Here, we report novel mechanistic insights into cellular and molecular targets of ZnO quantum dots (QDs) against Candida albicans, a representative of fungal pathogens. Stable, monodispersed 4-6 nm ZnO QDs were synthesized using a wet chemical route, which exhibited dose-dependent inhibition on the growth dynamics of Candida. Treatment with 200 μg/mL ZnO QDs revealed an aberrant morphology and a disrupted cellular ultrastructure in electron microscopy and led to a 23% reduction in ergosterol content and a 53% increase in intracellular reactive oxygen species. Significant increase in steady-state fluorescence polarization and fluorescence lifetime decay of membrane probe 1,6-diphenyl-1,3,5-hexatriene (DPH) in treated cells, respectively, implied reduction in membrane fluidity and enhanced microviscosity. The observed reduction in passive diffusion of fluorescent Rhodamine 6G across the membrane validated the intricate relationship between ergosterol, membrane fluidity, and microviscosity. An inverse relationship existing between ergosterol biosynthetic genes, ERG11 and ERG3 in treated cells, related well with displayed higher susceptibilities. Furthermore, treated cells exhibited impaired functionality and downregulation of ABC drug efflux pumps. Multiple cellular targets of ZnO QDs in Candida were validated by in silico molecular docking. Thus, targeting ERG11, ERG3, and ABC drug efflux pumps might emerge as a versatile, nano-ZnO-based strategy in fungal therapeutics to address the challenges of drug resistance.
Collapse
Affiliation(s)
- Preeti Chand
- Special Centre for Nano Sciences and AIRF, Jawaharlal Nehru University, New Delhi 110067, India
| | - Kritika Narula
- Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Radhakrishnan Vs
- Special Centre for Nano Sciences and AIRF, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shubham Sharma
- Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sangeeta Kumari
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Neelima Mondal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Surinder P Singh
- CSIR-National Physical Laboratory, Dr. K. S. Krishnan Marg, New Delhi 110012, India
| | - Prashant Mishra
- Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Tulika Prasad
- Special Centre for Nano Sciences and AIRF, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
33
|
Di Bella S, Sanson G, Monticelli J, Zerbato V, Principe L, Giuffrè M, Pipitone G, Luzzati R. Clostridioides difficile infection: history, epidemiology, risk factors, prevention, clinical manifestations, treatment, and future options. Clin Microbiol Rev 2024; 37:e0013523. [PMID: 38421181 PMCID: PMC11324037 DOI: 10.1128/cmr.00135-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
SUMMARYClostridioides difficile infection (CDI) is one of the major issues in nosocomial infections. This bacterium is constantly evolving and poses complex challenges for clinicians, often encountered in real-life scenarios. In the face of CDI, we are increasingly equipped with new therapeutic strategies, such as monoclonal antibodies and live biotherapeutic products, which need to be thoroughly understood to fully harness their benefits. Moreover, interesting options are currently under study for the future, including bacteriophages, vaccines, and antibiotic inhibitors. Surveillance and prevention strategies continue to play a pivotal role in limiting the spread of the infection. In this review, we aim to provide the reader with a comprehensive overview of epidemiological aspects, predisposing factors, clinical manifestations, diagnostic tools, and current and future prophylactic and therapeutic options for C. difficile infection.
Collapse
Affiliation(s)
- Stefano Di Bella
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| | - Gianfranco Sanson
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| | - Jacopo Monticelli
- Infectious Diseases
Unit, Trieste University Hospital
(ASUGI), Trieste,
Italy
| | - Verena Zerbato
- Infectious Diseases
Unit, Trieste University Hospital
(ASUGI), Trieste,
Italy
| | - Luigi Principe
- Microbiology and
Virology Unit, Great Metropolitan Hospital
“Bianchi-Melacrino-Morelli”,
Reggio Calabria, Italy
| | - Mauro Giuffrè
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
- Department of Internal
Medicine (Digestive Diseases), Yale School of Medicine, Yale
University, New Haven,
Connecticut, USA
| | - Giuseppe Pipitone
- Infectious Diseases
Unit, ARNAS Civico-Di Cristina
Hospital, Palermo,
Italy
| | - Roberto Luzzati
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| |
Collapse
|
34
|
Xu Z, Li Y, Xu A, Xue L, Soteyome T, Yuan L, Ma Q, Seneviratne G, Hong W, Mao Y, Kjellerup BV, Liu J. Differential alteration in Lactiplantibacillus plantarum subsp. plantarum quorum-sensing systems and reduced Candida albicans yeast survival and virulence gene expression in dual-species interaction. Microbiol Spectr 2024; 12:e0035324. [PMID: 38717160 PMCID: PMC11237386 DOI: 10.1128/spectrum.00353-24] [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: 02/08/2024] [Accepted: 04/15/2024] [Indexed: 06/06/2024] Open
Abstract
Candida albicans (C. albicans) and Lactiplantibacillus plantarum subsp. plantarum (L. plantarum) are frequently identified in various niches, but their dual-species interaction, especially with C. albicans in yeast form, remains unclear. This study aimed to investigate the dual-species interaction of L. plantarum and C. albicans, including proliferation, morphology, and transcriptomes examined by selective agar plate counting, microscopy, and polymicrobial RNA-seq, respectively. Maintaining a stable and unchanged growth rate, L. plantarum inhibited C. albicans yeast cell proliferation but not hyphal growth. Combining optical microscopy and atomic force microscopy, cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed during dual-species interaction. Reduced C. albicans yeast cell proliferation in mixed culture was partially due to L. plantarum cell-free culture supernatant but not the acidic environment. Upon polymicrobial transcriptomics analysis, interesting changes were identified in both L. plantarum and C. albicans gene expression. First, two L. plantarum quorum-sensing systems showed contrary changes, with the activation of lamBDCA and repression of luxS. Second, the upregulation of stress response-related genes and downregulation of cell cycle, cell survival, and cell integrity-related pathways were identified in C. albicans, possibly connected to the stress posed by L. plantarum and the reduced yeast cell proliferation. Third, a large scale of pathogenesis and virulence factors were downregulated in C. albicans, indicating the potential interruption of pathogenic activities by L. plantarum. Fourth, partial metabolism and transport pathways were changed in L. plantarum and C. albicans. The information in this study might aid in understanding the behavior of L. plantarum and C. albicans in dual-species interaction.IMPORTANCEThe anti-Candida albicans activity of Lactiplantibacillus plantarum has been explored in the past decades. However, the importance of C. albicans yeast form and the effect of C. albicans on L. plantarum had also been omitted. In this study, the dual-species interaction of L. plantarum and C. albicans was investigated with a focus on the transcriptomes. Cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed. Upon polymicrobial transcriptomics analysis, interesting changes were identified, including contrary changes in two L. plantarum quorum-sensing systems and reduced cell survival-related pathways and pathogenesis determinants in C. albicans.
Collapse
Affiliation(s)
- Zhenbo Xu
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yaqin Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou, China
| | - Liang Xue
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, China, Guangzhou, Guangdong
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qin Ma
- Key Laboratory of Functional Foods, Ministry of Agriculture, Guangdong Key Laboratory of Agricultural Products Processing, Sericultural and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | | | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuzhu Mao
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Birthe V. Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Junyan Liu
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, College of Light Industry and Food Science, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
35
|
Sahu SR, Dutta A, Peroumal D, Kumari P, Utakalaja BG, Patel SK, Acharya N. Immunogenicity and efficacy of CNA25 as a potential whole-cell vaccine against systemic candidiasis. EMBO Mol Med 2024; 16:1254-1283. [PMID: 38783167 PMCID: PMC11178797 DOI: 10.1038/s44321-024-00080-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Disseminated fungal infections account for ~1.5 million deaths per year worldwide, and mortality may increase further due to a rise in the number of immunocompromised individuals and drug-resistance fungal species. Since an approved antifungal vaccine is yet to be available, this study explored the immunogenicity and vaccine efficacy of a DNA polymerase mutant strain of Candida albicans. CNA25 is a pol32ΔΔ strain that exhibits growth defects and does not cause systemic candidiasis in mice. Immunized mice with live CNA25 were fully protected against C. albicans and C. parapsilosis but partially against C. tropicalis and C. glabrata infections. CNA25 induced steady expression of TLR2 and Dectin-1 receptors leading to a faster recognition and clearance by the immune system associated with the activation of protective immune responses mostly mediated by neutrophils, macrophages, NK cells, B cells, and CD4+ and CD8+ T cells. Molecular blockade of Dectin-1, IL-17, IFNγ, and TNFα abolished resistance to reinfection. Altogether, this study suggested that CNA25 collectively activates innate, adaptive, and trained immunity to be a promising live whole-cell vaccine against systemic candidiasis.
Collapse
Affiliation(s)
- Satya Ranjan Sahu
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Abinash Dutta
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Doureradjou Peroumal
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Premlata Kumari
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Bhabasha Gyanadeep Utakalaja
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Shraddheya Kumar Patel
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
36
|
Garcia-Bustos V, Rosario Medina I, Cabañero Navalón MD, Ruiz Gaitán AC, Pemán J, Acosta-Hernández B. Candida spp. in Cetaceans: Neglected Emerging Challenges in Marine Ecosystems. Microorganisms 2024; 12:1128. [PMID: 38930510 PMCID: PMC11205550 DOI: 10.3390/microorganisms12061128] [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: 05/13/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Cetaceans, which are crucial in marine ecosystems, act as sentinels for ecosystem and human-environmental health. However, emerging fungal infections, particularly by Candida spp., pose a growing concern in these marine mammals. This review consolidates current knowledge on the prevalence, clinical manifestations, species distribution, and antifungal resistance of Candida infections in cetaceans. We detail the diverse pathogenic impacts of Candida, including respiratory, dermal, and systemic afflictions, underscoring diagnostic and treatment challenges amid rising antifungal resistance. Our analysis extends beyond health concerns in captive cetaceans, where confinement stress heightens vulnerability, to encompass substantial ecological risks in wild populations. The review emphasizes the One Health perspective, linking cetacean health with broader environmental and human public health issues. We particularly focus on the potential zoonotic transmission of emerging fungal pathogens such as Candida auris and the role of environmental changes in fostering antifungal resistance. The study underscores the need for concerted, interdisciplinary efforts in veterinary, medical, and environmental sciences to enhance understanding and management of Candida infections in cetaceans. We advocate for comprehensive monitoring and collaborative research initiatives to mitigate the rising challenge of these infections. Addressing Candida spp. in cetaceans is not just a conservation priority but a critical step in safeguarding overall marine health and, by extension, human health in the context of evolving infectious diseases.
Collapse
Affiliation(s)
- Victor Garcia-Bustos
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Universidad de Las Palmas de Gran Canaria, 35001 Arucas, Spain;
- Severe Infection Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (M.D.C.N.); (A.C.R.G.); (J.P.)
| | - Inmaculada Rosario Medina
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Universidad de Las Palmas de Gran Canaria, 35001 Arucas, Spain;
| | - Marta Dafne Cabañero Navalón
- Severe Infection Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (M.D.C.N.); (A.C.R.G.); (J.P.)
| | - Alba Cecilia Ruiz Gaitán
- Severe Infection Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (M.D.C.N.); (A.C.R.G.); (J.P.)
| | - Javier Pemán
- Severe Infection Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (M.D.C.N.); (A.C.R.G.); (J.P.)
| | - Begoña Acosta-Hernández
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Universidad de Las Palmas de Gran Canaria, 35001 Arucas, Spain;
| |
Collapse
|
37
|
Yang Z, Zhang S, Ji N, Li J, Chen Q. The evil companion of OSCC: Candida albicans. Oral Dis 2024; 30:1873-1886. [PMID: 37530513 DOI: 10.1111/odi.14700] [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: 05/18/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE Microbial dysbiosis and microbiome-induced inflammation may play a role in the etiopathogenesis of oral squamous cell carcinoma (OSCC). Candida albicans (C. albicans) is the most prevalent opportunistic pathogenic fungus in the oral cavity, and Candida infection is considered as one of its high-risk factors. Although oral microbiota-host interactions are closely associated with the development of OSCC, the interrelationship between fungi and OSCC is poorly understood compared to that between bacteria and viruses. RESULTS We accumulated knowledge of the evidence, pathogenic factors, and possible multiple mechanisms by which C. albicans promotes malignant transformation of OSCC, focusing on the induction of epithelial damage, production of carcinogens, and regulation of the tumor microenvironment. In addition, we highlight the latest treatment strategies for Candida infection. CONCLUSION This review provides a new perspective on the interrelationship between C. albicans and OSCC and contributes to the establishment of a systematic and reliable clinical treatment system for OSCC patients with C. albicans infection.
Collapse
Affiliation(s)
- Zhixin Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Shiyu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| |
Collapse
|
38
|
Nenciarini S, Renzi S, di Paola M, Meriggi N, Cavalieri D. Ascomycetes yeasts: The hidden part of human microbiome. WIREs Mech Dis 2024; 16:e1641. [PMID: 38228159 DOI: 10.1002/wsbm.1641] [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: 05/17/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
The fungal component of the microbiota, the mycobiota, has been neglected for a long time due to its poor richness compared to bacteria. Limitations in fungal detection and taxonomic identification arise from using metagenomic approaches, often borrowed from bacteriome analyses. However, the relatively recent discoveries of the ability of fungi to modulate the host immune response and their involvement in human diseases have made mycobiota a fundamental component of the microbial communities inhabiting the human host, deserving some consideration in host-microbe interaction studies and in metagenomics. Here, we reviewed recent data on the identification of yeasts of the Ascomycota phylum across human body districts, focusing on the most representative genera, that is, Saccharomyces and Candida. Then, we explored the key factors involved in shaping the human mycobiota across the lifespan, ranging from host genetics to environment, diet, and lifestyle habits. Finally, we discussed the strengths and weaknesses of culture-dependent and independent methods for mycobiota characterization. Overall, there is still room for some improvements, especially regarding fungal-specific methodological approaches and bioinformatics challenges, which are still critical steps in mycobiota analysis, and to advance our knowledge on the role of the gut mycobiota in human health and disease. This article is categorized under: Immune System Diseases > Genetics/Genomics/Epigenetics Immune System Diseases > Environmental Factors Infectious Diseases > Environmental Factors.
Collapse
Affiliation(s)
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | - Monica di Paola
- Department of Biology, University of Florence, Florence, Italy
| | - Niccolò Meriggi
- Department of Biology, University of Florence, Florence, Italy
| | | |
Collapse
|
39
|
Spacova I, Allonsius CN, De Boeck I, Oerlemans E, Tuyaerts I, Van de Vliet N, van den Broek MFL, Jimenez L, Boyer M, Rodriguez B, Ballet N, Lebeer S. Multifactorial inhibition of Candida albicans by combinations of lactobacilli and probiotic Saccharomyces cerevisiae CNCM I-3856. Sci Rep 2024; 14:9365. [PMID: 38654026 DOI: 10.1038/s41598-024-59869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Strategies against the opportunistic fungal pathogen Candida albicans based on probiotic microorganisms represent a promising alternative to traditional antifungals. Here, we investigated the effects of Lactobacillaceae isolates from fermented foods or the human vagina, alone or in combination with the probiotic yeast Saccharomyces cerevisiae CNCM I-3856, against C. albicans in vitro. Nine out of nineteen tested strains of Lactobacillaceae inhibited growth of C. albicans with inhibition zones of 1-3 mm in spot assays. Five out of nineteen lactobacilli tested as such or in combination with S. cerevisiae CNCM I-3856 also significantly inhibited C. albicans hyphae formation, including Limosilactobacillus fermentum LS4 and L. fermentum LS5 resulting in respectively 62% and 78% hyphae inhibition compared to the control. Thirteen of the tested nineteen lactobacilli aggregated with the yeast form of C. albicans, with Lactiplantibacillus carotarum AMBF275 showing the strongest aggregation. The aggregation was enhanced when lactobacilli were combined with S. cerevisiae CNCM I-3856. No significant antagonistic effects were observed between the tested lactobacilli and S. cerevisiae CNCM I-3856. The multifactorial activity of Lactobacillaceae strains alone or combined with the probiotic S. cerevisiae CNCM I-3856 against C. albicans without antagonistic effects between the beneficial strains, paves the way for developing consortium probiotics for in vivo applications.
Collapse
Affiliation(s)
- Irina Spacova
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Camille Nina Allonsius
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Ilke De Boeck
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Eline Oerlemans
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Ines Tuyaerts
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Nele Van de Vliet
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Marianne F L van den Broek
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Luciana Jimenez
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Mickaël Boyer
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Bertrand Rodriguez
- Gnosis by Lesaffre, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Nathalie Ballet
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Sarah Lebeer
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium.
| |
Collapse
|
40
|
Moglad E, Elekhnawy E, Negm WA, Mokhtar FA, Binsuwaidan R, Attallah NGM, Ahmed E, Magdeldin S, Al-Fakhrany OM. Evaluation of Tamarix nilotica Fractions in Combating Candida albicans Infections. Expert Rev Anti Infect Ther 2024; 22:241-251. [PMID: 37387417 DOI: 10.1080/14787210.2023.2232112] [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: 03/10/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
OBJECTIVES Evaluation of the antifungal properties of Tamarix nilotica fractions against Candida albicans clinical isolates. METHODS The in vitro antifungal potential was evaluated by agar well diffusion and broth microdilution methods. The antibiofilm potential was assessed by crystal violet, scanning electron microscopy (SEM), and qRT-PCR. The in vivo antifungal activity was evaluated by determining the burden in the lung tissues of infected mice, histopathological, immunohistochemical studies, and ELISA. RESULTS Both the dichloromethane (DCM) and ethyl acetate (EtOAc) fractions had minimum inhibitory concentration (MIC) values of 64-256 and 128-1024 μg/mL, respectively. SEM examination showed that the DCM fraction decreased the biofilm formation capacity of the treated isolates. A significant decline in biofilm gene expression was observed in 33.33% of the DCM-treated isolates. A considerable decline in the CFU/g lung count in infected mice was observed, and histopathological examinations revealed that the DCM fraction maintained the lung tissue architecture. Immunohistochemical investigations indicated that the DCM fraction significantly (p < 0.05) decreased the expression of pro-inflammatory and inflammatory cytokines (TNF-α, NF-kB, COX-2, IL-6, and IL-1β) in the immunostained lung sections. The phytochemical profiling of DCM and EtOAc fractions was performed using Liquid chromatography-mass spectrometry (LC-ESI-MS/MS). CONCLUSION T. nilotica DCM fraction could be a significant source of natural products with antifungal activity against C. albicans infections.
Collapse
Affiliation(s)
- Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Fatma A Mokhtar
- Department of Pharmacognosy, Faculty of Pharmacy, El Saleheya El Gadida University, El Saleheya El Gadida, Egypt
| | - Reem Binsuwaidan
- Department of Pharmaceutical Science, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Eman Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital, Cairo, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital, Cairo, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | | |
Collapse
|
41
|
Vazquez-Munoz R, Thompson A, Sobue T, Dongari-Bagtzoglou A. Powder diet exacerbates oropharyngeal candidiasis in a mouse model. Appl Environ Microbiol 2024; 90:e0171323. [PMID: 38319097 PMCID: PMC10952443 DOI: 10.1128/aem.01713-23] [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: 09/28/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
This study reports on the influence of a powder diet in a mouse model of oropharyngeal candidiasis (OPC), a significant health concern caused primarily by Candida albicans. Despite identical nutritional composition, we found that a powdered diet significantly increased Candida burdens and oral lesions, and aggravated weight loss compared to a standard pelleted diet. High fungal burdens and severe oral lesions were accomplished within 48 hours after infection with only one dose of cortisone. Moreover, mice on a powder diet recovered a week after infection. Using a powder diet, we thus modified the cortisone OPC murine model in a way that simplifies the infection process, enhances reproducibility, and facilitates studies investigating both pathogenesis and recovery processes. Our findings also underscore the pivotal role of the physical form of the diet in the progression and severity of oral Candida infection in this model. Future research should investigate this relationship further to broaden our understanding of the underlying mechanisms, potentially leading to novel prevention strategies and improved disease management.IMPORTANCEOropharyngeal candidiasis (OPC) is a multifactorial disease and a significant health concern. We found that the physical form of the diet plays a critical role in the severity and progression of OPC. We developed a modified cortisone OPC murine model that facilitates studies investigating pathogenesis and recovery processes.
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
|
42
|
Le PH, Linklater DP, Medina AA, MacLaughlin S, Crawford RJ, Ivanova EP. Impact of multiscale surface topography characteristics on Candida albicans biofilm formation: From cell repellence to fungicidal activity. Acta Biomater 2024; 177:20-36. [PMID: 38342192 DOI: 10.1016/j.actbio.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/21/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
While there has been significant research conducted on bacterial colonization on implant materials, with a focus on developing surface modifications to prevent the formation of bacterial biofilms, the study of Candida albicans biofilms on implantable materials is still in its infancy, despite its growing relevance in implant-associated infections. C. albicans fungal infections represent a significant clinical concern due to their severity and associated high fatality rate. Pathogenic yeasts account for an increasing proportion of implant-associated infections, since Candida spp. readily form biofilms on medical and dental device surfaces. In addition, these biofilms are highly antifungal-resistant, making it crucial to explore alternative solutions for the prevention of Candida implant-associated infections. One promising approach is to modify the surface properties of the implant, such as the wettability and topography of these substrata, to prevent the initial Candida attachment to the surface. This review summarizes recent research on the effects of surface wettability, roughness, and architecture on Candida spp. attachment to implantable materials. The nanofabrication of material surfaces are highlighted as a potential method for the prevention of Candida spp. attachment and biofilm formation on medical implant materials. Understanding the mechanisms by which Candida spp. attach to surfaces will allow such surfaces to be designed such that the incidence and severity of Candida infections in patients can be significantly reduced. Most importantly, this approach could also substantially reduce the need to use antifungals for the prevention and treatment of these infections, thereby playing a crucial role in minimizing the possibility contributing to instances of antimicrobial resistance. STATEMENT OF SIGNIFICANCE: In this review we provide a systematic analysis of the role that surface characteristics, such as wettability, roughness, topography and architecture, play on the extent of C. albicans cells attachment that will occur on biomaterial surfaces. We show that exploiting bioinspired surfaces could significantly contribute to the prevention of antimicrobial resistance to antifungal and chemical-based preventive measures. By reducing the attachment and growth of C. albicans cells using surface structure approaches, we can decrease the need for antifungals, which are conventionally used to treat such infections.
Collapse
Affiliation(s)
- Phuc H Le
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia; ARC Research Hub for Australian Steel Manufacturing, Melbourne, VIC 3001, Australia
| | - Denver P Linklater
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia; ARC Research Hub for Australian Steel Manufacturing, Melbourne, VIC 3001, Australia; Department of Biomedical Engineering, The Graeme Clark Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | - Arturo Aburto Medina
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia
| | - Shane MacLaughlin
- ARC Research Hub for Australian Steel Manufacturing, Melbourne, VIC 3001, Australia; BlueScope Steel Research, Port Kembla, NSW 2505, Australia
| | - Russell J Crawford
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia
| | - Elena P Ivanova
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia; ARC Research Hub for Australian Steel Manufacturing, Melbourne, VIC 3001, Australia.
| |
Collapse
|
43
|
Valentine M, Rudolph P, Dietschmann A, Tsavou A, Mogavero S, Lee S, Priest EL, Zhurgenbayeva G, Jablonowski N, Timme S, Eggeling C, Allert S, Dolk E, Naglik JR, Figge MT, Gresnigt MS, Hube B. Nanobody-mediated neutralization of candidalysin prevents epithelial damage and inflammatory responses that drive vulvovaginal candidiasis pathogenesis. mBio 2024; 15:e0340923. [PMID: 38349176 PMCID: PMC10936171 DOI: 10.1128/mbio.03409-23] [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: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 03/14/2024] Open
Abstract
Candida albicans can cause mucosal infections in humans. This includes oropharyngeal candidiasis, which is commonly observed in human immunodeficiency virus infected patients, and vulvovaginal candidiasis (VVC), which is the most frequent manifestation of candidiasis. Epithelial cell invasion by C. albicans hyphae is accompanied by the secretion of candidalysin, a peptide toxin that causes epithelial cell cytotoxicity. During vaginal infections, candidalysin-driven tissue damage triggers epithelial signaling pathways, leading to hyperinflammatory responses and immunopathology, a hallmark of VVC. Therefore, we proposed blocking candidalysin activity using nanobodies to reduce epithelial damage and inflammation as a therapeutic strategy for VVC. Anti-candidalysin nanobodies were confirmed to localize around epithelial-invading C. albicans hyphae, even within the invasion pocket where candidalysin is secreted. The nanobodies reduced candidalysin-induced damage to epithelial cells and downstream proinflammatory responses. Accordingly, the nanobodies also decreased neutrophil activation and recruitment. In silico mathematical modeling enabled the quantification of epithelial damage caused by candidalysin under various nanobody dosing strategies. Thus, nanobody-mediated neutralization of candidalysin offers a novel therapeutic approach to block immunopathogenic events during VVC and alleviate symptoms.IMPORTANCEWorldwide, vaginal infections caused by Candida albicans (VVC) annually affect millions of women, with symptoms significantly impacting quality of life. Current treatments are based on anti-fungals and probiotics that target the fungus. However, in some cases, infections are recurrent, called recurrent VVC, which often fails to respond to treatment. Vaginal mucosal tissue damage caused by the C. albicans peptide toxin candidalysin is a key driver in the induction of hyperinflammatory responses that fail to clear the infection and contribute to immunopathology and disease severity. In this pre-clinical evaluation, we show that nanobody-mediated candidalysin neutralization reduces tissue damage and thereby limits inflammation. Implementation of candidalysin-neutralizing nanobodies may prove an attractive strategy to alleviate symptoms in complicated VVC cases.
Collapse
Affiliation(s)
- Marisa Valentine
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
| | - Paul Rudolph
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
| | - Antzela Tsavou
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, England, United Kingdom
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
| | - Sejeong Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, England, United Kingdom
| | - Emily L. Priest
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, England, United Kingdom
| | - Gaukhar Zhurgenbayeva
- Institute of Applied Optics and Biophysics, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| | - Nadja Jablonowski
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
| | - Sandra Timme
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
| | - Christian Eggeling
- Institute of Applied Optics and Biophysics, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
- Biophysical Imaging, Leibniz Institute of Photonic Technology, Jena, Germany
- Jena Center for Soft Matter (JCSM), Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
| | | | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, England, United Kingdom
| | - Marc T. Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| | - Mark S. Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
44
|
Garcia-Bustos V, Acosta-Hernández B, Cabañero-Navalón MD, Ruiz-Gaitán AC, Pemán J, Rosario Medina I. Potential Fungal Zoonotic Pathogens in Cetaceans: An Emerging Concern. Microorganisms 2024; 12:554. [PMID: 38543604 PMCID: PMC10972490 DOI: 10.3390/microorganisms12030554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 11/12/2024] Open
Abstract
Over 60% of emerging infectious diseases in humans are zoonotic, often originating from wild animals. This long-standing ecological phenomenon has accelerated due to human-induced environmental changes. Recent data show a significant increase in fungal infections, with 6.5 million cases annually leading to 3.7 million deaths, indicating their growing impact on global health. Despite the vast diversity of fungal species, only a few are known to infect humans and marine mammals. Fungal zoonoses, especially those involving marine mammals like cetaceans, are of global public health concern. Increased human-cetacean interactions, in both professional and recreational settings, pose risks for zoonotic disease transmission. This review focuses on the epidemiology, clinical manifestations, and zoonotic potential of major fungal pathogens shared in humans and cetaceans, highlighting their interspecies transmission capability and the challenges posed by antifungal resistance and environmental changes. It underscores the need for enhanced awareness and preventative measures in high-risk settings to protect public health and marine ecosystems.
Collapse
Affiliation(s)
- Victor Garcia-Bustos
- University Institute of Animal Health and Food Security (ULPGC-IUSA), University of Las Palmas de Gran Canaria, 35416 Arucas, Spain;
- Severe Infection Research Group, Health Research Institute La Fe, 46026 Valencia, Spain; (M.D.C.-N.); (A.C.R.-G.); (J.P.)
| | - Begoña Acosta-Hernández
- University Institute of Animal Health and Food Security (ULPGC-IUSA), University of Las Palmas de Gran Canaria, 35416 Arucas, Spain;
| | - Marta Dafne Cabañero-Navalón
- Severe Infection Research Group, Health Research Institute La Fe, 46026 Valencia, Spain; (M.D.C.-N.); (A.C.R.-G.); (J.P.)
| | - Alba Cecilia Ruiz-Gaitán
- Severe Infection Research Group, Health Research Institute La Fe, 46026 Valencia, Spain; (M.D.C.-N.); (A.C.R.-G.); (J.P.)
| | - Javier Pemán
- Severe Infection Research Group, Health Research Institute La Fe, 46026 Valencia, Spain; (M.D.C.-N.); (A.C.R.-G.); (J.P.)
| | - Inmaculada Rosario Medina
- University Institute of Animal Health and Food Security (ULPGC-IUSA), University of Las Palmas de Gran Canaria, 35416 Arucas, Spain;
| |
Collapse
|
45
|
Das S, Konwar BK. Inhibiting pathogenicity of vaginal Candida albicans by lactic acid bacteria and MS analysis of their extracellular compounds. APMIS 2024; 132:161-186. [PMID: 38168754 DOI: 10.1111/apm.13365] [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: 05/13/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
Maintaining healthy vaginal microflora post-puberty is critical. In this study we explore the potential of vaginal lactic acid bacteria (LAB) and their extracellular metabolites against the pathogenicity of Candida albicans. The probiotic culture free supernatant (PCFS) from Lactobacillus crispatus, L. gasseri, and L. vaginalis exhibit an inhibitory effect on budding, hyphae, and biofilm formation of C. albicans. LGPCFS manifested the best potential among the LAB PCFS, inhibiting budding for 24 h and restricting hyphae formation post-stimulation. LGPCFS also pre-eminently inhibited biofilm formation. Furthermore, L. gasseri itself grew under RPMI 1640 stimulation suppressing the biofilm formation of C. albicans. The PCFS from the LAB downregulated the hyphal genes of C. albicans, inhibiting the yeast transformation to fungi. Hyphal cell wall proteins HWP1, ALS3, ECE1, and HYR1 and transcription factors BCR1 and CPH1 were downregulated by the metabolites from LAB. Finally, the extracellular metabolome of the LAB was studied by LC-MS/MS analysis. L.gasseri produced the highest antifungal compounds and antibiotics, supporting its best activity against C. albicans. Vaginal LAB and their extracellular metabolites perpetuate C. albicans at an avirulent state. The metabolites produced by these LAB in vitro have been identified, and can be further exploited as a preventive measure against vaginal candidiasis.
Collapse
Affiliation(s)
- Shreaya Das
- Department of MBBT, Tezpur University, Napaam, Assam, India
| | | |
Collapse
|
46
|
Oh MJ, Yoon S, Babeer A, Liu Y, Ren Z, Xiang Z, Miao Y, Cormode DP, Chen C, Steager E, Koo H. Nanozyme-Based Robotics Approach for Targeting Fungal Infection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2300320. [PMID: 37141008 PMCID: PMC10624647 DOI: 10.1002/adma.202300320] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/29/2023] [Indexed: 05/05/2023]
Abstract
Fungal pathogens have been designated by the World Health Organization as microbial threats of the highest priority for global health. It remains a major challenge to improve antifungal efficacy at the site of infection while avoiding off-target effects, fungal spreading, and drug tolerance. Here, a nanozyme-based microrobotic platform is developed that directs localized catalysis to the infection site with microscale precision to achieve targeted and rapid fungal killing. Using electromagnetic field frequency modulation and fine-scale spatiotemporal control, structured iron oxide nanozyme assemblies are formed that display tunable dynamic shape transformation and catalysis activation. The catalytic activity varies depending on the motion, velocity, and shape providing controllable reactive oxygen species (ROS) generation. Unexpectedly, nanozyme assemblies bind avidly to fungal (Candida albicans) surfaces to enable concentrated accumulation and targeted ROS-mediated killing in situ. By exploiting these tunable properties and selective binding to fungi, localized antifungal activity is achieved using in vivo-like cell spheroid and animal tissue infection models. Structured nanozyme assemblies are directed to Candida-infected sites using programmable algorithms to perform precisely guided spatial targeting and on-site catalysis resulting in fungal eradication within 10 min. This nanozyme-based microrobotics approach provides a uniquely effective and targeted therapeutic modality for pathogen elimination at the infection site.
Collapse
Affiliation(s)
- Min Jun Oh
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemical and Biomolecular Engineering, School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seokyoung Yoon
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, Philadelphia, PA 19104, USA
| | - Alaa Babeer
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Endodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Oral Biology, King Abdulaziz University, Jeddah 21589, KSA
| | - Yuan Liu
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Preventive & Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhi Ren
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhenting Xiang
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yilan Miao
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David P. Cormode
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edward Steager
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- GRASP Laboratory, School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hyun Koo
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
47
|
Schimanski J, Gresnigt MS, Brunner E, Werz O, Hube B, Garscha U. Hyphal-associated protein expression is crucial for Candida albicans-induced eicosanoid biosynthesis in immune cells. Eur J Immunol 2024; 54:e2350743. [PMID: 38233139 DOI: 10.1002/eji.202350743] [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: 08/29/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
Candida albicans causes opportunistic infections ranging from mucosal mycoses to life-threatening systemic infections in immunocompromised patients. During C. albicans infection, leukotrienes and prostaglandins are formed from arachidonic acid by 5-lipoxygenase (5-LOX) and cyclooxygenases, respectively to amplify inflammatory conditions, but also to initiate macrophage infiltration to achieve tissue homeostasis. Since less is known about the cellular mechanisms triggering such lipid mediator biosynthesis, we investigated the eicosanoid formation in monocyte-derived M1 and M2 macrophages, neutrophils and HEK293 cells transfected with 5-LOX and 5-LOX-activating protein (FLAP) in response to C. albicans yeast or hyphae. Leukotriene biosynthesis was exclusively induced by hyphae in neutrophils and macrophages, whereas prostaglandin E2 was also formed in response to yeast cells by M1 macrophages. Eicosanoid biosynthesis was significantly higher in M1 compared to M2 macrophages. In HEK_5-LOX/FLAP cells only hyphae activated the essential 5-LOX translocation to the nuclear membrane. Using yeast-locked C. albicans mutants, we demonstrated that hyphal-associated protein expression is critical in eicosanoid formation. For neutrophils and HEK_5-LOX/FLAP cells, hyphal wall protein 1 was identified as the essential surface protein that stimulates leukotriene biosynthesis. In summary, our data suggest that hyphal-associated proteins of C. albicans are central triggers of eicosanoid biosynthesis in human phagocytes.
Collapse
Affiliation(s)
- Jana Schimanski
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Greifswald University, Greifswald, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Elena Brunner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller University Jena, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller University Jena, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
- Institute for Microbiology, Friedrich Schiller University, Jena, Germany
| | - Ulrike Garscha
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Greifswald University, Greifswald, Germany
| |
Collapse
|
48
|
Zhang TY, Chen YQ, Tan JC, Zhou JA, Chen WN, Jiang T, Zha JY, Zeng XK, Li BW, Wei LQ, Zou Y, Zhang LY, Hong YM, Wang XL, Zhu RZ, Xu WX, Xi J, Wang QQ, Pan L, Zhang J, Luan Y, Zhu RX, Wang H, Chen C, Liu NN. Global fungal-host interactome mapping identifies host targets of candidalysin. Nat Commun 2024; 15:1757. [PMID: 38413612 PMCID: PMC10899660 DOI: 10.1038/s41467-024-46141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/15/2024] [Indexed: 02/29/2024] Open
Abstract
Candidalysin, a cytolytic peptide toxin secreted by the human fungal pathogen Candida albicans, is critical for fungal pathogenesis. Yet, its intracellular targets have not been extensively mapped. Here, we performed a high-throughput enhanced yeast two-hybrid (HT-eY2H) screen to map the interactome of all eight Ece1 peptides with their direct human protein targets and identified a list of potential interacting proteins, some of which were shared between the peptides. CCNH, a regulatory subunit of the CDK-activating kinase (CAK) complex involved in DNA damage repair, was identified as one of the host targets of candidalysin. Mechanistic studies revealed that candidalysin triggers a significantly increased double-strand DNA breaks (DSBs), as evidenced by the formation of γ-H2AX foci and colocalization of CCNH and γ-H2AX. Importantly, candidalysin binds directly to CCNH to activate CAK to inhibit DNA damage repair pathway. Loss of CCNH alleviates DSBs formation under candidalysin treatment. Depletion of candidalysin-encoding gene fails to induce DSBs and stimulates CCNH upregulation in a murine model of oropharyngeal candidiasis. Collectively, our study reveals that a secreted fungal toxin acts to hijack the canonical DNA damage repair pathway by targeting CCNH and to promote fungal infection.
Collapse
Affiliation(s)
- Tian-Yi Zhang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yao-Qi Chen
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing-Cong Tan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jin-An Zhou
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wan-Ning Chen
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Department of Bioinformatics, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Tong Jiang
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Unit of Pathogenic Fungal Infection & Host Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jin-Yin Zha
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Xiang-Kang Zeng
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Science, Shanghai, China
| | - Bo-Wen Li
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lu-Qi Wei
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yun Zou
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Unit of Pathogenic Fungal Infection & Host Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lu-Yao Zhang
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Unit of Pathogenic Fungal Infection & Host Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue-Mei Hong
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiu-Li Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Run-Ze Zhu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wan-Xing Xu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Xi
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qin-Qin Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Pan
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Science, Shanghai, China
| | - Jian Zhang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Yang Luan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui-Xin Zhu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Department of Bioinformatics, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Changbin Chen
- The Center for Microbes, Development, and Health, Key Laboratory of Molecular Virology and Immunology, Unit of Pathogenic Fungal Infection & Host Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
49
|
Huang X, Lin R, Mao B, Tang X, Zhao J, Zhang Q, Cui S. Lactobacillus crispatus CCFM1339 Inhibits Vaginal Epithelial Barrier Injury Induced by Gardnerella vaginalis in Mice. Biomolecules 2024; 14:240. [PMID: 38397477 PMCID: PMC10886512 DOI: 10.3390/biom14020240] [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: 01/21/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The vaginal epithelial barrier, which integrates mechanical, immune, chemical, and microbial defenses, is pivotal in safeguarding against external pathogens and upholding the vaginal microecological equilibrium. Although the widely used metronidazole effectively curtails Gardnerella vaginalis, a key pathogen in bacterial vaginosis, it falls short in restoring the vaginal barrier or reducing recurrence rates. Our prior research highlighted Lactobacillus crispatus CCFM1339, a vaginally derived Lactobacillus strain, for its capacity to modulate the vaginal epithelial barrier. In cellular models, L. crispatus CCFM1339 fortified the integrity of the cellular monolayer, augmented cellular migration, and facilitated repair. Remarkably, in animal models, L. crispatus CCFM1339 substantially abated the secretion of the barrier disruption biomarker E-cadherin (from 101.45 to 82.90 pg/mL) and increased the anti-inflammatory cytokine IL-10 (35.18% vs. the model), consequently mitigating vaginal inflammation in mice. Immunological assays in vaginal tissues elucidated increased secretory IgA levels (from 405.56 to 740.62 ng/mL) and curtailed IL-17 gene expression. Moreover, L. crispatus CCFM1339 enhanced Lactobacilli abundance and attenuated Enterobacterium and Enterococcus within the vaginal microbiome, underscoring its potential in probiotic applications for vaginal barrier regulation.
Collapse
Affiliation(s)
- Xiaoyan Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
| | - Rumeng Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
50
|
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
|