1
|
Oshika, Bari VK. Molecular mechanism of host-yeast interactions and prevention by nanoformulation approaches. Microb Pathog 2025:107663. [PMID: 40339625 DOI: 10.1016/j.micpath.2025.107663] [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: 10/30/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
Fungal infections are a major source of morbidity and mortality in people with compromised immune systems, such as those with human immunodeficiency virus, cancer, organ transplant recipients, and patients undergoing chemotherapy in healthcare settings. According to a recent World Health Organization (WHO) fungal priority pathogens list, Cryptococcus spp., Candida spp., Aspergillus spp., and Candia auris cause severe invasive infections in humans. These opportunistic pathogens cause a significant number of mycoses, which affect over a billion people annually. Around two million infections can be fatal, especially for those with compromised immune systems. To diagnose and treat mycoses, we need to understand the complex interactions between the fungus and the host during pathogenesis, the virulence-causing traits of the fungus, and how the host fights infection through the immune system. Although several antifungal drugs are available to treat fungal infections, their effectiveness is highly variable with adverse effects. In addition, the increasing resistance to traditional antifungal treatments poses serious risks to the healthcare industry. Therefore, new therapeutic strategies are required to combat these potentially fatal fungal infections. Nanostructure-based formulations can improve the therapeutic efficacy of conventional medications by broadening their activities, decreasing toxicity, enhancing bioactivity, and improving biodistribution. The review highlights host and fungus interaction and how nanoformulations can be targeted against fungal infections.
Collapse
Affiliation(s)
- Oshika
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO- Ghudda, Bathinda -India
| | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO- Ghudda, Bathinda -India.
| |
Collapse
|
2
|
Lortal L, Lyon CM, Sprague JL, Sonnberger J, Paulin OKA, Wickramasinghe DN, Richardson JP, Hube B, Naglik JR. Candidalysin biology and activation of host cells. mBio 2025:e0060324. [PMID: 40293285 DOI: 10.1128/mbio.00603-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can cause life-threatening systemic infections and distressing mucosal infections. A major breakthrough in understanding C. albicans pathogenicity was the discovery of candidalysin, the first cytolytic peptide toxin identified in a human pathogenic fungus. Secreted by C. albicans hyphae and encoded by the ECE1 gene, this 31-amino acid peptide integrates into and permeabilizes host cell membranes, causing damage across diverse cell types. Beyond its cytolytic activity, candidalysin can trigger potent innate immune responses in epithelial cells, macrophages, and neutrophils. Additionally, candidalysin plays a key role in nutrient acquisition during infection. This review explores the biology of candidalysin, its role in host cell activation, and extends the discussion to non-candidalysin Ece1p peptides, shedding light on their emerging significance.
Collapse
Affiliation(s)
- Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Claire M Lyon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Johannes Sonnberger
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Olivia K A Paulin
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Don N Wickramasinghe
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Schille TB, Sprague JL, Naglik JR, Brunke S, Hube B. Commensalism and pathogenesis of Candida albicans at the mucosal interface. Nat Rev Microbiol 2025:10.1038/s41579-025-01174-x. [PMID: 40247134 DOI: 10.1038/s41579-025-01174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
Fungi are important and often underestimated human pathogens. Infections with fungi mostly originate from the environment, from soil or airborne spores. By contrast, Candida albicans, one of the most common and clinically important fungal pathogens, permanently exists in the vast majority of healthy individuals as a member of the human mucosal microbiota. Only under certain circumstances will these commensals cause infections. However, although the pathogenic behaviour and disease manifestation of C. albicans have been at the centre of research for many years, its asymptomatic colonization of mucosal surfaces remains surprisingly understudied. In this Review, we discuss the interplay of the fungus, the host and the microbiome on the dualism of commensal and pathogenic life of C. albicans, and how commensal growth is controlled and permitted. We explore hypotheses that could explain how the mucosal environment shapes C. albicans adaptations to its commensal lifestyle, while still maintaining or even increasing its pathogenic potential.
Collapse
Affiliation(s)
- Tim B Schille
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
4
|
de Mello Fiallos N, Zanin dos Santos IJ, Brunson DN, Kajfasz JK, Zeng L, de Aguiar Cordeiro R, Lemos JA, Abranches J. Candida albicans impacts carbohydrate metabolism of Enterococcus faecalis in interkingdom biofilms. J Oral Microbiol 2025; 17:2492194. [PMID: 40247862 PMCID: PMC12004723 DOI: 10.1080/20002297.2025.2492194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Objectives This study investigated the transcriptional and physiological responses of Enterococcus faecalis, an opportunistic pathogen linked to endodontic infections, when cultivated in dual-species biofilms with Candida albicans, a yeast pathobiont found in the oral cavity. Methods Forty-eight-hour E. faecalis OG1RF biofilms were developed in BHI medium as mono- or dual-species with C. albicans SC5314. Biofilms were assessed for biomass, colony-forming units (CFUs), and architecture using confocal microscopy. RNA sequencing was performed on an Illumina platform. Mannose-PTS activity and glycerol quantification assays were conducted to investigate changes in carbohydrate metabolism. Results Transcriptomic analysis revealed 149 E. faecalis genes differentially expressed in dual-species biofilms. Genes linked to mannose-PTS and glycerol metabolism were notably upregulated. Mannose-PTS activity was significantly higher in dual-species biofilms. Mannose, as the sole carbohydrate source, increased E. faecalis CFUs and decreased C. albicans CFUs in co-culture, while glucose had no effect. As C. albicans is a glycerol net producer, glycerol levels were always higher when C. albicans was present, likely contributing to the upregulation of glycerol metabolism genes in E. faecalis when in co-cultures. Conclusions The presence of C. albicans alters E. faecalis gene expression and metabolism, suggesting metabolic crosstalk that may influence their pathogenicity and role in oral infections.
Collapse
Affiliation(s)
- Nicole de Mello Fiallos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
- Department of Pathology and Legal Medicine, Federal University of Ceará, College of Medicine, Fortaleza, Brazil
| | - Iriana J. Zanin dos Santos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
- Department of Cariology and Operative Dentistry, Federal University of Ceará, College of Dentistry, Sobral, Brazil
| | - Debra N. Brunson
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jessica K. Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Rossana de Aguiar Cordeiro
- Department of Pathology and Legal Medicine, Federal University of Ceará, College of Medicine, Fortaleza, Brazil
| | - José A. Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jacqueline Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
5
|
Zhang Q, Li G, Wang Y, Yang C, Bai W, Li Q, Zhang J, Zhang P. Cas5 Regulates the Exposure of β-Glucan, the Cell Surface Hydrophobicity, and the Expression of Cell Wall Proteins to Remodel the Candida albicans Cell Wall and Participates in the Recruitment of Neutrophils. Microorganisms 2025; 13:683. [PMID: 40142575 PMCID: PMC11944837 DOI: 10.3390/microorganisms13030683] [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: 02/06/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Candida albicans (C. albicans) is a major opportunistic fungal pathogen that causes life-threatening infections, particularly in immunocompromised individuals, underscoring the critical need to understand its pathogenic mechanisms. This study investigates the role of Cas5, a key transcription factor, in regulating C. albicans cell wall remodeling, virulence, and host interactions. Genetic manipulation and biochemical assays were used to examine the effects of Cas5 depletion on C. albicans cell wall structure, adhesion to host cells, morphology transition, innate immune cells recruitment, and pathogenicity in a BALB/C mouse model of oropharyngeal candidiasis (OPC). The results showed that the Cas5 depletion mediated β-glucan exposure and enhanced C. albicans's ability to recruit neutrophils in vivo. Additionally, Cas5-mediated changes in cell surface hydrophobicity (CSH), CWP expressions, and morphological transition promoted C. albicans adhesion to biologically active surfaces (host cells) and increased fungal burden in the mouse model of OPC. In conclusion, Cas5 modulates C. albicans cell wall remodeling by masking cell wall β-glucan, altering CSH, and regulating the expression of cell wall proteins (CWPs). Additionally, Cas5 participates in inhibiting neutrophil recruitment and enhancing the C. albicans adhesion to host cells, as well as facilitating morphological transitions. These actions promote the colonization and invasion of C. albicans in OPC pathogenesis.
Collapse
Affiliation(s)
- Qiyue Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Guanglin Li
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yanmei Wang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Chen Yang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Wenhui Bai
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Qingqing Li
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| | - Peipei Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (Q.Z.); (G.L.); (Y.W.); (C.Y.); (W.B.); (Q.L.); (J.Z.)
- Institute of Pharmaceutical Science and Technology, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
6
|
Mishra A, Solis NV, Dietz SM, Crouch AL, Filler SG, Anderson MZ. Strain background interacts with chromosome 7 aneuploidy to determine commensal and virulence phenotypes in Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634449. [PMID: 39896449 PMCID: PMC11785170 DOI: 10.1101/2025.01.23.634449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The human fungal pathobiont Candida albicans displays extensive genomic plasticity, including large-scale chromosomal changes such as aneuploidy. Chromosome trisomy appears frequently in natural and laboratory strains of C. albicans. Trisomy of specific chromosomes has been linked to large phenotypic effects, such as increased murine gut colonization by strains trisomic for chromosome 7 (Chr7). However, studies of whole-chromosome aneuploidy are generally limited to the SC5314 genome reference strain, making it unclear whether the imparted phenotypes are conserved across C. albicans genetic backgrounds. Here, we report the presence of a Chr7 trisomy in the "commensal-like" oral candidiasis strain, 529L, and dissect the contribution of Chr7 trisomy to colonization and virulence in 529L and SC5314. These experiments show that strain background and homolog identity (i.e., AAB vs ABB) interact with Chr7 trisomy to alter commensal and virulence phenotypes in multiple host niches. In vitro filamentation was the only phenotype altered by Chr7 trisomy in similar ways across the two strain backgrounds. Oral colonization of mice was increased by the presence of a Chr7 trisomy in 529L but not SC5314; conversely, virulence during systemic infection was reduced by Chr7 trisomy in SC5314 but not 529L. Strikingly, the AAB Chr7 trisomy in the SC5314 background rendered this strain avirulent in murine systemic infection. Increased dosage of NRG1 failed to reproduce most of the Chr7 trisomy phenotypes. Our results demonstrate that aneuploidy interacts with background genetic variation to produce complex phenotypic patterns that deviate from our current understanding in the genome reference strain.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
- Center for Genomic Science Innovation, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Norma V. Solis
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Siobhan M. Dietz
- Cellular and Molecular Pathology, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Audra L. Crouch
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Scott G. Filler
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Matthew Z. Anderson
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
- Center for Genomic Science Innovation, University of Wisconsin - Madison, Madison, WI, 53706, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, WI, 53706, USA
| |
Collapse
|
7
|
Cheng H, Tian G, Liu H, Bai D, Zhang Y, Wang Q, Zhao M, Cao S, Deng D, Wang X. A molybdenum sulfide based nitric oxide controlled release oral gel for rapid healing of oral mucosal ulcers. J Colloid Interface Sci 2025; 678:560-571. [PMID: 39214008 DOI: 10.1016/j.jcis.2024.08.210] [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: 07/04/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Oral mucosal ulcer is the most prevalent oral mucosal lesion, affecting over 25 % of general population. The current treatment regimens lack efficacy in addressing challenges such as wound bleeding, bacterial infection and inflammation on a continuous basis. Hence, a multi-functional oral gel (termed MPCST) with a long-acting duration is designed. It is based on a tannic acid-thioctic acid (TATA) supramolecular hydrogel which absorbs tissue exudate while exhibiting robust tissue adhesion properties. To form MPCST, TATA is loaded with MPCS, which are composed of polydopamine (PDA)-coated molybdenum disulfide (MoS2) nanoflakes (MoS2@PDA) with high photothermal conversion efficiency, nitric oxide (NO) precursor nitroprusside (SNP) and cerium oxide (CeO2) with high reactive oxygen species (ROS) scavenging rate. Upon exposure to 808 nm near-infrared (NIR) irradiation, MPCS rapidly heats up and releases NO to promote angiogenesis, while exhibiting strong ROS scavenging, antibacterial (including oral common Streptococcus mutans), and anti-inflammatory properties. Animal experiments show that the MPCST oral gel, composed of MPCS and TATA hydrogel, exhibits superior therapeutic efficacy compared to the commonly used dexamethasone patch.
Collapse
Affiliation(s)
- Haoxin Cheng
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Guangqi Tian
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen 518000, PR China
| | - Hongmei Liu
- School of the First Clinical Medica, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Danmeng Bai
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Yue Zhang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Qingqing Wang
- School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Mengzhen Zhao
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Shuangyuan Cao
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Dan Deng
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, PR China
| | - Xiaolei Wang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, PR China; The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, PR China.
| |
Collapse
|
8
|
Hitzler SUJ, Fernández-Fernández C, Montaño DE, Dietschmann A, Gresnigt MS. Microbial adaptive pathogenicity strategies to the host inflammatory environment. FEMS Microbiol Rev 2025; 49:fuae032. [PMID: 39732621 PMCID: PMC11737513 DOI: 10.1093/femsre/fuae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 12/30/2024] Open
Abstract
Pathogenic microorganisms can infect a variety of niches in the human body. During infection, these microbes can only persist if they adapt adequately to the dynamic host environment and the stresses imposed by the immune system. While viruses entirely rely on host cells to replicate, bacteria and fungi use their pathogenicity mechanisms for the acquisition of essential nutrients that lie under host restriction. An inappropriate deployment of pathogenicity mechanisms will alert host defence mechanisms that aim to eradicate the pathogen. Thus, these adaptations require tight regulation to guarantee nutritional access without eliciting strong immune activation. To work efficiently, the immune system relies on a complex signalling network, involving a myriad of immune mediators, some of which are quite directly associated with imminent danger for the pathogen. To manipulate the host immune system, viruses have evolved cytokine receptors and viral cytokines. However, among bacteria and fungi, selected pathogens have evolved the capacity to use these inflammatory response-specific signals to regulate their pathogenicity. In this review, we explore how bacterial and fungal pathogens can sense the immune system and use adaptive pathogenicity strategies to evade and escape host defence to ensure their persistence in the host.
Collapse
Affiliation(s)
- Sophia U J Hitzler
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Candela Fernández-Fernández
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Dolly E Montaño
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| |
Collapse
|
9
|
Sawicka-Gutaj N, Stańska A, Stański M, Gruszczyński D, Zawalna N, Pochylski M, Ruchała M. Elimination of oral foci of infection might lead to clinical improvement of Graves' orbitopathy. Graefes Arch Clin Exp Ophthalmol 2025:10.1007/s00417-024-06716-2. [PMID: 39751637 DOI: 10.1007/s00417-024-06716-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 10/14/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
PURPOSE Graves' disease (GD) and Graves' orbitopathy (GO) are multifactorial disorders with links to the gut microbiome and autoimmunity. It is observed that patients with GD exhibit altered gut microbiome diversity. However, little is known about the role of oral microbiota in GD and GO. This study aims to investigate the impact of oral health and oral sanitation on the clinical course of GO in patients disqualified from glucocorticoid treatment due to oral infections. METHODS We reviewed 188 admissions of 127 patients with GO, hospitalized in a tertiary university hospital. Clinical, biochemical, imaging, ophthalmological, and oral health assessment data from each admission were analyzed. Patients excluded from the glucocorticoids (GCs) therapy due to oral foci of infection had the clinical activity score (CAS) reassessed after three months, and they were divided into two groups: with and without improvement. RESULTS Finishing dental treatment in the meantime was the only factor significantly correlated with improvement in these patients (p = 0.041). The secondary finding was that anti-thyroid peroxidase antibodies titer was significantly higher in the group with oral foci of infection considered as a contraindication for GCs (medians 28.50 vs 128.00; p = 0.026), and those patients were more likely to smoke than the group without oral issues (p = 0.024). CONCLUSIONS The results of our study suggest that monitoring and treating oral diseases may be pertinent in patients with GO and might serve as a supportive treatment strategy for managing the condition. KEY MESSAGES What is known: There is a recognized link between gut dysbiosis and the autoimmune processes in Graves' Disease (GD) and Graves' Orbitopathy (GO). WHAT IS NEW Elevated levels of TPOAb have been observed in patients with GO who also have oral foci of infection. Dental treatment has been shown to lead to significant clinical improvements in patients with GO. Maintaining oral hygiene might serve as a supportive treatment strategy for managing GO.
Collapse
Affiliation(s)
- Nadia Sawicka-Gutaj
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Alicja Stańska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Marcin Stański
- Department of General Radiology and Neuroradiology, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Dawid Gruszczyński
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Natalia Zawalna
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Mateusz Pochylski
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland
| |
Collapse
|
10
|
Zhang W, Zhao J, Zou X, Yu J, Liao J, Huang F. Multifunctional hydrogels for the healing of oral ulcers. J Biomed Mater Res A 2025; 113:e37776. [PMID: 39210659 DOI: 10.1002/jbm.a.37776] [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: 04/21/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Oral ulcers are one of the most common oral diseases in clinical practice. Its etiology is complex and varied. Due to the dynamic nature of the oral environment, the wound surface is painful due to contact and wear, which seriously affects the quality of life of patients. Oral ulcers are often treated with topical drug therapy. Studies have shown that functional hydrogels play a positive role in promoting wound healing, showing unique advantages in wound dressings. In this paper, the causes and healing characteristics of oral ulcers are discussed in depth, and then the common treatment methods for oral ulcers are summarized and compared. Finally, the potential of functional hydrogels in the treatment of oral ulcers is discussed and projected through a review of the literature in recent years.
Collapse
Affiliation(s)
- Wenjie Zhang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jie Zhao
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinxin Zou
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jingrong Yu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jinlong Liao
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fengjie Huang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
De Lima Gualque MW, Vaso CO, dos Santos KS, Galeane MC, Gomes PC, Palma MS, Soares Mendes Giannini MJ, Moroz A, Fusco Almeida AM. Peptides from Galleria mellonella against Cryptococcus spp: toxicity in three-dimensional cell cultures and G. mellonella. Future Microbiol 2025; 20:11-21. [PMID: 39552598 PMCID: PMC11731228 DOI: 10.1080/17460913.2024.2421632] [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: 06/10/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
Aim: This work aimed to test peptides against the planktonic and biofilm form of Cryptococcus spp. and in vitro toxicity using three-dimensional (3D) cells characterized and evaluate in vivo toxicity in Galleria mellonella.Materials & methods: Susceptibility tests were conducted on the planktonic form and biofilm formation. The toxicity of the peptides was evaluated in lung and brain cells in monolayer (2D) and 3D mono- and co-culture, in addition to in vivo analysis with G. mellonella.Results: Susceptibility values ranged from 31.25 to over 250 µg/ml with a fungicidal profile. Regarding toxicity, the PepM2 peptide was not toxic in 3D culture (500 µg/ml). G. mellonella, showed a survival rate of more than 85% In assays with brain and lung cell lines, concentrations ranged from 4 × 104 to 4 × 103 cells/well for brain cells and 1 × 103 cells/well for lung cells. Cocultures used 1 × 105 brain and 1 × 103 lung cells.Conclusion: This study shows that the peptides have great potential against cryptococcosis, and all spheroids were characterized as having a spheroidal and compact structure.
Collapse
Affiliation(s)
- Marcos William De Lima Gualque
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Carolina Orlando Vaso
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Kelvin Sousa dos Santos
- Laboratory for Monoclonal Antibodies, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Mariana Cristina Galeane
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Paulo César Gomes
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Mario Sérgio Palma
- Department of Basic & Applied Biology/LSBZ, Institute of Biosciences, São Paulo State University-UNESP, Rio Claro, 13506-900, São Paulo, Brazil
| | - Maria José Soares Mendes Giannini
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Andrei Moroz
- Laboratory for Monoclonal Antibodies, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Ana Marisa Fusco Almeida
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
12
|
Jørgensen MR. Pathophysiological microenvironments in oral candidiasis. APMIS 2024; 132:956-973. [PMID: 38571459 DOI: 10.1111/apm.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024]
Abstract
Oral candidiasis (OC), a prevalent opportunistic infection of the oral mucosa, presents a considerable health challenge, particularly in individuals with compromised immune responses, advanced age, and local predisposing conditions. A considerable part of the population carries Candida in the oral cavity, but only few develop OC. Therefore, the pathogenesis of OC may depend on factors other than the attributes of the fungus, such as host factors and other predisposing factors. Mucosal trauma and inflammation compromise epithelial integrity, fostering a conducive environment for fungal invasion. Molecular insights into the immunocompromised state reveal dysregulation in innate and adaptive immunity, creating a permissive environment for Candida proliferation. Detailed examination of Candida species (spp.) and their virulence factors uncovers a nuanced understanding beyond traditional C. albicans focus, which embrace diverse Candida spp. and their strategies, influencing adhesion, invasion, immune evasion, and biofilm formation. Understanding the pathophysiological microenvironments in OC is crucial for the development of targeted therapeutic interventions. This review aims to unravel the diverse pathophysiological microenvironments influencing OC development focusing on microbial, host, and predisposing factors, and considers Candida resistance to antifungal therapy. The comprehensive approach offers a refined perspective on OC, seeking briefly to identify potential therapeutic targets for future effective management.
Collapse
Affiliation(s)
- Mette Rose Jørgensen
- Section of Oral Pathology and Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Sabbatini S, Zatini L, Narducci E, Rosati L, Ardizzoni A, Mencacci A, Rende M, Pericolini E, Galli F, Bartolini D, Monari C. Modulation of C. albicans-Induced Immune Response in Vaginal Epithelial Cells by Garcinoic Acid. Microorganisms 2024; 12:2455. [PMID: 39770658 PMCID: PMC11678841 DOI: 10.3390/microorganisms12122455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Vulvovaginal candidiasis (VVC) is a prevalent women's infection characterized by excessive inflammation and damage of the vaginal epithelium that, in its recurrent form (RVVC), causes more than three symptomatic episodes per year, impacting nearly 8% of women globally. Current antifungal treatments alleviate symptoms but often fail to restore the inflammatory homeostasis of mucosal tissue and prevent recurrences. α-Tocopherol (α-TOH) and garcinoic acid (GA), a vitamin E metabolite, with immunomodulatory properties, were investigated for the first time in vaginal epithelial cells exposed to C. albicans infection to assess their effects on inflammatory signaling parameters important to restore cellular homeostasis. For this purpose, the protein kinases MKK3/6, p38 stress kinase (SAPK), and ERK1/2 were studied together with c-Fos transcription factor and IL-6, IL-1α, and IL-1β secretion in A-431 vaginal epithelial cells pre-treated with GA or with α-TOH and then infected with C. albicans. GA, differently from α-TOH, significantly reduced the C. albicans-induced activation of p38-SAPK while increasing pro-survival MAPK ERK1/2 activity. This resulted in a significant reduction in the secretion levels of the inflammatory cytokines IL-6 and IL-1α, as well as IL-1β. Overall, our data indicate that GA holds potential for restoring the immuno-metabolic properties of the vaginal epithelium exposed to C. albicans infection, which may help to treat inflammatory symptoms in VVC/RVVC.
Collapse
Affiliation(s)
- Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| | - Linda Zatini
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy; (L.Z.); (F.G.)
| | - Eleonora Narducci
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| | - Lucrezia Rosati
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy;
| | - Andrea Ardizzoni
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.P.)
| | - Antonella Mencacci
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy;
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.P.)
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy; (L.Z.); (F.G.)
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy; (L.Z.); (F.G.)
| | - Claudia Monari
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy; (E.N.); (A.M.); (C.M.)
| |
Collapse
|
14
|
Kashyap B, Padala SR, Kaur G, Kullaa A. Candida albicans Induces Oral Microbial Dysbiosis and Promotes Oral Diseases. Microorganisms 2024; 12:2138. [PMID: 39597528 PMCID: PMC11596246 DOI: 10.3390/microorganisms12112138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Candida albicans are ubiquitous fungal organisms that colonize the oral cavity of healthy individuals without causing disease. C. albicans is an opportunistic microorganism with several virulent factors that influence the inflammatory process and allow it to invade tissues, evade host defense mechanisms, and release toxins, facilitating proliferation and degradation. At present, increasing emphasis is placed on polymicrobial interactions between C. albicans and various bacterial pathogens. Such interaction is mutually beneficial for both parties: it is competitive and antagonistic. Their complex interaction and colonization in the oral cavity serve as the basis for several oral diseases. The dispersion of C. albicans in saliva and the systemic circulation is noted in association with other bacterial populations, suggesting their virulence in causing disease. Hence, it is necessary to understand fungal-bacterial interactions for early detection and the development of novel therapeutic strategies to treat oral diseases. In this paper, we review the mutualistic interaction of C. albicans in oral biofilm formation and polymicrobial interactions in oral diseases. In addition, C. albicans virulence in causing biofilm-related oral diseases and its presence in saliva are discussed.
Collapse
Affiliation(s)
- Bina Kashyap
- Institute of Dentistry, University of Eastern Finland, 70211 Kuopio, Finland;
| | | | - Gaganjot Kaur
- Shaheed Kartar Singh Sarabha Dental College & Hospital, Ludhiana 141105, India;
| | - Arja Kullaa
- Institute of Dentistry, University of Eastern Finland, 70211 Kuopio, Finland;
| |
Collapse
|
15
|
Kim MJ, Cravener M, Solis N, Filler SG, Mitchell AP. A Brg1-Rme1 circuit in Candida albicans hyphal gene regulation. mBio 2024; 15:e0187224. [PMID: 39078139 PMCID: PMC11389389 DOI: 10.1128/mbio.01872-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
Major Candida albicans virulence traits include its ability to make hyphae, to produce a biofilm, and to damage host cells. These traits depend upon expression of hypha-associated genes. A gene expression comparison among clinical isolates suggested that transcription factor Rme1, established by previous studies to be a positive regulator of chlamydospore formation, may also be a negative regulator of hypha-associated genes. Engineered RME1 overexpression supported this hypothesis, but no relevant rme1Δ/Δ mutant phenotype was detected. We reasoned that Rme1 may function within a specific regulatory pathway. This idea was supported by our finding that an rme1Δ/Δ mutation relieves the need for biofilm regulator Brg1 in biofilm formation. The impact of the rme1Δ/Δ mutation is most prominent under static or "biofilm-like" growth conditions. RNA sequencing (RNA-seq) of cells grown under biofilm-like conditions indicates that Brg1 activates hypha-associated genes indirectly via repression of RME1: hypha-associated gene expression levels are substantially reduced in a brg1Δ/Δ mutant and partially restored in a brg1Δ/Δ rme1Δ/Δ double mutant. An rme1Δ/Δ mutation does not simply bypass Brg1, because iron homeostasis genes depend upon Brg1 regardless of Rme1. Rme1 thus connects Brg1 to the targets relevant to hypha and biofilm formation under biofilm growth conditions.IMPORTANCECandida albicans is a major fungal pathogen of humans, and its ability to grow as a surface-associated biofilm on implanted devices is a common cause of infection. Here, we describe a new regulator of biofilm formation, RME1, whose activity is most prominent under biofilm-like growth conditions.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Max Cravener
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Norma Solis
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Scott G Filler
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Aaron P Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
16
|
Ramachandran R, Manan A, Kim J, Choi S. NLRP3 inflammasome: a key player in the pathogenesis of life-style disorders. Exp Mol Med 2024; 56:1488-1500. [PMID: 38945951 PMCID: PMC11297159 DOI: 10.1038/s12276-024-01261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 07/02/2024] Open
Abstract
Proinflammatory cytokines and chemokines play a crucial role in regulating the inflammatory response, which is essential for the proper functioning of our immune system. When infections or threats to the body's defense mechanisms are detected, the innate immune system takes the lead. However, an excessive inflammatory response can lead to the production of high concentrations of cytotoxic molecules, resulting in tissue damage. Inflammasomes are significant contributors to innate immunity, and one of the most extensively studied inflammasome complexes is NOD-like receptor 3 (NLRP3). NLRP3 has a wide range of recognition mechanisms that streamline immune activation and eliminate pathogens. These cytosolic multiprotein complexes are composed of effector, adaptor, and sensor proteins, which are crucial for identifying intracellular bacterial breakdown products and initiating an innate immune cascade. To understand the diverse behavior of NLRP3 activation and its significance in the development of lifestyle-related diseases, one must delve into the study of the immune response and apoptosis mediated by the release of proinflammatory cytokines. In this review, we briefly explore the immune response in the context of lifestyle associated disorders such as obesity, hyperlipidemia, diabetes, chronic respiratory disease, oral disease, and cardiovascular disease.
Collapse
Affiliation(s)
- Rajath Ramachandran
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.
| | - Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Jei Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon, 16502, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon, 16502, Korea.
| |
Collapse
|
17
|
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
|
18
|
Wu H, Li C, Wang Y, Zhang M, Wu D, Shao J, Wang T, Wang C. Transcriptomics Reveals Effect of Pulsatilla Decoction Butanol Extract in Alleviating Vulvovaginal Candidiasis by Inhibiting Neutrophil Chemotaxis and Activation via TLR4 Signaling. Pharmaceuticals (Basel) 2024; 17:594. [PMID: 38794163 PMCID: PMC11124330 DOI: 10.3390/ph17050594] [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: 04/09/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
The Pulsatilla decoction is a well-known herbal remedy used in clinical settings for treating vulvovaginal candidiasis (VVC). However, the specific mechanism that makes it effective is still unclear. Recent studies have shown that in cases of VVC, neutrophils recruited to the vagina, influenced by heparan sulfate (HS), do not successfully engulf Candida albicans (C. albicans). Instead, they release many inflammatory factors that cause damage to the vaginal mucosa. This study aims to understand the molecular mechanism by which the n-butanol extract of Pulsatilla decoction (BEPD) treats VVC through transcriptomics. High-performance liquid chromatography was used to identify the primary active components of BEPD. A VVC mouse model was induced using an estrogen-dependent method and the mice were treated daily with BEPD (20 mg/kg, 40 mg/kg, and 80 mg/kg) for seven days. The vaginal lavage fluid of the mice was analyzed for various experimental indices, including fungal morphology, fungal burden, degree of neutrophil infiltration, and cytokines. Various assessments were then performed on mouse vaginal tissues, including pathological assessment, immunohistochemistry, immunofluorescence, Western blot (WB), quantitative real-time PCR, and transcriptome assays. Our results showed that BEPD reduced vaginal redness and swelling, decreased white discharge, inhibited C. albicans hyphae formation, reduced neutrophil infiltration and fungal burden, and attenuated vaginal tissue damage compared with the VVC model group. The high-dose BEPD group even restored the damaged vaginal tissue to normal levels. The medium- and high-dose groups of BEPD also significantly reduced the levels of IL-1β, IL-6, TNF-α, and LDH. Additionally, transcriptomic results showed that BEPD regulated several chemokine (CXCL1, CXCL3, and CXCL5) and S100 alarmin (S100A8 and S100A9) genes, suggesting that BEPD may treat VVC by affecting chemokine- and alarmin-mediated neutrophil chemotaxis. Finally, we verified that BEPD protects the vaginal mucosa of VVC mice by inhibiting neutrophil recruitment and chemotaxis in an animal model of VVC via the TLR4/MyD88/NF-κB pathway. This study provides further evidence to elucidate the mechanism of BEPD treatment of VVC.
Collapse
Affiliation(s)
- Hui Wu
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
| | - Can Li
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
| | - Yemei Wang
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Mengxiang Zhang
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Daqiang Wu
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Jing Shao
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Tianming Wang
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Changzhong Wang
- School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (H.W.); (C.L.); (Y.W.); (M.Z.); (D.W.); (J.S.); (T.W.)
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
19
|
Nenciarini S, Renzi S, di Paola M, Meriggi N, Cavalieri D. The yeast-human coevolution: Fungal transition from passengers, colonizers, and invaders. WIREs Mech Dis 2024; 16:e1639. [PMID: 38146626 DOI: 10.1002/wsbm.1639] [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: 05/19/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/27/2023]
Abstract
Fungi are the cause of more than a billion infections in humans every year, although their interactions with the host are still neglected compared to bacteria. Major systemic fungal infections are very unusual in the healthy population, due to the long history of coevolution with the human host. Humans are routinely exposed to environmental fungi and can host a commensal mycobiota, which is increasingly considered as a key player in health and disease. Here, we review the current knowledge on host-fungi coevolution and the factors that regulate their interaction. On one hand, fungi have learned to survive and inhabit the host organisms as a natural ecosystem, on the other hand, the host immune system finely tunes the response toward fungi. In turn, recognition of fungi as commensals or pathogens regulates the host immune balance in health and disease. In the human gut ecosystem, yeasts provide a fingerprint of the transient microbiota. Their status as passengers or colonizers is related to the integrity of the gut barrier and the risk of multiple disorders. Thus, the study of this less known component of the microbiota could unravel the rules of the transition from passengers to colonizers and invaders, as well as their dependence on the innate component of the host's immune response. This article is categorized under: Infectious Diseases > Environmental Factors Immune System Diseases > Environmental Factors Infectious Diseases > Molecular and Cellular Physiology.
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
|
20
|
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
|
21
|
Chen J, Ren J, Wu Y, Hu N, Zhao F, Zhang L. Wet adhesive hydrogels based on niobium carbide for experimental research of oral mucosal impairment. RSC Adv 2024; 14:12935-12946. [PMID: 38650683 PMCID: PMC11033722 DOI: 10.1039/d4ra01352b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Oral mucosal impairment is a prevalent oral disease that frequently causes pain for patients. Conventional treatments have limited effectiveness and can cause adverse reactions. Furthermore, the moist and dynamic nature of the oral mucosal environment makes persistent adherence of conventional materials challenging, which can affect treatment efficacy. In this study, we investigated the potential of a NbC/TA-GelMA hydrogel system, where niobium carbide (NbC) and tannic acid (TA) were added to gelatin methacryloyl (GelMA), for repairing oral mucosal impairment. The wet adhesion properties of NbC/TA-GelMA hydrogels were confirmed by the inclusion of TA with a catechol-rich group. In addition, the photothermal effect of NbC/TA-GelMA hydrogel under near-infrared light, synergizing with TA, provided sustained antibacterial action. Furthermore, the NbC/TA-GelMA hydrogel effectively healed damaged oral mucosa of rats.
Collapse
Affiliation(s)
- Jiayuan Chen
- First Affiliated Hospital of Harbin Medical University, College of Stomatology, Harbin Medical University No. 143 Yiman Street, Nangang District Harbin 150001 China
| | - Junyu Ren
- Oral Implant Center, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Harbin Heilongjiang China
| | - Yingjie Wu
- Key Laboratory of Microsystems and Microstructures Manufacturing (Ministry of Education), School of Medicine and Health, Harbin Institute of Technology No. 92 XiDaZhi Street Harbin 150001 China
| | - Narisu Hu
- Oral Implant Center, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Harbin Heilongjiang China
| | - Fang Zhao
- Department of Dentistry, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Harbin Heilongjiang China
| | - Lin Zhang
- First Affiliated Hospital of Harbin Medical University, College of Stomatology, Harbin Medical University No. 143 Yiman Street, Nangang District Harbin 150001 China
| |
Collapse
|
22
|
Su K, Li J, Wu X, Deng D, Gu H, Sun Y, Wang X, Huang W, Wang Y, Shang X, Xue C, Liang L, Li X, Li D, Ang S, Zhang K, Wu P, Wu K. One-Step Synthesis of Hydrogel Adhesive with Acid-Responsive Tannin Release for Diabetic Oral Mucosa Defects Healing. Adv Healthc Mater 2024; 13:e2303252. [PMID: 38245866 DOI: 10.1002/adhm.202303252] [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: 09/25/2023] [Revised: 01/12/2024] [Indexed: 01/22/2024]
Abstract
The complex preparation, weak wet tissue adhesion, and limited biological activity of traditional oral wound dressings usually impede their efficient treatment and healing for diabetic oral mucosal defects. To overcome these problems, a novel hydrogel adhesive (named CFT hydrogel) is rapidly constructed using a one-step method based on dual-dynamic covalent cross-linking. Compared with the commercial oral patches, the CFT hydrogel shows superior in vivo (rat tongue) wet tissue adhesion performance. Additionally, the CFT hydrogel exhibits unique acid-responsive properties, thereby facilitating the release of bioactive molecule tannic acid in the acidic diabetic wound microenvironment. And a series of in vitro experiments substantiate the favorable biocompatibility and bioactivity properties (including antibacterial, antioxidative, anti-inflammatory, and angiogenetic effects) exhibited by CFT hydrogel. Moreover, in vivo experiments conducted on a diabetic rat model with oral mucosal defects demonstrate that the CFT hydrogel exhibits significant efficacy in protecting against mucosal wounds, alleviating inflammatory reactions, thereby facilitating the wound-healing process. Taken together, this study provides a promising and comprehensive therapeutic option with great potential for the clinical management of oral mucosa defects in diabetic patients.
Collapse
Affiliation(s)
- Kaize Su
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Jinxuan Li
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Xiaoxian Wu
- Instrumental Analysis and Research Center, South China Agricultural University, Guangzhou, 510642, China
| | - Duanyu Deng
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Han Gu
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Ying Sun
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Xu Wang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Wenhuan Huang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Yan Wang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Xiangcun Shang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Cuiyu Xue
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Lihua Liang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Xiaofang Li
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Dongli Li
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Song Ang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Kun Zhang
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Panpan Wu
- School of Pharmacy and Food Engineering, Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529040, P. R. China
| | - Keke Wu
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511495, P. R. China
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Ashrafi S, Amini AA, Karimi P, Bagherian M, Adibzadeh Sereshgi MM, Asgarhalvaei F, Ahmadi K, Yazdi MH, Jahantigh HR, Mahdavi M, Sarrami Forooshani R. Candidiasis in breast cancer: Tumor progression or not? IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1346-1356. [PMID: 39386227 PMCID: PMC11459349 DOI: 10.22038/ijbms.2024.75408.16379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/06/2024] [Indexed: 10/12/2024]
Abstract
Candida albicans is an "opportunistic fungal agent" in cancer patients that can become colonized in both mucosal and deep tissues and cause severe infections. Most evidence has shown that C. albicans can enhance the progress of different cancers by several mechanisms such as generating virulence factors, participation in endogenous production of pro-inflammatory mediators, and stimulating a wide range of immune cells in the host. The main idea of this review is to describe a range of Candida-used mechanisms that are important in candidiasis-associated malignant processes and cancer development, particularly breast cancer. This review intends to provide a detailed discussion on different regulatory mechanisms of C. albicans that undoubtedly help to open new therapeutic horizons of cancer therapy in patients with fungal infection. The current therapeutic approach is not fully effective in immunocompromised and cancer patients, and further studies are required to find new products with effective antifungal properties and minimal side effects to increase the susceptibility of opportunistic fungal infections to conventional antifungal agents. So, in this situation, a special therapy should be considered to control the infection and simultaneously have the most therapeutic index on tumor patients.
Collapse
Affiliation(s)
- Somayeh Ashrafi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, Faculty of Advanced Sciences & Technology, Tehran Medical Sciences, Islamic Azad University, (IAUPS), Tehran, Iran
- These authors contributed eqully to this work
| | - Abbas Ali Amini
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Pegah Karimi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Biochemistry, Faculty of Basic Sciences, Islamic Azad University, Central Tehran Branch, Tehran, Iran
- These authors contributed eqully to this work
| | - Maryam Bagherian
- Department of Hematology and Oncology and Stem Cell Transplantation, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Asgarhalvaei
- Department of Microbiology, Faculty of Advanced Sciences & Technology, Tehran Medical Sciences, Islamic Azad University, (IAUPS), Tehran, Iran
| | - Khadijeh Ahmadi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Hossein Yazdi
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Jahantigh
- Animal Health and Zoonosis PhD Course, Department of Veterinary Medicine, University of Bari, Bari, Italy
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Sarrami Forooshani
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| |
Collapse
|
26
|
Kabir AR, Chaudhary AA, Aladwani MO, Podder S. Decoding the host-pathogen interspecies molecular crosstalk during oral candidiasis in humans: an in silico analysis. Front Genet 2023; 14:1245445. [PMID: 37900175 PMCID: PMC10603195 DOI: 10.3389/fgene.2023.1245445] [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: 06/23/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: The objective of this study is to investigate the interaction between Candida albicans and human proteins during oral candidiasis, with the aim of identifying pathways through which the pathogen subverts host cells. Methods: A comprehensive list of interactions between human proteins and C. albicans was obtained from the Human Protein Interaction Database using specific screening criteria. Then, the genes that exhibit differential expression during oral candidiasis in C. albicans were mapped with the list of human-Candida interactions to identify the corresponding host proteins. The identified host proteins were further compared with proteins specific to the tongue, resulting in a final list of 99 host proteins implicated in oral candidiasis. The interactions between host proteins and C. albicans proteins were analyzed using the STRING database, enabling the construction of protein-protein interaction networks. Similarly, the gene regulatory network of Candida proteins was reconstructed using data from the PathoYeastract and STRING databases. Core module proteins within the targeted host protein-protein interaction network were identified using ModuLand, a Cytoscape plugin. The expression levels of the core module proteins under diseased conditions were assessed using data from the GSE169278 dataset. To gain insights into the functional characteristics of both host and pathogen proteins, ontology analysis was conducted using Enrichr and YeastEnrichr, respectively. Result: The analysis revealed that three Candida proteins, HHT21, CYP5, and KAR2, interact with three core host proteins, namely, ING4 (in the DNMT1 module), SGTA, and TOR1A. These interactions potentially impair the immediate immune response of the host against the pathogen. Additionally, differential expression analysis of fungal proteins and their transcription factors in Candida-infected oral cell lines indicated that Rob1p, Tye7p, and Ume6p could be considered candidate transcription factors involved in instigating the pathogenesis of oral candidiasis during host infection. Conclusion: Our study provides a molecular map of the host-pathogen interaction during oral candidiasis, along with potential targets for designing regimens to overcome oral candidiasis, particularly in immunocompromised individuals.
Collapse
Affiliation(s)
- Ali Rejwan Kabir
- Computational and System Biology Lab, Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Malak O Aladwani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Soumita Podder
- Computational and System Biology Lab, Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| |
Collapse
|
27
|
Taylor TC, Coleman BM, Arunkumar SP, Dey I, Dillon JT, Ponde NO, Poholek AC, Schwartz DM, McGeachy MJ, Conti HR, Gaffen SL. IκBζ is an essential mediator of immunity to oropharyngeal candidiasis. Cell Host Microbe 2023; 31:1700-1713.e4. [PMID: 37725983 PMCID: PMC10591851 DOI: 10.1016/j.chom.2023.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/28/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023]
Abstract
Fungal infections are a global threat; yet, there are no licensed vaccines to any fungal pathogens. Th17 cells mediate immunity to Candida albicans, particularly oropharyngeal candidiasis (OPC), but essential downstream mechanisms remain unclear. In the murine model of OPC, IκBζ (Nfkbiz, a non-canonical NF-κB transcription factor) was upregulated in an interleukin (IL)-17-dependent manner and was essential to prevent candidiasis. Deletion of Nfkbiz rendered mice highly susceptible to OPC. IκBζ was dispensable in hematopoietic cells and acted partially in the suprabasal oral epithelium to control OPC. One prominent IκBζ-dependent gene target was β-defensin 3 (BD3) (Defb3), an essential antimicrobial peptide. Human oral epithelial cells required IκBζ for IL-17-mediated induction of BD2 (DEFB4A, human ortholog of mouse Defb3) through binding to the DEFB4A promoter. Unexpectedly, IκBζ regulated the transcription factor Egr3, which was essential for C. albicans induction of BD2/DEFB4A. Accordingly, IκBζ and Egr3 comprise an antifungal signaling hub mediating mucosal defense against oral candidiasis.
Collapse
Affiliation(s)
- Tiffany C Taylor
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Bianca M Coleman
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Samyuktha P Arunkumar
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ipsita Dey
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John T Dillon
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Nicole O Ponde
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Amanda C Poholek
- Department of Pediatrics, University of Pittsburgh, Children's Hospital of UPMC, Pittsburgh, PA 15224, USA
| | - Daniella M Schwartz
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mandy J McGeachy
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Heather R Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
28
|
Liang X, Chen D, Wang J, Liao B, Shen J, Ye X, Wang Z, Zhu C, Gou L, Zhou X, Cheng L, Ren B, Zhou X. Artemisinins inhibit oral candidiasis caused by Candida albicans through the repression on its hyphal development. Int J Oral Sci 2023; 15:40. [PMID: 37699886 PMCID: PMC10497628 DOI: 10.1038/s41368-023-00245-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Candida albicans is the most abundant fungal species in oral cavity. As a smart opportunistic pathogen, it increases the virulence by switching its forms from yeasts to hyphae and becomes the major pathogenic agent for oral candidiasis. However, the overuse of current clinical antifungals and lack of new types of drugs highlight the challenges in the antifungal treatments because of the drug resistance and side effects. Anti-virulence strategy is proved as a practical way to develop new types of anti-infective drugs. Here, seven artemisinins, including artemisinin, dihydroartemisinin, artemisinic acid, dihydroartemisinic acid, artesunate, artemether and arteether, were employed to target at the hyphal development, the most important virulence factor of C. albicans. Artemisinins failed to affect the growth, but significantly inhibited the hyphal development of C. albicans, including the clinical azole resistant isolates, and reduced their damage to oral epithelial cells, while arteether showed the strongest activities. The transcriptome suggested that arteether could affect the energy metabolism of C. albicans. Seven artemisinins were then proved to significantly inhibit the productions of ATP and cAMP, while reduced the hyphal inhibition on RAS1 overexpression strain indicating that artemisinins regulated the Ras1-cAMP-Efg1 pathway to inhibit the hyphal development. Importantly, arteether significantly inhibited the fungal burden and infections with no systemic toxicity in the murine oropharyngeal candidiasis models in vivo caused by both fluconazole sensitive and resistant strains. Our results for the first time indicated that artemisinins can be potential antifungal compounds against C. albicans infections by targeting at its hyphal development.
Collapse
Affiliation(s)
- Xiaoyue Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingchen Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengguang Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Chen X, Wang J, Chen J, Wang G, Zhang R, Qiu J. Vaginal homeostasis features of Vulvovaginal Candidiasis through vaginal metabolic profiling. Med Mycol 2023; 61:myad085. [PMID: 37573133 DOI: 10.1093/mmy/myad085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/25/2023] [Accepted: 08/11/2023] [Indexed: 08/14/2023] Open
Abstract
Vulvovaginal candidiasis (VVC) is an inflammatory disease primarily infected by Candida albicans. The condition has good short-term treatment effects, high recurrence, and seriously affects the quality of life of women. Metabolomics has been applied to research a variety of inflammatory diseases. In the present study, the vaginal metabolic profiles of VVC patients and healthy populations (Cnotrol (CTL)) were explored by a non-targeted metabolomics approach. In total, 211 differential metabolites were identified, with the VVC group having 128 over-expressed and 83 under-expressed metabolites compared with healthy individuals. Functional analysis showed that these metabolites were mainly involved in amino acid metabolism and lipid metabolism. In addition, network software analysis indicated that the differential metabolites were associated with mitogen-activated protein kinase (MAPK) signaling and NF-κB signaling. Further molecular docking suggested that linoleic acid can bind to the acyl-CoA synthetase 1 (ACSL1) protein, which has been shown to be associated with multiple inflammatory diseases and is an upstream regulator of the MAPK and NF-κB signaling pathways that mediate inflammation. Therefore, our preliminary analysis results suggest that VVC has a unique metabolic profile. Linoleic acid, a significantly elevated unsaturated fatty acid in the VVC group, may promote VVC development through the ACSL1/MAPK and ACSL1/NF-κB signaling pathways. This study's findings contribute to further exploring the mechanism of VVC infection and providing new perspectives for the treatment of Candida albicans vaginal infection.
Collapse
Affiliation(s)
- Xinyi Chen
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinbo Wang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanghua Wang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runjie Zhang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Qiu
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Rapala-Kozik M, Surowiec M, Juszczak M, Wronowska E, Kulig K, Bednarek A, Gonzalez-Gonzalez M, Karkowska-Kuleta J, Zawrotniak M, Satała D, Kozik A. Living together: The role of Candida albicans in the formation of polymicrobial biofilms in the oral cavity. Yeast 2023; 40:303-317. [PMID: 37190878 DOI: 10.1002/yea.3855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
The oral cavity of humans is colonized by diversity of microbial community, although dominated by bacteria, it is also constituted by a low number of fungi, often represented by Candida albicans. Although in the vast minority, this usually commensal fungus under certain conditions of the host (e.g., immunosuppression or antibiotic therapy), can transform into an invasive pathogen that adheres to mucous membranes and also to medical or dental devices, causing mucosal infections. This transformation is correlated with changes in cell morphology from yeast-like cells to hyphae and is supported by numerous virulence factors exposed by C. albicans cells at the site of infection, such as multifunctional adhesins, degradative enzymes, or toxin. All of them affect the surrounding host cells or proteins, leading to their destruction. However, at the site of infection, C. albicans can interact with different bacterial species and in its filamentous form may produce biofilms-the elaborated consortia of microorganisms, that present increased ability to host colonization and resistance to antimicrobial agents. In this review, we highlight the modification of the infectious potential of C. albicans in contact with different bacterial species, and also consider the mutual bacterial-fungal relationships, involving cooperation, competition, or antagonism, that lead to an increase in the propagation of oral infection. The mycofilm of C. albicans is an excellent hiding place for bacteria, especially those that prefer low oxygen availability, where microbial cells during mutual co-existence can avoid host recognition or elimination by antimicrobial action. However, these microbial relationships, identified mainly in in vitro studies, are modified depending on the complexity of host conditions and microbial dominance in vivo.
Collapse
Affiliation(s)
- Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Magdalena Surowiec
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Aneta Bednarek
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Miriam Gonzalez-Gonzalez
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satała
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
31
|
Qiu XR, Shen CR, Jiang LW, Ji P, Zhang Y, Hou WT, Zhang W, Shen H, An MM. Ssa1-targeted antibody prevents host invasion by Candida albicans. Front Microbiol 2023; 14:1182914. [PMID: 37560525 PMCID: PMC10407798 DOI: 10.3389/fmicb.2023.1182914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/20/2023] [Indexed: 08/11/2023] Open
Abstract
INTRODUCTION Candida albicans is a commensal fungus that colonizes most healthy individuals' skin and mucosal surfaces but can also cause life-threatening invasive infections, particularly in immunocompromised patients. Despite antifungal treatment availability, drug resistance is increasing, and mortality rates remain unacceptably high. Heat shock protein Ssa1, a conserved member of the Hsp70 family in yeast, is a novel invasin that binds to host cell cadherins, induces host cell endocytosis, and enables C. albicans to cause maximal damage to host cells and induces disseminated and oropharyngeal disease. RESULT Here we discovered a mouse monoclonal antibody (mAb 13F4) that targeting C. albicans Ssa1 with high affinity (EC50 = 39.78 ng/mL). mAb 13F4 prevented C. albicans from adhering to and invading human epithelial cells, displayed antifungal activity, and synergized with fluconazole in proof of concept in vivo studies. mAb 13F4 significantly prolonged the survival rate of the hematogenous disseminated candidiasis mice to 75%. We constructed a mAb 13F4 three-dimensional structure using homology modeling methods and found that the antigen-binding fragment (Fab) interacts with the Ssa1 N-terminus. DISCUSSION These results suggest that blocking Ssa1 cell surface function may effectively control invasive C. albicans infections and provide a potential new treatment strategy for invasive fungal infections.
Collapse
Affiliation(s)
- Xi-Ran Qiu
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen-Rui Shen
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li-Wen Jiang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Ji
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Zhang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei-Tong Hou
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Zhang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Shen
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mao-Mao An
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Gaziano R, Sabbatini S, Monari C. The Interplay between Candida albicans, Vaginal Mucosa, Host Immunity and Resident Microbiota in Health and Disease: An Overview and Future Perspectives. Microorganisms 2023; 11:1211. [PMID: 37317186 DOI: 10.3390/microorganisms11051211] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Vulvovaginal candidiasis (VVC), which is primarily caused by Candida albicans, is an infection that affects up to 75% of all reproductive-age women worldwide. Recurrent VVC (RVVC) is defined as >3 episodes per year and affects nearly 8% of women globally. At mucosal sites of the vagina, a delicate and complex balance exists between Candida spp., host immunity and local microbial communities. In fact, both immune response and microbiota composition play a central role in counteracting overgrowth of the fungus and maintaining homeostasis in the host. If this balance is perturbed, the conditions may favor C. albicans overgrowth and the yeast-to-hyphal transition, predisposing the host to VVC. To date, the factors that affect the equilibrium between Candida spp. and the host and drive the transition from C. albicans commensalism to pathogenicity are not yet fully understood. Understanding the host- and fungus-related factors that drive VVC pathogenesis is of paramount importance for the development of adequate therapeutic interventions to combat this common genital infection. This review focuses on the latest advances in the pathogenic mechanisms implicated in the onset of VVC and also discusses novel potential strategies, with a special focus on the use of probiotics and vaginal microbiota transplantation in the treatment and/or prevention of recurrent VVC.
Collapse
Affiliation(s)
- Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06132 Perugia, Italy
| | - Claudia Monari
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06132 Perugia, Italy
| |
Collapse
|
33
|
Qi W, Dong N, Wu L, Zhang X, Li H, Wu H, Ward N, Yu J, Liu H, Wang J, Deng X, Zhao RC. Promoting oral mucosal wound healing using a DCS-RuB2A2 hydrogel based on a photoreactive antibacterial and sustained release of BMSCs. Bioact Mater 2023; 23:53-68. [DOI: 10.1016/j.bioactmat.2022.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
|
34
|
Liu H, Li Q, Xu Y, Sun Y, Fan X, Fang H, Hu B, Huang L, Liao L, Wang X. Dual-light defined in situ oral mucosal lesion therapy through a mode switchable anti-bacterial and anti-inflammatory mucoadhesive hydrogel. Biomater Sci 2023; 11:3180-3196. [PMID: 36920078 DOI: 10.1039/d2bm01721k] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Oral mucosal ulcer is the most prevalent oral mucosal lesion, affecting the quality of life. Due to the moist and highly dynamic oral lining, the existing oral mucoadhesives are unable to serially address the challenges of residency, hemorrhage, bacterial infection and inflammatory reaction. Herein, a dual-light defined oral mucoadhesive (ZPTA-G/HMA) was proposed, with a methacrylate gelatin-methacrylate hyaluronic acid (GelMA-HAMA, G/HMA) double network hydrogel as a matrix, tannic acid (TA) as a high content anchor moiety provider for the moist oral mucosa, and polydopamine modified zinc oxide (ZnO@PDA, ZP) as a photocatalytic antibacterial substance. This platform had good adhesive and hemostatic properties both in vitro and in vivo. Under 520 nm green light (GL) irradiation, ZPTA-G/HMA would anchor to the wet mucosa surface by crosslinking and exert broad-spectrum antibacterial ability (even including Candida albicans) by in situ producing reactive oxygen species (ROS). Moreover, under 808 nm near-infrared (NIR) irradiation, the increased release of TA combined with the photothermal effect of ZP endowed ZPTA-G/HMA with enhanced anti-inflammatory and pro-healing performance. Collectively, ZPTA-G/HMA could be switched by light sources to achieve the dual-mode real-time adjustment of in situ anti-bacterial function and controlled anti-inflammation, combined with ideal mucosal residence, thus promising in developing personalized sequential strategies for varied oral mucosal lesions.
Collapse
Affiliation(s)
- Huijie Liu
- The Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China. .,The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Qun Li
- The Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China. .,The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Yingying Xu
- The Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China. .,The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Yue Sun
- College of Chemistry and Chemical Engineering of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China.
| | - Xin Fan
- The Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| | - Huaqiang Fang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Binbin Hu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Li Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Lan Liao
- The Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| | - Xiaolei Wang
- College of Chemistry and Chemical Engineering of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China. .,The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| |
Collapse
|
35
|
Of Mycelium and Men: Inherent Human Susceptibility to Fungal Diseases. Pathogens 2023; 12:pathogens12030456. [PMID: 36986378 PMCID: PMC10058615 DOI: 10.3390/pathogens12030456] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
In medical mycology, the main context of disease is iatrogenic-based disease. However, historically, and occasionally, even today, fungal diseases affect humans with no obvious risk factors, sometimes in a spectacular fashion. The field of “inborn errors of immunity” (IEI) has deduced at least some of these previously enigmatic cases; accordingly, the discovery of single-gene disorders with penetrant clinical effects and their immunologic dissection have provided a framework with which to understand some of the key pathways mediating human susceptibility to mycoses. By extension, they have also enabled the identification of naturally occurring auto-antibodies to cytokines that phenocopy such susceptibility. This review provides a comprehensive update of IEI and autoantibodies that inherently predispose humans to various fungal diseases.
Collapse
|
36
|
Bellotti D, D’Accolti M, Pula W, Huang N, Simeliere F, Caselli E, Esposito E, Remelli M. Calcitermin-Loaded Smart Gels Activity against Candida albicans: A Preliminary In Vitro Study. Gels 2023; 9:gels9020165. [PMID: 36826335 PMCID: PMC9957098 DOI: 10.3390/gels9020165] [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: 01/27/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Calcitermin is an antimicrobial peptide of 15 amino acids found in human nasal fluid characterized by antifungal and antibacterial properties. Candida albicans is the most common human fungal pathogen affecting many tissues, such as vaginal mucosa. In this study a formulation suitable for calcitermin administration on vaginal mucosa was developed for the treatment of fungal infections. To favor topical application, mucosal adhesion, and permanence, gels based on poloxamer 407 and xanthan gum were designed and compared with regard to their rheological behavior, erosion, and leakage. The selected gel was loaded with calcitermin, whose release kinetic was evaluated in vitro by Franz cells. An antifungal activity assay was conducted to assess the calcitermin anticandidal potential and the effect of its inclusion in the selected gel. The rheological study revealed the elastic and viscous moduli behavior as a function of poloxamer 407 and xanthan gum concentration. Xanthan gum presence decreased the transition temperature of the gel, while prolonging its erosion and leakage. Particularly, poloxamer 407, 18% and xanthan gum 0.4% were chosen. The calcitermin loading in the selected gel resulted in a transparent and homogeneous formulation and in a 4-fold decrease of the release rate with respect to the calcitermin solution, as evidenced by Franz cell study. The anticandidal activity tests demonstrated that calcitermin-loaded gel was more active against Candida albicans with respect to the peptide solution.
Collapse
Affiliation(s)
- Denise Bellotti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland
| | - Maria D’Accolti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Walter Pula
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Nicolas Huang
- Institut Galien Paris-Saclay (CNRS UMR 8612), Faculté de Pharmacie, Bâtiment Henri Moissan, Université Paris-Saclay, 91400 Orsay, France
| | - Fanny Simeliere
- Institut Galien Paris-Saclay (CNRS UMR 8612), Faculté de Pharmacie, Bâtiment Henri Moissan, Université Paris-Saclay, 91400 Orsay, France
| | - Elisabetta Caselli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy
- Correspondence: (E.C.); (E.E.)
| | - Elisabetta Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy
- Correspondence: (E.C.); (E.E.)
| | - Maurizio Remelli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy
| |
Collapse
|
37
|
The Candida glabrata Parent Strain Trap: How Phenotypic Diversity Affects Metabolic Fitness and Host Interactions. Microbiol Spectr 2023; 11:e0372422. [PMID: 36633405 PMCID: PMC9927409 DOI: 10.1128/spectrum.03724-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Reference strains improve reproducibility by standardizing observations and methodology, which has ultimately led to important insights into fungal pathogenesis. However, recent investigations have highlighted significant genotypic and phenotypic heterogeneity across isolates that influence genetic circuitry and virulence within a species. Candida glabrata is the second leading cause of candidiasis, a life-threatening infection, and undergoes extensive karyotype and phenotypic changes in response to stress. Much of the work conducted on this pathogen has focused on two sequenced strains, CBS138 (ATCC 2001) and BG2. Few studies have compared these strains in detail, but key differences include mating type and altered patterns of expression of EPA adhesins. In fact, most C. glabrata isolates and BG2 are MATa, while CBS138 is MATα. However, it is not known if other phenotypic differences between these strains play a role in our understanding of C. glabrata pathogenesis. Thus, we set out to characterize metabolic, cell wall, and host-interaction attributes for CBS138 and BG2. We found that BG2 utilized a broader range of nitrogen sources and had reduced cell wall size and carbohydrate exposure than CBS138, which we hypothesized results in differences in innate immune interactions and virulence. We observed that, although both strains were phagocytosed to a similar extent, BG2 replicated to higher numbers in macrophages and was more virulent during Galleria mellonella infection than CBS138 in a dose-dependent manner. Interestingly, deletion of SNF3, a major nutrient sensor, did not affect virulence in G. mellonella for BG2, but significantly enhanced larval killing in the CBS138 background compared to the parent strain. Understanding these fundamental differences in metabolism and host interactions will allow more robust conclusions to be drawn in future studies of C. glabrata pathogenesis. IMPORTANCE Reference strains provide essential insights into the mechanisms underlying virulence in fungal pathogens. However, recent studies in Candida albicans and other species have revealed significant genotypic and phenotypic diversity within clinical isolates that are challenging paradigms regarding key virulence factors and their regulation. Candida glabrata is the second leading cause of candidiasis, and many studies use BG2 or CBS138 for their investigations. Therefore, we aimed to characterize important virulence-related phenotypes for both strains that might alter conclusions about C. glabrata pathogenesis. Our study provides context for metabolic and cell wall changes and how these may influence host interaction phenotypes. Understanding these differences is necessary to support robust conclusions about how virulence factors may function in these and other very different strain backgrounds.
Collapse
|
38
|
Kurakado S, Matsumoto Y, Yamada T, Shimizu K, Wakasa S, Sugita T. Tacrolimus inhibits stress responses and hyphal formation via the calcineurin signaling pathway in Trichosporon asahii. Microbiol Immunol 2023; 67:49-57. [PMID: 36398783 DOI: 10.1111/1348-0421.13039] [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: 08/05/2022] [Revised: 10/09/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
The pathogenic fungus Trichosporon asahii causes fatal deep-seated mycosis in immunocompromised patients. Calcineurin, which is widely conserved in eukaryotes, regulates cell growth and various stress responses in fungi. Tacrolimus (FK506), a calcineurin inhibitor, induces sensitivity to compounds that cause stress on the cell membrane and cell wall integrity. In this study, we demonstrated that FK506 affects stress responses and hyphal formation in T. asahii. In silico structural analysis revealed that amino acid residues in the binding site of the calcineurin-FKBP12 complex that interact with FK506 are conserved in T. asahii. The growth of T. asahii was delayed by FK506 in the presence of SDS or Congo red but not in the presence of calcium chloride. FK506 also inhibited hyphal formation in T. asahii. A mutant deficient of the cnb gene, which encodes the regulatory subunit B of calcineurin, exhibited stress sensitivities on exposure to SDS and Congo red and reduced the hyphal forming ability of T. asahii. In the cnb-deficient mutant, FK506 did not increase the stress sensitivity or reduce hyphal forming ability. These results suggest that FK506 affects stress responses and hyphal formation in T. asahii via the calcineurin signaling pathway.
Collapse
Affiliation(s)
- Sanae Kurakado
- Department of Microbiology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yasuhiko Matsumoto
- Department of Microbiology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Tsuyoshi Yamada
- Teikyo University Institute of Medical Mycology, Tokyo, Japan.,Asia International Institute of Infectious Disease Control, Teikyo University, Tokyo, Japan
| | - Kiminori Shimizu
- Department of Biological Science and Technology, Tokyo University of Science, Tokyo, Japan.,Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Shogo Wakasa
- Department of Microbiology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
39
|
Russell CM, Rybak JA, Miao J, Peters BM, Barrera FN. Candidalysin: Connecting the pore forming mechanism of this virulence factor to its immunostimulatory properties. J Biol Chem 2023; 299:102829. [PMID: 36581211 PMCID: PMC9852700 DOI: 10.1016/j.jbc.2022.102829] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
Candida albicans is a deadly pathogen responsible for millions of mucosal and systemic infections per year. The pathobiology of C. albicans is largely dependent on the damaging and immunostimulatory properties of the peptide candidalysin (CL), a key virulence factor. When CL forms pores in the plasma membrane of epithelial cells, it activates a response network grounded in activation of the epidermal growth factor receptor. Prior reviews have characterized the resulting CL immune activation schemas but lacked insights into the molecular mechanism of CL membrane damage. We recently demonstrated that CL functions by undergoing a unique self-assembly process; CL forms polymers and loops in aqueous solution prior to inserting and forming pores in cell membranes. This mechanism, the first of its kind to be observed, informs new therapeutic avenues to treat Candida infections. Recently, variants of CL were identified in other Candida species, providing an opportunity to identify the residues that are key for CL to function. In this review, we connect the ability of CL to damage cell membranes to its immunostimulatory properties.
Collapse
Affiliation(s)
- Charles M Russell
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Jennifer A Rybak
- School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, USA
| | - Jian Miao
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA.
| |
Collapse
|
40
|
Khan A, Moni SS, Ali M, Mohan S, Jan H, Rasool S, Kamal MA, Alshahrani S, Halawi M, Alhazmi HA. Antifungal Activity of Plant Secondary Metabolites on Candida albicans: An Updated Review. Curr Mol Pharmacol 2023; 16:15-42. [PMID: 35249516 DOI: 10.2174/1874467215666220304143332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/24/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Fungal infections have been increasing continuously worldwide, especially in immunocompromised individuals. Fungi, regarded as eukaryotic pathogens, have many similarities to the host cells, which inhibit anti-fungal drug development progress. Various fungal model systems have been studied, and it was concluded that Candida spp. is the most common disease-causing fungus. Candida species are well known to cause infections not only in our mouth, skin, and vagina, but they are also a frequent cause of life-threatening hospital bloodstream infections. The morphological and developmental pathways of Candida have been studied extensively, providing insight into the fungus development. Candida albicans is known to be the most pathogenic species responsible for a variety of infections in humans. Conventional anti-fungal drugs, mainly azoles drugs available in the market, have been used for years developing resistance in C. albicans. Hence, the production of new anti-fungal drugs, which require detailed molecular knowledge of fungal pathogenesis, needs to be encouraged. Therefore, this review targets the new approach of "Green Medicines" or the phytochemicals and their secondary metabolites as a source of novel anti-fungal agents to overcome the drug resistance of C. albicans, their mechanism of action, and their combined effects with the available anti-fungal drugs.
Collapse
Affiliation(s)
- Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | | | - M Ali
- Department of Pharmacognosy, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, 45142, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Huma Jan
- Department of Clinical Biochemistry, University of Kashmir, Hazratbal, Srinagar -190006, J&K, India
| | - Saiema Rasool
- Department of School Education, Govt. of Jammu & Kashmir, Srinagar, 190001 J&K, India
| | - Mohammad A Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589. Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
- Enzymoics, 7 Peterlee place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Maryam Halawi
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, 45142, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| |
Collapse
|
41
|
Beute JE, Kim AY, Park JJ, Yang A, Torres-Shafer K, Mullins DW, Sundstrom P. The IL-20RB receptor and the IL-20 signaling pathway in regulating host defense in oral mucosal candidiasis. Front Cell Infect Microbiol 2022; 12:979701. [PMID: 36225230 PMCID: PMC9548646 DOI: 10.3389/fcimb.2022.979701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Pseudomembranous candidiasis (thrush), erythematous candidiasis, and fungal esophagitis are infections of the barrier mucosa of the upper gastrointestinal tract. The majority of these infections are caused by Candida albicans, an opportunistic fungal pathogen that frequently exists as a harmless commensal on mucosal surfaces lining the gastrointestinal tract. Oral infections are initiated in the superficial stratified squamous epithelium, in which keratinocytes are the most abundant host cells and are the initial points of contact with C. albicans present in saliva. Intrinsic features of oral keratinocytes are likely to play important roles in host defense and tissue homeostasis in oral candidiasis. One understudied pathway that may be important for modulating oral candidiasis is the IL-20 cytokine signaling pathway that employs keratinocyte IL-20RB receptors as ligands for IL-19, IL-20, and IL-24. We report that production of human oral keratinocyte il24 mRNA and protein are stimulated during co-culture with C. albicans. To test the role of the IL-20 family signaling pathway in oral candidiasis, Il20rb-/- mice (lacking the IL-20RB receptor) were compared to wild-type mice in a murine model of oropharyngeal candidiasis. Fungal burdens and percent loss in body weight were determined. Despite comparable fungal burdens, the Il20rb-/- mice exhibited less weight loss over the course of their infection compared to the B6 mice, suggestive of reduced overall disease consequences in the mutant mice. Interference with IL-20 family cytokine signaling may be useful for augmenting the ability of the host to defend itself against pathogens.
Collapse
Affiliation(s)
| | - Alex Y. Kim
- Dartmouth College, Hanover, NH, United States
| | | | - Allen Yang
- Dartmouth College, Hanover, NH, United States
| | - Keshia Torres-Shafer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - David W. Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Paula Sundstrom
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- *Correspondence: Paula Sundstrom,
| |
Collapse
|
42
|
Lin Y, Yin Q, Zhuge D, Hu Y, Yang X, Tian D, Li L, Wang H, Liu S, Weng C, Zhang X, Wen B, Wang F, Yan L, Chen M, Wang L, Chen Y. Enhanced Targeting, Retention, and Penetration of Amphotericin B Through a Biomimetic Strategy to Treat Against Vulvovaginal Candidiasis. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yijing Lin
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Qingqing Yin
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Deli Zhuge
- Department of Pharmaceutics School of Pharmaceutical Sciences of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Yiqin Hu
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Xuewei Yang
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Dongyan Tian
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Li Li
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Haonan Wang
- Department of Pharmaceutics School of Pharmaceutical Sciences of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Shuangshuang Liu
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Cuiye Weng
- Laboratory Animal Center of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Xufei Zhang
- Laboratory Animal Center of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Bin Wen
- Laboratory Animal Center of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Fang Wang
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Linzhi Yan
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Mengchun Chen
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
- Department of Pharmaceutics School of Pharmaceutical Sciences of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
- Department of Pharmacy The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Ledan Wang
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
| | - Yijie Chen
- Department of Obstetrics and Gynecology The Second Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325027 P. R. China
- Department of Pharmaceutics School of Pharmaceutical Sciences of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| |
Collapse
|
43
|
Iwasawa MT, Miyachi H, Wakabayashi S, Sugihira T, Aoyama R, Nakagawa S, Katayama Y, Yoneyama M, Hara H, Iwakura Y, Matsumoto M, Inohara N, Koguchi-Yoshioka H, Fujimoto M, Núñez G, Matsue H, Nakamura Y, Saijo S. Epidermal clearance of Candida albicans is mediated by IL-17 but independent of fungal innate immune receptors. Int Immunol 2022; 34:409-420. [PMID: 35641096 PMCID: PMC9317997 DOI: 10.1093/intimm/dxac019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 05/27/2022] [Indexed: 11/12/2022] Open
Abstract
IL-17 plays important roles in host defense against Candida albicans at barrier surfaces and during invasive infection. However, the role of IL-17 in host defense after colonization of the epidermis, a main site of C. albicans infection, remains poorly understood. Using a murine model of epicutaneous candidiasis without skin abrasion, we found that skin inflammation triggered by epidermal C. albicans colonization was self-limiting with fungal clearance completed by day 7 after inoculation in wild-type mice or animals deficient in IL-17A or IL-17F. In contrast, marked neutrophilic inflammation in the epidermis and impaired fungal clearance were observed in mice lacking both IL-17A and IL-17F. Clearance of C. albicans was independent of Dectin-1, Dectin-2, CARD9 (caspase-recruitment domain family, member 9), TLR2 (Toll-like receptor 2) and MyD88 in the epidermal colonization model. We found that group 3 innate lymphoid cells (ILC3s) and γδT cells were the major IL-17 producers in the epicutaneous candidiasis model. Analyses of Rag2-/- mice and Rag2-/-Il2rg-/- mice revealed that production of IL-17A and IL-17F by ILC3s was sufficient for C. albicans clearance. Finally, we found that depletion of neutrophils impaired C. albicans clearance in the epidermal colonization model. Taken together, these findings indicate a critical and redundant function of IL-17A and IL-17F produced by ILC3s in host defense against C. albicans in the epidermis. The results also suggest that epidermal C. albicans clearance is independent of innate immune receptors or that these receptors act redundantly in fungal recognition and clearance.
Collapse
Affiliation(s)
- Mari T Iwasawa
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan
| | - Hideaki Miyachi
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan
| | - Seiichiro Wakabayashi
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan
| | - Takashi Sugihira
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Suita-shi, Osaka 565-0871, Japan
| | - Reika Aoyama
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Suita-shi, Osaka 565-0871, Japan
| | - Seitaro Nakagawa
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan
| | - Yuki Katayama
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan
| | - Mitsutoshi Yoneyama
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University , Chiba-shi, Chiba 260-8673, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-shi, Kagoshima 890-8544, Japan
| | - Yoichiro Iwakura
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University , Chiba-shi, Chiba 260-8673, Japan.,Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.,Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
| | - Masanori Matsumoto
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Naohiro Inohara
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Suita-shi, Osaka 565-0871, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Suita-shi, Osaka 565-0871, Japan.,Cutaneous Immunology, Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka 565-0871, Japan
| | - Gabriel Núñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hiroyuki Matsue
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan
| | - Yuumi Nakamura
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba-shi, Chiba 260-8670, Japan.,Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Suita-shi, Osaka 565-0871, Japan.,Cutaneous Immunology, Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka 565-0871, Japan
| | - Shinobu Saijo
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University , Chiba-shi, Chiba 260-8673, Japan
| |
Collapse
|
44
|
Holzknecht J, Dubrac S, Hedtrich S, Galgóczy L, Marx F. Small, Cationic Antifungal Proteins from Filamentous Fungi Inhibit Candida albicans Growth in 3D Skin Infection Models. Microbiol Spectr 2022; 10:e0029922. [PMID: 35499318 PMCID: PMC9241769 DOI: 10.1128/spectrum.00299-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
The emerging resistance of human-pathogenic fungi to antifungal drugs urges the development of alternative therapeutic strategies. The small, cationic antifungal proteins (AFPs) from filamentous ascomycetes represent promising candidates for next-generation antifungals. These bio-molecules need to be tested for tolerance in the host and efficacy against fungal pathogens before they can be safely applied in humans. Testing of the efficacy and possible adverse effects of new drug candidates in three-dimensional (3D) human-cell based models represents an advantageous alternative to animal experiments. In, this study, as a proof-of-principle, we demonstrate the usefulness of 3D skin infection models for screening new antifungal drug candidates for topical application. We established a cutaneous infection with the opportunistic human-pathogenic yeast Candida albicans in a commercially available 3D full-thickness (FT) skin model to test the curative potential of distinct AFPs from Penicillium chrysogenum (PAFopt, PAFB, and PAFC) and Neosartorya (Aspergillus) fischeri (NFAP2) in vitro. All tested AFPs were comparably well tolerated by the skin models. The infected 3D models exhibited reduced epidermal permeability barriers, allowing C. albicans to colonize the epidermal and dermal layers, and showed increased secretion of the pro-inflammatory cytokine IL-6 and the chemokine IL-8. AFP treatment diminished the fungal burden and penetration depth of C. albicans in the infected models. The epidermal permeability barrier was restored and the secretion of IL-8 was decreased following AFP treatment. In summary, our study proves that the tested AFPs exhibit antifungal potential against cutaneous C. albicans infection in a 3D FT skin model. IMPORTANCE Candida albicans represents one of the most prevalent opportunistic fungal pathogens, causing superficial skin and mucosal infections in humans with certain predisposing health conditions and life-threatening systemic infections in immunosuppressed patients. The emerging drug resistance of this human-pathogenic yeast and the limited number of antifungal drugs for prevention and treatment of infections urgently demands the identification of new antifungal compounds with novel mechanisms of action. Small, cationic antifungal proteins (AFPs) from filamentous fungi represent promising candidates for next-generation antifungals for topical application. These bio-molecules need to be tested for tolerance by the host and efficacy in pathogen clearance prior to being involved in clinical trials. In a proof-of-principle study, we provide evidence for the suitability of 3D human-cell based models as advantageous alternatives to animal experiments. We document the tolerance of specific AFPs and their curative efficacy against cutaneous C. albicans infection in a 3D skin model.
Collapse
Affiliation(s)
- Jeanett Holzknecht
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
45
|
The Bacillary Postbiotics, Including 2-Undecanone, Suppress the Virulence of Pathogenic Microorganisms. Pharmaceutics 2022; 14:pharmaceutics14050962. [PMID: 35631548 PMCID: PMC9143114 DOI: 10.3390/pharmaceutics14050962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/11/2022] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
Secreted molecules from probiotic Bacilli have often been considered potential pharmaceuticals to fight infections caused by bacterial or yeast pathogens. In the present study, we investigated the antagonistic potential of secreted probiotic filtrates (hereafter, postbiotics) derived from Lactobacillus plantarum cells against pathogenic microorganisms, such as Escherichia coli, Staphylococcus aureus, and Candida albicans. We found that the postbiotics mitigate the biofilms of the tested pathogens with no notable effect on their planktonic growth. In addition, the postbiotics suppressed some virulence traits, for instance, the dendrite swarming motility of E. coli and yeast-to-hyphal switch in C. albicans. Further assays with an active constituent produced by the L. plantarum cells–2-undecanone revealed two significant findings: (i) 2-undecanone inhibits C. albicans biofilms and hyphae in vitro and in a Caenorhabditis elegans model, and (ii) it interacts specifically with Gln 58 amino acid residue of hyphal wall protein-1 (Hwp-1) in molecular docking analysis. The results suggest the targeted mode of antagonistic action of 2-undecanone against C. albicans biofilm. In total, the findings of the study depict an appealing strategy to use postbiotics, including specific ketone molecules, produced by L. plantarum for developing novel antibiofilm and anti-hyphal pharmaceuticals.
Collapse
|
46
|
Nikou SA, Zhou C, Griffiths JS, Kotowicz NK, Coleman BM, Green MJ, Moyes DL, Gaffen SL, Naglik JR, Parker PJ. The Candida albicans toxin candidalysin mediates distinct epithelial inflammatory responses through p38 and EGFR-ERK pathways. Sci Signal 2022; 15:eabj6915. [PMID: 35380879 PMCID: PMC7612652 DOI: 10.1126/scisignal.abj6915] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The fungal pathogen Candida albicans secretes the peptide toxin candidalysin, which damages epithelial cells and drives an innate inflammatory response mediated by the epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase (MAPK) pathways and the transcription factor c-Fos. In cultured oral epithelial cells, candidalysin activated the MAPK p38, which resulted in heat shock protein 27 (Hsp27) activation, IL-6 release, and EGFR phosphorylation without affecting the induction of c-Fos. p38 activation was not triggered by EGFR but by two nonredundant pathways involving MAPK kinases (MKKs) and the kinase Src, which differentially controlled p38 signaling outputs. Whereas MKKs mainly promoted p38-dependent release of IL-6, Src promoted p38-mediated phosphorylation of EGFR in a ligand-independent fashion. In parallel, candidalysin also activated the EGFR-ERK pathway in a ligand-dependent manner, resulting in c-Fos activation and release of the neutrophil-activating chemokines G-CSF and GM-CSF. In mice, early clearance events of oral C. albicans infection required p38 but not c-Fos. These findings delineate how candidalysin activates the pathways downstream of the MAPKs p38 and ERK that differentially contribute to immune activation during C. albicans infection.
Collapse
Affiliation(s)
- Spyridoula-Angeliki Nikou
- Protein Phosphorylation Lab, The Francis Crick Institute; London, UK
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London; London, UK
| | - Chunsheng Zhou
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh; Pittsburgh, USA
| | - James S. Griffiths
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London; London, UK
| | - Natalia K. Kotowicz
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London; London, UK
| | - Bianca M. Coleman
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh; Pittsburgh, USA
| | - Mary J. Green
- Experimental Histopathology Lab, The Francis Crick Institute; London, UK
| | - David L. Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London; London, UK
| | - Sarah L. Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh; Pittsburgh, USA
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London; London, UK
| | - Peter J. Parker
- Protein Phosphorylation Lab, The Francis Crick Institute; London, UK
- School of Cancer and Pharmaceutical Sciences, New Hunt’s House, King’s College London; London, UK
| |
Collapse
|
47
|
Oral Cavity and Candida albicans: Colonisation to the Development of Infection. Pathogens 2022; 11:pathogens11030335. [PMID: 35335659 PMCID: PMC8953496 DOI: 10.3390/pathogens11030335] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Candida colonisation of the oral cavity increases in immunocompromised individuals which leads to the development of oral candidiasis. In addition, host factors such as xerostomia, smoking, oral prostheses, dental caries, diabetes and cancer treatment accelerate the disease process. Candida albicans is the primary causative agent of this infection, owing to its ability to form biofilm and hyphae and to produce hydrolytic enzymes and candialysin. Although mucosal immunity is activated, from the time hyphae-associated toxin is formed by the colonising C. albicans cells, an increased number and virulence of this pathogenic organism collectively leads to infection. Prevention of the development of infection can be achieved by addressing the host physiological factors and habits. For maintenance of oral health, conventional oral hygiene products containing antimicrobial compounds, essential oils and phytochemicals can be considered, these products can maintain the low number of Candida in the oral cavity and reduce their virulence. Vulnerable patients should be educated in order to increase compliance.
Collapse
|
48
|
Pimentel BNADS, Marin-Dett FH, Assis M, Barbugli PA, Longo E, Vergani CE. Antifungal Activity and Biocompatibility of α-AgVO 3, α-Ag 2WO 4, and β-Ag 2MoO 4 Using a Three-Dimensional Coculture Model of the Oral Mucosa. Front Bioeng Biotechnol 2022; 10:826123. [PMID: 35237581 PMCID: PMC8883331 DOI: 10.3389/fbioe.2022.826123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 11/18/2022] Open
Abstract
Fungal infections have become a major concern in the medical community, especially those caused by Candida spp. Within this species, Candida albicans stands out for being an opportunistic commensal fungus that can cause superficial and invasive infections. Current antifungal therapy involves the local and/or systemic use of drugs such as azoles, polyenes, and echinocandins. These antifungals are based on highly specific target sites, and the development of resistance may occur with changes in the enzymatic pathways that serve as the drug targets. Thus, the development of new antifungal drugs is highly recommended to prevent drug resistance. The present investigation evaluated the antifungal activity of silver-containing microcrystals such as silver vanadate (α-AgVO3), silver tungstate (α-Ag2WO4), and silver molybdate (β-Ag2MoO4). In addition to having antimicrobial activity, such compounds should not cause damage to underlying tissues. Thus, to better assess the biocompatibility of new compounds, a new three-dimensional (3D) coculture model involving three cell lines was developed. The validation of the model was based on fluorescent markers and confocal laser microscopy. The biocompatibility of silver-containing microcrystals was evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. 3D coculture was infected with C. albicans biofilm and challenged with α-AgVO3, α-Ag2WO4, and β-Ag2MoO4. The action of microcrystals on C. albicans biofilm was evaluated by colony-forming units (CFU/ml) and LIVE/DEAD staining. In addition, production of proinflammatory cytokines interleukin 6 (IL-6), IL-8, IL-1β, and tumor necrosis factor α (TNF-α) was measured by cytometric bead array kit using flow cytometry. The 3D coculture model described here proved to be adequate to assess both the biocompatibility of the new materials and the infectious processes. Regarding the biocompatibility of the microcrystals, only α-AgVO3 (15.62 µg/ml) showed a decrease in cell viability. The antibiofilm activity of α-Ag2WO4 was similar to that of the standard drug (fluconazole). Although α-Ag2WO4 was able to induce the production of IL-6, IL-8, and IL-1β, no differences in cytokine production were observed between noninfected and infected models treated with this microcrystal. β-Ag2MoO4 inhibits the production of TNF-α in the infected model; however, it showed no antibiofilm activity. Based on the biocompatibility and antifungal findings, α-Ag2WO4 is a promising material for treating C. albicans infection.
Collapse
Affiliation(s)
- Bruna Natália Alves da Silva Pimentel
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Freddy Humberto Marin-Dett
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Marcelo Assis
- CDMF, LIEC, Chemistry Department, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Paula Aboud Barbugli
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil.,Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Elson Longo
- CDMF, LIEC, Chemistry Department, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Carlos Eduardo Vergani
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
49
|
林 冬, 杨 利, 王 智. [Research Updates: The Role of Interaction between Oral Microbiota, Immune Cells, and Epithelial Barrier in Oral Mucosal Homeostasis and Pathogenesis]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2022; 53:188-193. [PMID: 35332716 PMCID: PMC10409361 DOI: 10.12182/20220360501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Indexed: 06/14/2023]
Abstract
In a healthy state, the interaction between the oral microorganisms, mucosal immune cells and epithelial barrier can maintain the oral microecological stability. However, the oral microecology is disrupted under a diseased state and various pathogenic bacteria and their virulence factors and metabolites irritate the immune system, which causes direct or indirect damage to the epithelial barrier, promotes the pathogenesis and progression of oral mucosal diseases, and triggers immune inflammatory response or the irreversible transformation from inflammation into cancer. We herein reviewed the interaction between oral microorganisms, immune cells and epithelial barrier from two perspectives, the maintenance of the oral homeostasis and the pathogenesis of oral mucosal diseases. We intended to gain further understanding of the oral mucosal homeostasis and the mechanism of action of the pathogenesis and progression of oral mucosal diseases, and to provide thereby ideas and scientific and theoretical basis for developing new strategies for the diagnosis and treatment of oral mucosal diseases through re-establishing mucosal homeostasis.
Collapse
Affiliation(s)
- 冬佳 林
- 中山大学光华口腔医学院·附属口腔医院 (广州 510055)Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- 广东省口腔医学重点实验室 (广州 510055)Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - 利洒 杨
- 中山大学光华口腔医学院·附属口腔医院 (广州 510055)Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- 广东省口腔医学重点实验室 (广州 510055)Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - 智 王
- 中山大学光华口腔医学院·附属口腔医院 (广州 510055)Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- 广东省口腔医学重点实验室 (广州 510055)Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
50
|
Mussi MCM, Fernandes KS, Gallottini MHC. A call for further research on the relation between type 2 diabetes and oral candidiasis. Oral Surg Oral Med Oral Pathol Oral Radiol 2022; 134:206-212. [PMID: 35422408 DOI: 10.1016/j.oooo.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE This study aimed to evaluate the relationship between oral candidiasis with salivary features, neutrophil function, and glycemic control in type 2 diabetes (T2D). DESIGN Twenty-nine individuals were included, 16 with T2D and 13 without the disease. The participants underwent clinical examination, neutrophilic function tests, fasting glycemia and glycated hemoglobin (A1c), stimulated and unstimulated saliva collection, and swab and exfoliative cytology. Salivary flow, pH, and total fungi count were evaluated on saliva, and identification of the Candida species was performed in saliva and swab samples. RESULTS There was no difference in unstimulated salivary flow and pH of the stimulated and unstimulated saliva for participants with T2D and controls (P > .05). Individuals from both groups presented no candidal lesions. The salivary fungal growth in the T2D group was higher than that in controls (P < .05). Only individuals with T2D presented alterations in the neutrophilic functions (14/16; 87.5%; P < .05). There was no relationship between high A1c values and neutrophil dysfunction with the presence of Candida spp. in both saliva and mucosa (P > .05). CONCLUSIONS High A1c level, reduction in neutrophil activity, salivary flow and pH, and increase in total salivary Candida spp. counts were not related to oral candidiasis in patients with T2D.
Collapse
|