1
|
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
|
2
|
Esfandiary MA, Khosravi AR, Asadi S, Nikaein D, Hassan J, Sharifzadeh A. Antimicrobial and anti-biofilm properties of oleuropein against Escherichia coli and fluconazole-resistant isolates of Candida albicans and Candida glabrata. BMC Microbiol 2024; 24:154. [PMID: 38704559 PMCID: PMC11069153 DOI: 10.1186/s12866-024-03305-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Side effects associated with antimicrobial drugs, as well as their high cost, have prompted a search for low-cost herbal medicinal substances with fewer side effects. These substances can be used as supplements to medicine or to strengthen their effects. The current study investigated the effect of oleuropein on the inhibition of fungal and bacterial biofilm in-vitro and at the molecular level. MATERIALS AND METHODS In this experimental study, antimicrobial properties were evaluated using microbroth dilution method. The effect of oleuropein on the formation and eradication of biofilm was assessed on 96-well flat bottom microtiter plates and their effects were observed through scanning electron microscopy (SEM). Its effect on key genes (Hwp1, Als3, Epa1, Epa6, LuxS, Pfs) involved in biofilm formation was investigated using the quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) method. RESULTS The minimum inhibitory concentration (MIC) and minimum fungicidal/bactericidal concentration (MFC/MBC) for oleuropein were found to be 65 mg/ml and 130 mg/ml, respectively. Oleuropein significantly inhibited biofilm formation at MIC/2 (32.5 mg/ml), MIC/4 (16.25 mg/ml), MIC/8 (8.125 mg/ml) and MIC/16 (4.062 mg/ml) (p < 0.0001). The anti-biofilm effect of oleuropein was confirmed by SEM. RT-qPCR indicated significant down regulation of expression genes involved in biofilm formation in Candida albicans (Hwp1, Als3) and Candida glabrata (Epa1, Epa6) as well as Escherichia coli (LuxS, Pfs) genes after culture with a MIC/2 of oleuropein (p < 0.0001). CONCLUSIONS The results indicate that oleuropein has antifungal and antibacterial properties that enable it to inhibit or destroy the formation of fungal and bacterial biofilm.
Collapse
Affiliation(s)
- Mohammad Ali Esfandiary
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, PO Box: 14155-6453, Tehran, Iran
| | - Ali Reza Khosravi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, PO Box: 14155-6453, Tehran, Iran.
| | - Sepideh Asadi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, PO Box: 14155-6453, Tehran, Iran
| | - Donya Nikaein
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, PO Box: 14155-6453, Tehran, Iran
| | - Jalal Hassan
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Aghil Sharifzadeh
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, PO Box: 14155-6453, Tehran, Iran
| |
Collapse
|
3
|
Pijuan J, Moreno DF, Yahya G, Moisa M, Ul Haq I, Krukiewicz K, Mosbah R, Metwally K, Cavalu S. Regulatory and pathogenic mechanisms in response to iron deficiency and excess in fungi. Microb Biotechnol 2023; 16:2053-2071. [PMID: 37804207 PMCID: PMC10616654 DOI: 10.1111/1751-7915.14346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023] Open
Abstract
Iron is an essential element for all eukaryote organisms because of its redox properties, which are important for many biological processes such as DNA synthesis, mitochondrial respiration, oxygen transport, lipid, and carbon metabolism. For this reason, living organisms have developed different strategies and mechanisms to optimally regulate iron acquisition, transport, storage, and uptake in different environmental responses. Moreover, iron plays an essential role during microbial infections. Saccharomyces cerevisiae has been of key importance for decrypting iron homeostasis and regulation mechanisms in eukaryotes. Specifically, the transcription factors Aft1/Aft2 and Yap5 regulate the expression of genes to control iron metabolism in response to its deficiency or excess, adapting to the cell's iron requirements and its availability in the environment. We also review which iron-related virulence factors have the most common fungal human pathogens (Aspergillus fumigatus, Cryptococcus neoformans, and Candida albicans). These factors are essential for adaptation in different host niches during pathogenesis, including different fungal-specific iron-uptake mechanisms. While being necessary for virulence, they provide hope for developing novel antifungal treatments, which are currently scarce and usually toxic for patients. In this review, we provide a compilation of the current knowledge about the metabolic response to iron deficiency and excess in fungi.
Collapse
Affiliation(s)
- Jordi Pijuan
- Laboratory of Neurogenetics and Molecular MedicineInstitut de Recerca Sant Joan de DéuBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadridSpain
| | - David F. Moreno
- Department of Molecular Cellular and Developmental BiologyYale UniversityNew HavenConnecticutUSA
- Systems Biology InstituteYale UniversityWest HavenConnecticutUSA
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of PharmacyZagazig UniversityAl SharqiaEgypt
| | - Mihaela Moisa
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| | - Ihtisham Ul Haq
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Programa de Pós‐graduação em Inovação TecnológicaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Centre for Organic and Nanohybrid ElectronicsSilesian University of TechnologyGliwicePoland
| | - Rasha Mosbah
- Infection Control UnitHospitals of Zagazig UniversityZagazigEgypt
| | - Kamel Metwally
- Department of Medicinal Chemistry, Faculty of PharmacyUniversity of TabukTabukSaudi Arabia
- Department of Pharmaceutical Medicinal Chemistry, Faculty of PharmacyZagazig UniversityZagazigEgypt
| | - Simona Cavalu
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| |
Collapse
|
4
|
Schena NC, Baker KM, Stark AA, Thomas DP, Cleary IA. Constitutive ALS3 expression in Candida albicans enhances adhesion and biofilm formation of efg1, but not cph1 mutant strains. PLoS One 2023; 18:e0286547. [PMID: 37440498 PMCID: PMC10343153 DOI: 10.1371/journal.pone.0286547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/17/2023] [Indexed: 07/15/2023] Open
Abstract
Adhesion to living and non-living surfaces is an important virulence trait of the fungal pathogen Candida albicans. Biofilm formation in this organism depends on the expression of a number of cell surface proteins including the hypha-specific protein Als3p. Loss of ALS3 impairs biofilm formation and decreases cell-cell adhesion. We wanted to test whether constitutively expressing ALS3 could compensate for defects in adhesion and biofilm formation observed in mutant strains that lack key transcriptional regulators of biofilm formation Efg1p and Cph1p. We found that ALS3 improved adhesion and biofilm formation in the efg1Δ and efg1Δ cph1Δ mutant strains, but had less effect on the cph1Δ strain.
Collapse
Affiliation(s)
- Nicholas C. Schena
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Kassandra M. Baker
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Anna A. Stark
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Derek P. Thomas
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Ian A. Cleary
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| |
Collapse
|
5
|
Martorano-Fernandes L, Goodwine JS, Ricomini-Filho AP, Nobile CJ, Del Bel Cury AA. Candida albicans Adhesins Als1 and Hwp1 Modulate Interactions with Streptococcus mutans. Microorganisms 2023; 11:1391. [PMID: 37374893 DOI: 10.3390/microorganisms11061391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Candida albicans and Streptococcus mutans are known to synergistically interact with each other in the oral cavity. For example, glucosyltransferase B (GtfB), secreted by S. mutans, can bind to the C. albicans cell surface, promoting dual-species biofilm formation. However, the fungal factors mediating interactions with S. mutans are unknown. The C. albicans adhesins Als1, Als3, and Hwp1 are key players in C. albicans single-species biofilm formation, but their roles, if any, in interacting with S. mutans have not been assessed. Here, we investigated the roles of the C. albicans cell wall adhesins Als1, Als3, and Hwp1 on forming dual-species biofilms with S. mutans. We assessed the abilities of the C. albicans wild-type als1Δ/Δ, als3Δ/Δ, als1Δ/Δ/als3Δ/Δ, and hwp1Δ/Δ strains to form dual-species biofilms with S. mutans by measuring optical density, metabolic activity, cell enumeration, biomass, thickness, and architecture of the biofilms. We observed that the C. albicans wild-type strain formed enhanced dual-species biofilms in the presence of S. mutans in these different biofilm assays, confirming that C. albicans and S. mutans synergistically interact in the context of biofilms. Our results reveal that C. albicans Als1 and Hwp1 are major players in interacting with S. mutans, since dual-species biofilm formation was not enhanced when the als1Δ/Δ or hwp1Δ/Δ strains were cultured with S. mutans in dual-species biofilms. Als3, however, does not seem to play a clear role in interacting with S. mutans in dual-species biofilm formation. Overall, our data suggest that the C. albicans adhesins Als1 and Hwp1 function to modulate interactions with S. mutans and could be potential targets for future therapeutics.
Collapse
Affiliation(s)
- Loyse Martorano-Fernandes
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
| | - James S Goodwine
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
| | - Antônio Pedro Ricomini-Filho
- Department of Physiological Science, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
- Health Sciences Research Institute, University of California Merced, Merced, CA 95343, USA
| | - Altair Antoninha Del Bel Cury
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil
| |
Collapse
|
6
|
Kanchanapiboon J, Maiuthed A, Rukthong P, Thunyaharn S, Tuntoaw S, Poonsatha S, Santimaleeworagun W. Metabolomics profiling of culture medium reveals association of methionine and vitamin B metabolisms with virulent phenotypes of clinical bloodstream-isolated Candida albicans. Res Microbiol 2023; 174:104009. [PMID: 36403754 DOI: 10.1016/j.resmic.2022.104009] [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: 07/25/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
Candida albicans is a predominant species causing candidemia in hospitalized patients. This study aimed to investigate the association of culture medium metabolomic profiles with biofilm formation and invasion properties of clinical bloodstream-isolated C. albicans. A total of twelve isolates and two reference strains were identified by virulent phenotypes. Their susceptibility was determined by the microdilution method, following EUCAST guidelines. Biofilm formation was evaluated with metabolic activity, morphology and agglutinin-like sequence 3 (ALS3) mRNA expression. Invasion into the vascular endothelial EA.hy926 cells was determined by lactate dehydrogenase release and internalization assay. Their metabolomic profiles were assessed by high-resolution accurate-mass spectrometry (HRAMS). The results showed four different phenotypes of C. albicans: high-biofilm/invasive (50%), high-biofilm/non-invasive (7%), low-biofilm/invasive (36%) and low-biofilm/non-invasive (7%). The metabolomic profiles of the culture medium determined strong correlation of the virulent phenotypes and the alteration of metabolites in the methionine metabolism pathway, such as homocysteine, 5-methyltetrahydrofolate and S-adenosylmethioninamine. Moreover, thiamine and biotin levels were significantly increased in Isolate03, representative of a high-biofilm/invasive phenotype. These results suggest that methionine and vitamin B metabolism pathways might be influenced by their virulent phenotypes and pathogenic traits. Therefore, their metabolism pathways might be a potential target for reducing virulence of C. albicans bloodstream infections.
Collapse
Affiliation(s)
- Jamras Kanchanapiboon
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi 11000, Thailand.
| | - Arnatchai Maiuthed
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Pattarawit Rukthong
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Srinakharinwirot University, Nakornnayok 26120, Thailand; Center for Excellence in Plant and Herbal Innovation Research, Strategic Wisdom and Research Institute, Srinakharinwirot University, Nakornnayok 26120, Thailand.
| | - Sudaluck Thunyaharn
- Department of Medical Technology, Faculty of Allied Health Sciences, Nakhonratchasima College, Nakhon Ratchasima 30000, Thailand.
| | - Sasiwan Tuntoaw
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi 11000, Thailand.
| | - Subhadhcha Poonsatha
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi 11000, Thailand.
| | - Wichai Santimaleeworagun
- Department of Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
7
|
Cravener MV, Do E, May G, Zarnowski R, Andes DR, McManus CJ, Mitchell AP. Reinforcement amid genetic diversity in the Candida albicans biofilm regulatory network. PLoS Pathog 2023; 19:e1011109. [PMID: 36696432 PMCID: PMC9901766 DOI: 10.1371/journal.ppat.1011109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/06/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Biofilms of the fungal pathogen Candida albicans include abundant long filaments called hyphae. These cells express hypha-associated genes, which specify diverse virulence functions including surface adhesins that ensure biofilm integrity. Biofilm formation, virulence, and hypha-associated gene expression all depend upon the transcription factor Efg1. This transcription factor has been characterized extensively in the C. albicans type strain SC5314 and derivatives, but only recently has its function been explored in other clinical isolates. Here we define a principal set of Efg1-responsive genes whose expression is significantly altered by an efg1Δ/Δ mutation across 17 clinical isolates. This principal gene set includes 68 direct Efg1 targets, whose 5' regions are bound by Efg1 in five clinical isolates, and 42 indirect Efg1 targets, whose 5' regions are not detectably bound by Efg1. Three direct Efg1 target genes encode transcription factors-BRG1, UME6, and WOR3 -whose increased expression in an efg1Δ/Δ mutant restores expression of multiple indirect and direct principal targets, as well as biofilm formation ability. Although BRG1 and UME6 are well known positive regulators of hypha-associated genes and biofilm formation, WOR3 is best known as an antagonist of Efg1 in the sexual mating pathway. We confirm the positive role of WOR3 in biofilm formation with the finding that a wor3Δ/Δ mutation impairs biofilm formation in vitro and in an in vivo biofilm model. Positive control of Efg1 direct target genes by other Efg1 direct target genes-BRG1, UME6, and WOR3 -may buffer principal Efg1-responsive gene expression against the impact of genetic variation in the C. albicans species.
Collapse
Affiliation(s)
- Max V. Cravener
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Eunsoo Do
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Gemma May
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Robert Zarnowski
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - David R. Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
8
|
Chudzik-Rząd B, Zalewski D, Kasela M, Sawicki R, Szymańska J, Bogucka-Kocka A, Malm A. The Landscape of Gene Expression during Hyperfilamentous Biofilm Development in Oral Candida albicans Isolated from a Lung Cancer Patient. Int J Mol Sci 2022; 24:ijms24010368. [PMID: 36613809 PMCID: PMC9820384 DOI: 10.3390/ijms24010368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The filamentation ability of Candida albicans represents one of the main virulence factors allowing for host tissue penetration and biofilm formation. The aim of this paper was to study the genetic background of the hyperfilamentous biofilm development in vitro in C. albicans isolated from the oral cavity of a lung cancer patient. Analyzed C. albicans isolates (CA1, CA2, CA3) were chosen based on their different structures of mature biofilm. The CA3 isolate possessing hyperfilamentation properties and forming high biofilm was compared with CA1 and CA2 isolates exhibiting low or average biofilm-forming ability, respectively. The detailed biofilm organization was studied with the use of confocal scanning laser microscopy. The whole transcriptome analysis was conducted during three stages of biofilm development (24 h, 48 h, 72 h). In contrast to CA1 and/or CA2 isolate, the CA3 isolate was characterized by a significant upregulation of genes encoding for cell wall proteins (HWP1, PGA13, PGA44, ALS3) and candidalysin (ECE1), as well as being involved in iron metabolism (FRE1, ALS3), sulfur metabolism (HAL21), the degradation of aromatic compounds (HQD2), and membrane transport (DIP5, PHO89, TNA1). In contrast, some genes (SCW11, FGR41, RBE1) in the CA3 were downregulated. We also observed the overexpression of a few genes over time-mainly FRE1, ATX1, CSA2 involved in iron metabolism. This is the first insight into the potential function of multiple genes in the hyperfilamentous biofilm formation in C. albicans, primarily isolated from host tissue, which may have an important clinical impact on cancer patients. Moreover, the presented data can lay the foundation for further research on novel pathogen-specific targets for antifungal drugs.
Collapse
Affiliation(s)
- Beata Chudzik-Rząd
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
| | - Daniel Zalewski
- Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Martyna Kasela
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
- Correspondence: (M.K.); (A.M.); Tel.: +48-81448-7100 (M.K. & A.M.)
| | - Rafał Sawicki
- Department of Biochemistry and Biotechnology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
| | - Jolanta Szymańska
- Department of Comprehensive Paediatric and Adult Dentistry, Medical University of Lublin, 6 Chodźki St., 20-093 Lublin, Poland
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
- Correspondence: (M.K.); (A.M.); Tel.: +48-81448-7100 (M.K. & A.M.)
| |
Collapse
|
9
|
Arvizu-Rubio VJ, García-Carnero LC, Mora-Montes HM. Moonlighting proteins in medically relevant fungi. PeerJ 2022; 10:e14001. [PMID: 36117533 PMCID: PMC9480056 DOI: 10.7717/peerj.14001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/13/2022] [Indexed: 01/19/2023] Open
Abstract
Moonlighting proteins represent an intriguing area of cell biology, due to their ability to perform two or more unrelated functions in one or many cellular compartments. These proteins have been described in all kingdoms of life and are usually constitutively expressed and conserved proteins with housekeeping functions. Although widely studied in pathogenic bacteria, the information about these proteins in pathogenic fungi is scarce, but there are some reports of their functions in the etiological agents of the main human mycoses, such as Candida spp., Paracoccidioides brasiliensis, Histoplasma capsulatum, Aspergillus fumigatus, Cryptococcus neoformans, and Sporothrix schenckii. In these fungi, most of the described moonlighting proteins are metabolic enzymes, such as enolase and glyceraldehyde-3-phosphate dehydrogenase; chaperones, transcription factors, and redox response proteins, such as peroxiredoxin and catalase, which moonlight at the cell surface and perform virulence-related processes, contributing to immune evasion, adhesions, invasion, and dissemination to host cells and tissues. All moonlighting proteins and their functions described in this review highlight the limited information about this biological aspect in pathogenic fungi, representing this a relevant opportunity area that will contribute to expanding our current knowledge of these organisms' pathogenesis.
Collapse
|
10
|
Rivera-Yañez CR, Ruiz-Hurtado PA, Reyes-Reali J, Mendoza-Ramos MI, Vargas-Díaz ME, Hernández-Sánchez KM, Pozo-Molina G, Méndez-Catalá CF, García-Romo GS, Pedroza-González A, Méndez-Cruz AR, Nieto-Yañez O, Rivera-Yañez N. Antifungal Activity of Mexican Propolis on Clinical Isolates of Candida Species. Molecules 2022; 27:molecules27175651. [PMID: 36080417 PMCID: PMC9457601 DOI: 10.3390/molecules27175651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Infections caused by micro-organisms of the genus Candida are becoming a growing health problem worldwide. These fungi are opportunistic commensals that can produce infections—clinically known as candidiasis—in immunocompromised individuals. The indiscriminate use of different anti-fungal treatments has triggered the resistance of Candida species to currently used therapies. In this sense, propolis has been shown to have potent antimicrobial properties and thus can be used as an approach for the inhibition of Candida species. Therefore, this work aims to evaluate the anti-Candida effects of a propolis extract obtained from the north of Mexico on clinical isolates of Candida species. Candida species were specifically identified from oral lesions, and both the qualitative and quantitative anti-Candida effects of the Mexican propolis were evaluated, as well as its inhibitory effect on C. albicans isolate’s germ tube growth and chemical composition. Three Candida species were identified, and our results indicated that the inhibition halos of the propolis ranged from 7.6 to 21.43 mm, while that of the MFC and FC50 ranged from 0.312 to 1.25 and 0.014 to 0.244 mg/mL, respectively. Moreover, the propolis was found to inhibit germ tube formation (IC50 ranging from 0.030 to 1.291 mg/mL). Chemical composition analysis indicated the presence of flavonoids, including pinocembrin, baicalein, pinobanksin chalcone, rhamnetin, and biochanin A, in the Mexican propolis extract. In summary, our work shows that Mexican propolis presents significant anti-Candida effects related to its chemical composition, and also inhibits germ tube growth. Other Candida species virulence factors should be investigated in future research in order to determine the mechanisms associated with antifungal effects against them.
Collapse
Affiliation(s)
- Claudia Rebeca Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Porfirio Alonso Ruiz-Hurtado
- Laboratorio de Toxicología de Productos Naturales, Departamento de Farmacia, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Av. Wilfrido Massieu, Gustavo A. Madero 07738, Mexico
| | - Julia Reyes-Reali
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - María Isabel Mendoza-Ramos
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - María Elena Vargas-Díaz
- Laboratorio de Química de Productos Naturales, Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala, Ciudad de México 11340, Mexico
| | - Karla Mariela Hernández-Sánchez
- Laboratorio de Química de Productos Naturales, Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala, Ciudad de México 11340, Mexico
| | - Glustein Pozo-Molina
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Claudia Fabiola Méndez-Catalá
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Gina Stella García-Romo
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Alexander Pedroza-González
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Adolfo René Méndez-Cruz
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Oscar Nieto-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (O.N.-Y.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| | - Nelly Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (O.N.-Y.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| |
Collapse
|
11
|
Rapid Hypothesis Testing in Candida albicans Clinical Isolates Using a Cloning-Free, Modular, and Recyclable System for CRISPR-Cas9 Mediated Mutant and Revertant Construction. Microbiol Spectr 2022; 10:e0263021. [PMID: 35612314 PMCID: PMC9241802 DOI: 10.1128/spectrum.02630-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
As increasing evidence emerges that interstrain genetic diversity among Candida albicans clinical isolates underpins phenotypic variation compared to the reference isolate SC5314, new genetic tools are required to interrogate gene function across strain backgrounds. Here, the SAT1-flipper plasmid was reengineered to contain a C. albicans codon optimized hygromycin B resistance gene (CaHygB). Cassettes were PCR-amplified from both SAT1-flipper and CaHygB-flipper plasmids using primers with homologous sequences flanking target genes of interest to serve as repair templates. Ribonucleoprotein (RNP) complexes containing proprietary CRISPR RNAs (crRNAs), universal transactivating CRISPR RNA (tracrRNA), and Cas9 protein were assembled in vitro and transformed, along with both repair templates, by electroporation into C. albicans. Homozygous deletion of the ADE2 gene results in red-pigmented colonies and this gene was used to validate our approach. Both in SC5314 and a variety of clinical isolates (529L, JS15, SJCA1, TW1), homozygous gene targeting was nearly 100% when plating on media containing nourseothricin and hygromycin B with transformation efficiencies exceeding 104 homozygous deletion mutants per μg of DNA. A gene reversion system was also employed with plasmids pDUP3 and pDIS3 engineered to contain the ADH1 terminator and an overlap extension PCR-mediated approach combined with CRISPR-Cas9 targeting at the NEUT5 neutral locus. A variety of single or compound mutants (Δ/Δals3, Δ/Δcph1 Δ/Δefg1, Δ/Δece1) and their revertant strains were constructed and phenotypically validated by a variety of assays, including biofilm formation, hyphal growth, and macrophage IL-1β response. Thus, we have established a cloning-free, modular system for highly efficient homozygous gene deletion and reversion in diverse isolates. IMPORTANCE Recently, phenotypic heterogeneity in Candida albicans isolates has been recognized as an underappreciated factor contributing to gene diversification and broadly impacts strain-to-strain antifungal resistance, fitness, and pathogenicity. We have designed a cloning-free genetic system for rapid gene deletion and reversion in C. albicans clinical isolates that interlaces established recyclable genetic systems with CRISPR-Cas9 technology. The SAT1-flipper was reengineered to contain CaHygB encoding resistance to hygromycin B. Using a modular PCR-mediated approach coupled with in vitro ribonucleoprotein assembly with commercial reagents, both SAT1- and CaHygB-flipper cassettes were simultaneously integrated at loci with high efficiency (104 transformants per μg DNA) and upward of 99% homozygous gene targeting across a collection of diverse isolates of various anatomical origin. Revertant strains were constructed by overlap extension PCR with CRISPR-Cas9 targeted integration at the NEUT5 locus. Thus, this facile system will aid in unraveling the genetic factors contributing to the complexity of intraspecies diversity.
Collapse
|
12
|
Pócsi I, Szigeti ZM, Emri T, Boczonádi I, Vereb G, Szöllősi J. Use of red, far-red, and near-infrared light in imaging of yeasts and filamentous fungi. Appl Microbiol Biotechnol 2022; 106:3895-3912. [PMID: 35599256 PMCID: PMC9200671 DOI: 10.1007/s00253-022-11967-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/02/2022] [Accepted: 05/07/2022] [Indexed: 11/30/2022]
Abstract
Abstract While phototoxicity can be a useful therapeutic modality not only for eliminating malignant cells but also in treating fungal infections, mycologists aiming to observe morphological changes or molecular events in fungi, especially when long observation periods or high light fluxes are warranted, encounter problems owed to altered regulatory pathways or even cell death caused by various photosensing mechanisms. Consequently, the ever expanding repertoire of visible fluorescent protein toolboxes and high-resolution microscopy methods designed to investigate fungi in vitro and in vivo need to comply with an additional requirement: to decrease the unwanted side effects of illumination. In addition to optimizing exposure, an obvious solution is red-shifted illumination, which, however, does not come without compromises. This review summarizes the interactions of fungi with light and the various molecular biology and technology approaches developed for exploring their functions on the molecular, cellular, and in vivo microscopic levels, and outlines the progress towards reducing phototoxicity through applying far-red and near-infrared light. Key points • Fungal biological processes alter upon illumination, also under the microscope • Red shifted fluorescent protein toolboxes decrease interference by illumination • Innovations like two-photon, lightsheet, and near IR microscopy reduce phototoxicity
Collapse
Affiliation(s)
- István Pócsi
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary.
| | - Zsuzsa M Szigeti
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary
| | - Tamás Emri
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary
| | - Imre Boczonádi
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary.,MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary.,Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary.,MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032, Debrecen, Hungary
| |
Collapse
|
13
|
Man A, Mare AD, Mares M, Ruta F, Pribac M, Maier AC, Cighir A, Ciurea CN. Antifungal and anti-virulence activity of six essential oils against important Candida species - a preliminary study. Future Microbiol 2022; 17:737-753. [PMID: 35531749 DOI: 10.2217/fmb-2021-0296] [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: 11/21/2022] Open
Abstract
Opportunistic infections with Candida species are becoming more problematic, considering their increasing virulence and resistance to antifungal drugs. AIM To assess the antifungal and anti-virulence activity of basil, cinnamon, clove, melaleuca, oregano and thyme essential oils (EOs) on five Candida species (C. albicans, C. auris, C. krusei C. parapsilosis and C. guillermondii). METHODS The MIC, growth rate, antibiofilm activity, regulation of gene expression (ALS3, SAP2, HSP70) and germ-tube formation were evaluated by specific methods. RESULTS Most EOs inhibited Candida species growth and reduced the expression of some virulence factors. Cinnamon and clove EO showed the most significant inhibitory effects. CONCLUSIONS The tested EOs are promising agents for facilitating the management of some Candida infections.
Collapse
Affiliation(s)
- Adrian Man
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mureș, 540142, Romania
| | - Anca-Delia Mare
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mureș, 540142, Romania
| | - Mihai Mares
- Laboratory of Antimicrobial Chemotherapy, Ion Ionescu de la Brad University of Life Sciences of Iași, Iași, 700490, Romania
| | - Florina Ruta
- Department of Community Nutrition & Food Safety, George Emil Palade University of Medicine, Pharmacy, Science, & Technology of Târgu Mureș, Târgu Mureș, 540142, Romania
| | - Mirela Pribac
- Nutrition & Holistic Health, Holomed, Târgu Mureș, 540272, Romania
| | - Adrian-Cornel Maier
- Department of Urology, "Dunarea de Jos" University of Galați, Galați, 800008, Romania
| | - Anca Cighir
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mureș, 540142, Romania.,Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science, & Technology of Târgu Mureș, Târgu Mureș, 540142, Romania
| | - Cristina-Nicoleta Ciurea
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mureș, 540142, Romania.,Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science, & Technology of Târgu Mureș, Târgu Mureș, 540142, Romania
| |
Collapse
|
14
|
Glazier VE. EFG1, Everyone’s Favorite Gene in Candida albicans: A Comprehensive Literature Review. Front Cell Infect Microbiol 2022; 12:855229. [PMID: 35392604 PMCID: PMC8980467 DOI: 10.3389/fcimb.2022.855229] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/18/2022] Open
Abstract
Candida sp. are among the most common fungal commensals found in the human microbiome. Although Candida can be found residing harmlessly on the surface of the skin and mucosal membranes, these opportunistic fungi have the potential to cause superficial skin, nail, and mucus membrane infections as well as life threatening systemic infections. Severity of infection is dependent on both fungal and host factors including the immune status of the host. Virulence factors associated with Candida sp. pathogenicity include adhesin proteins, degradative enzymes, phenotypic switching, and morphogenesis. A central transcriptional regulator of morphogenesis, the transcription factor Efg1 was first characterized in Candida albicans in 1997. Since then, EFG1 has been referenced in the Candida literature over three thousand times, with the number of citations growing daily. Arguably one of the most well studied genes in Candida albicans, EFG1 has been referenced in nearly all contexts of Candida biology from the development of novel therapeutics to white opaque switching, hyphae morphology to immunology. In the review that follows we will synthesize the research that has been performed on this extensively studied transcription factor and highlight several important unanswered questions.
Collapse
|
15
|
Fahim A, Himratul-Aznita WH, Abdul-Rahman PS, Alam MK. Efficacy of bakuchiol-garlic combination against virulent genes of C andida albicans. PeerJ 2022; 9:e12251. [PMID: 35036111 PMCID: PMC8734458 DOI: 10.7717/peerj.12251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022] Open
Abstract
Background Polymicrobial biofilms are notorious for causing intraoral tissue destruction. Streptococcus sanguinis and Streptococcus mitis, commensals of oral cavities, have been found co-existing with C. albicans in resistant oral infections. There is an urgent need to find alternative treatment options. This study aims to assess the efficacy of garlic (G) and bakuchiol (Bk) combination against candida virulent genes and their subsequently secreted proteins. Methods In vitro single species biofilms of C. albicans, and mixed species biofilms formed in combination with streptococci were exposed to bakuchiol and garlic extract (Bk+G). Gene expression of agglutinin-like sequence (ALS1), (ALS3), adhesin-like wall proteins (HWP1) and aspartyl proteinases (SAP5) were determined using qPCR and their subsequent proteins were assessed through Western blotting. Results Virulent genes were significantly downregulated in single species biofilms when they were treated with Bk+G combination. However, Bk+G did not have significant effect on ALS1 and HWP1 gene in polymicrobial biofilms. ALS3 and SAP5 were significantly downregulated in Bk+G treated polymicrobial biofilm. Similar results were portrayed in Western blotting. Conclusion Bk+G combination exhibited antimicrobial effects against single and mixed species biofilms. The findings might provide insights for treating resistant candida infections. This combination could potentially serve as an herbal alternative to traditional antifungals following further research.
Collapse
Affiliation(s)
- Ayesha Fahim
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Department of Oral Biology, University College of Dentistry, University of Lahore, Lahore, Pakistan
| | - Wan Harun Himratul-Aznita
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Puteri Shafinaz Abdul-Rahman
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,University of Malaya Centre for Proteomics Research, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohammad K Alam
- Orthodontic Division, Preventive Dentistry Department, College of Dentistry, Jouf University, Jouf, Saudi Arabia
| |
Collapse
|
16
|
Ibe C, Munro CA. Fungal Cell Wall Proteins and Signaling Pathways Form a Cytoprotective Network to Combat Stresses. J Fungi (Basel) 2021; 7:jof7090739. [PMID: 34575777 PMCID: PMC8466366 DOI: 10.3390/jof7090739] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/13/2022] Open
Abstract
Candida species are part of the normal flora of humans, but once the immune system of the host is impaired and they escape from commensal niches, they shift from commensal to pathogen causing candidiasis. Candida albicans remains the primary cause of candidiasis, accounting for about 60% of the global candidiasis burden. The cell wall of C. albicans and related fungal pathogens forms the interface with the host, gives fungal cells their shape, and also provides protection against stresses. The cell wall is a dynamic organelle with great adaptive flexibility that allows remodeling, morphogenesis, and changes in its components in response to the environment. It is mainly composed of the inner polysaccharide rich layer (chitin, and β-glucan) and the outer protein coat (mannoproteins). The highly glycosylated protein coat mediates interactions between C. albicans cells and their environment, including reprograming of wall architecture in response to several conditions, such as carbon source, pH, high temperature, and morphogenesis. The mannoproteins are also associated with C. albicans adherence, drug resistance, and virulence. Vitally, the mannoproteins contribute to cell wall construction and especially cell wall remodeling when cells encounter physical and chemical stresses. This review describes the interconnected cell wall integrity (CWI) and stress-activated pathways (e.g., Hog1, Cek1, and Mkc1 mediated pathways) that regulates cell wall remodeling and the expression of some of the mannoproteins in C. albicans and other species. The mannoproteins of the surface coat is of great importance to pathogen survival, growth, and virulence, thus understanding their structure and function as well as regulatory mechanisms can pave the way for better management of candidiasis.
Collapse
Affiliation(s)
- Chibuike Ibe
- Department of Microbiology, Faculty of Biological Sciences, Abia State University, Uturu 441107, Nigeria
- Correspondence:
| | - Carol A. Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB24 3FX, UK;
| |
Collapse
|
17
|
Development of Carbazole Derivatives Compounds against Candida albicans: Candidates to Prevent Hyphal Formation via the Ras1-MAPK Pathway. J Fungi (Basel) 2021; 7:jof7090688. [PMID: 34575726 PMCID: PMC8466151 DOI: 10.3390/jof7090688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/03/2022] Open
Abstract
Morphogenesis contributes to the virulence of the opportunistic human fungal pathogen Candida albicans. Ras1-MAPK pathways play a critical role in the virulence of C. albicans by regulating cell growth, morphogenesis, and biofilm formation. Ume6 acts as a transcription factor, and Nrg1 is a transcriptional repressor for the expression of hyphal-specific genes in morphogenesis. Azoles or echinocandin drugs have been extensively prescribed for C. albicans infections, which has led to the development of drug-resistant strains. Therefore, it is necessary to develop new molecules to effectively treat fungal infections. Here, we showed that Molecule B and Molecule C, which contained a carbazole structure, attenuated the pathogenicity of C. albicans through inhibition of the Ras1/MAPK pathway. We found that Molecule B and Molecule C inhibit morphogenesis through repressing protein and RNA levels of Ras/MAPK-related genes, including UME6 and NRG1. Furthermore, we determined the antifungal effects of Molecule B and Molecule C in vivo using a candidiasis murine model. We anticipate our findings are that Molecule B and Molecule C, which inhibits the Ras1/MAPK pathway, are promising compounds for the development of new antifungal agents for the treatment of systemic candidiasis and possibly for other fungal diseases.
Collapse
|
18
|
Liu C, Xu C, Du Y, Liu J, Ning Y. Role of agglutinin-like sequence protein 3 (Als3) in the structure and antifungal resistance of Candida albicans biofilms. FEMS Microbiol Lett 2021; 368:6316779. [PMID: 34232317 DOI: 10.1093/femsle/fnab089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Agglutinin-like sequence protein 3 (Als3) is a cell surface glycoprotein of Candida albicans that plays essential roles in the processes of adherence and biofilm formation in vitro. In this study, we focused on the contribution of Als3 to the structure and drug susceptibility of biofilms. The C. albicans wild-type (WT) strain DAY185, the als3Δ/Δ null strain and the als3Δ/Δ + pALS3 complemented strain were used. Colony-forming unit enumeration, crystal violet and cell surface hydrophobicity assays, scanning electron microscopy and confocal laser scanning microscopy coupled with analyses using COMSTAT software were performed to evaluate the biomass and architecture of the biofilms. The detailed architectural analysis showed a significant variation in the biofilm parameters of the als3Δ/Δ biofilms compared with those of the WT biofilms. Fluconazole, miconazole and amphotericin B were selected as the antifungal agents for the antimycotic susceptibility test, and increased susceptibility was found with the ALS3 deletion biofilms. A quantitative real-time polymerase chain reaction analysis showed downregulation of biofilm formation-related genes (ALS1, EFG1, HWP1 and CSH1) and drug resistance-related genes (ERG11, CDR1, CDR2 and MDR1) in the als3Δ/Δ biofilms. We concluded that Als3 contributes to biofilm formation by changing the biofilm architecture and is involved in the antifungal resistance of C. albicans biofilms.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Cheng Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Yu Du
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Jia Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Yang Ning
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou 510055, China
| |
Collapse
|
19
|
Computational Drug Repurposing Resources and Approaches for Discovering Novel Antifungal Drugs against Candida albicans N-Myristoyl Transferase. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is a yeast that is an opportunistic fungal pathogen and also identified as ubiquitous polymorphic species that is mainly linked with major fungal infections in humans, particularly in the immunocompromised patients including transplant recipients, chemotherapy patients, HIV-infected patients as well as in low-birth-weight infants. Systemic Candida infections have a high mortality rate of around 29 to 76%. For reducing its infection, limited drugs are existing such as caspofungin, fluconazole, terbinafine, and amphotericin B, etc. which contain unlikable side effects and also toxic. This review intends to utilize advanced bioinformatics technologies such as Molecular docking, Scaffold hopping, Virtual screening, Pharmacophore modeling, Molecular dynamics (MD) simulation for the development of potentially new drug candidates with a drug-repurpose approach against Candida albicans within a limited time frame and also cost reductive.
Collapse
|
20
|
Ponde NO, Lortal L, Ramage G, Naglik JR, Richardson JP. Candida albicans biofilms and polymicrobial interactions. Crit Rev Microbiol 2021; 47:91-111. [PMID: 33482069 PMCID: PMC7903066 DOI: 10.1080/1040841x.2020.1843400] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/05/2020] [Accepted: 10/25/2020] [Indexed: 12/16/2022]
Abstract
Candida albicans is a common fungus of the human microbiota. While generally a harmless commensal in healthy individuals, several factors can lead to its overgrowth and cause a range of complications within the host, from localized superficial infections to systemic life-threatening disseminated candidiasis. A major virulence factor of C. albicans is its ability to form biofilms, a closely packed community of cells that can grow on both abiotic and biotic substrates, including implanted medical devices and mucosal surfaces. These biofilms are extremely hard to eradicate, are resistant to conventional antifungal treatment and are associated with high morbidity and mortality rates, making biofilm-associated infections a major clinical challenge. Here, we review the current knowledge of the processes involved in C. albicans biofilm formation and development, including the central processes of adhesion, extracellular matrix production and the transcriptional network that regulates biofilm development. We also consider the advantages of the biofilm lifestyle and explore polymicrobial interactions within multispecies biofilms that are formed by C. albicans and selected microbial species.
Collapse
Affiliation(s)
- Nicole O. Ponde
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Gordon Ramage
- School of Medicine, Dentistry & Nursing, Glasgow Dental School and Hospital, Faculty of Medicine, University of Glasgow, G2 3JZ, United Kingdom
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| |
Collapse
|
21
|
Evolutionary Overview of Molecular Interactions and Enzymatic Activities in the Yeast Cell Walls. Int J Mol Sci 2020; 21:ijms21238996. [PMID: 33256216 PMCID: PMC7730094 DOI: 10.3390/ijms21238996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022] Open
Abstract
Fungal cell walls are composed of a polysaccharide network that serves as a scaffold in which different glycoproteins are embedded. Investigation of fungal cell walls, besides simple identification and characterization of the main cell wall building blocks, covers the pathways and regulations of synthesis of each individual component of the wall and biochemical reactions by which they are cross-linked and remodeled in response to different growth phase and environmental signals. In this review, a survey of composition and organization of so far identified and characterized cell wall components of different yeast genera including Saccharomyces, Candida, Kluyveromyces, Yarrowia, and Schizosaccharomyces are presented with the focus on their cell wall proteomes.
Collapse
|
22
|
Kanchanapiboon J, Kongsa U, Pattamadilok D, Kamponchaidet S, Wachisunthon D, Poonsatha S, Tuntoaw S. Boesenbergia rotunda extract inhibits Candida albicans biofilm formation by pinostrobin and pinocembrin. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:113193. [PMID: 32730867 DOI: 10.1016/j.jep.2020.113193] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Boesenbergia rotunda (L.) Mansf. (Zingiberaceae) is an indigenous plant of Southeast Asia. Based on ethnopharmacological use, the rhizome is recommended in the treatment of stomachache, leukoplakia, abscesses, and leukorrhea in Thailand primary health care system. Candida albicans often causes leukorrhea, and infection of many mucosal sites. Its infection leads to serious illness. AIM OF THE STUDY This study aimed to investigate the effects of the ethanolic extract of the B. rotunda rhizome on C. albicans ATCC10231 in the stages of planktonic and biofilm formation and to explore the underlying mechanisms. MATERIALS AND METHODS The chemical composition of the extract was determined using ultra-performance liquid chromatography (UPLC). The planktonic growth of C. albicans was evaluated by the microdilution method, following EUCAST guidelines. For each stage of biofilm formation, the biofilm was assessed by the MTT assay. The biofilm structure was examined under a light microscope. The degree of cell surface hydrophobicity was measured. The mRNA levels of ALS1, ALS3, and ACT1 were determined by RT-qPCR. RESULTS The extract of B. rotunda consisted of 25% (w/w) pinostrobin and 12% (w/w) pinocembrin. All stages of C. albicans biofilm formation were significantly inhibited by the extract, whereas the planktonic growth did not change. Biofilm development greatly decreased due to the extract in a concentration-dependent manner, with an IC50 value of 17.7 μg/mL. Pinostrobin and pinocembrin demonstrated inhibitory effects during this stage. These results were in accordance with the microscopic evaluation. The filamentous form decreased with pinocembrin rather than pinostrobin. Moreover, the cell surface hydrophobicity was significantly decreased by 6.25 and 12.5 μg/mL of the extract and 100 μM of pinocembrin. The ALS3 mRNA level was noticeably decreased by 12.5 μg/mL of the extract, 100 μM of pinostrobin, and 100 μM of pinocembrin. The ACT1 mRNA level decreased significantly with pinocembrin. However, the ALS1 mRNA level was not altered following all treatments. CONCLUSION The ethanolic extract of B. rotunda could inhibit biofilm formation of C. albicans, especially during the biofilm development stage, by means of reducing the cell surface hydrophobicity and suppressing the ALS3 mRNA expression. Pinocembrin had a stronger effect on ALS3 mRNA expression than pinostrobin. Only pinocembrin significantly decreased the ACT1 mRNA level.
Collapse
Affiliation(s)
- Jamras Kanchanapiboon
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand.
| | - Ubonphan Kongsa
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Duangpen Pattamadilok
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Sunisa Kamponchaidet
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Detmontree Wachisunthon
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Subhadhcha Poonsatha
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Sasiwan Tuntoaw
- Medicinal Plant Research Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, 11000, Thailand
| |
Collapse
|
23
|
Jung P, Mischo CE, Gunaratnam G, Spengler C, Becker SL, Hube B, Jacobs K, Bischoff M. Candida albicans adhesion to central venous catheters: Impact of blood plasma-driven germ tube formation and pathogen-derived adhesins. Virulence 2020; 11:1453-1465. [PMID: 33108253 PMCID: PMC7595616 DOI: 10.1080/21505594.2020.1836902] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Candida albicans-related bloodstream infections are often associated with infected central venous catheters (CVC) triggered by microbial adhesion and biofilm formation. We utilized single-cell force spectroscopy (SCFS) and flow chamber models to investigate the adhesion behavior of C. albicans yeast cells and germinated cells to naïve and human blood plasma (HBP)-coated CVC tubing. Germinated cells demonstrated up to 56.8-fold increased adhesion forces to CVC surfaces when compared to yeast cells. Coating of CVCs with HBP significantly increased the adhesion of 60-min germinated cells but not of yeast cells and 30-min germinated cells. Under flow conditions comparable to those in major human veins, germinated cells displayed a flow directional-orientated adhesion pattern to HBP-coated CVC material, suggesting the germ tip to serve as the major adhesive region. None of the above-reported phenotypes were observed with germinated cells of an als3Δ deletion mutant, which displayed similar adhesion forces to CVC surfaces as the isogenic yeast cells. Germinated cells of the als3Δ mutant also lacked a clear flow directional-orientated adhesion pattern on HBP-coated CVC material, indicating a central role for Als3 in the adhesion of germinated C. albicans cells to blood exposed CVC surfaces. In the common model of C. albicans, biofilm formation is thought to be mediated primarily by yeast cells, followed by surface-triggered the formation of hyphae. We suggest an extension of this model in which C. albicans germ tubes promote the initial adhesion to blood-exposed implanted medical devices via the germ tube-associated adhesion protein Als3.
Collapse
Affiliation(s)
- Philipp Jung
- Institute for Medical Microbiology and Hygiene, Saarland University , Homburg, Germany
| | - Clara E Mischo
- Institute for Medical Microbiology and Hygiene, Saarland University , Homburg, Germany
| | - Gubesh Gunaratnam
- Institute for Medical Microbiology and Hygiene, Saarland University , Homburg, Germany
| | | | - Sören L Becker
- Institute for Medical Microbiology and Hygiene, Saarland University , Homburg, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena (HKI) , Jena, Germany.,Institute of Microbiology, Friedrich Schiller University , Jena, Germany
| | - Karin Jacobs
- Experimental Physics, Saarland University , Saarbrücken, Germany.,Max Planck School Matter to Life , Heidelberg, Jahnstr. 29, D-69120, Germany
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygiene, Saarland University , Homburg, Germany
| |
Collapse
|
24
|
Advances in Fungal Peptide Vaccines. J Fungi (Basel) 2020; 6:jof6030119. [PMID: 32722452 PMCID: PMC7558412 DOI: 10.3390/jof6030119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/09/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Vaccination is one of the greatest public health achievements in the past century, protecting and improving the quality of life of the population worldwide. However, a safe and effective vaccine for therapeutic or prophylactic treatment of fungal infections is not yet available. The lack of a vaccine for fungi is a problem of increasing importance as the incidence of diverse species, including Paracoccidioides, Aspergillus, Candida, Sporothrix, and Coccidioides, has increased in recent decades and new drug-resistant pathogenic fungi are emerging. In fact, our antifungal armamentarium too frequently fails to effectively control or cure mycoses, leading to high rates of mortality and morbidity. With this in mind, many groups are working towards identifying effective and safe vaccines for fungal pathogens, with a particular focus of generating vaccines that will work in individuals with compromised immunity who bear the major burden of infections from these microbes. In this review, we detail advances in the development of vaccines for pathogenic fungi, and highlight new methodologies using immunoproteomic techniques and bioinformatic tools that have led to new vaccine formulations, like peptide-based vaccines.
Collapse
|
25
|
Böttcher B, Hoffmann B, Garbe E, Weise T, Cseresnyés Z, Brandt P, Dietrich S, Driesch D, Figge MT, Vylkova S. The Transcription Factor Stp2 Is Important for Candida albicans Biofilm Establishment and Sustainability. Front Microbiol 2020; 11:794. [PMID: 32425915 PMCID: PMC7203782 DOI: 10.3389/fmicb.2020.00794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/03/2020] [Indexed: 01/12/2023] Open
Abstract
The fungal pathogen Candida albicans forms polymorphic biofilms where hyphal morphogenesis and metabolic adaptation are tightly coordinated by a complex intertwined network of transcription factors. The sensing and metabolism of amino acids play important roles during various phases of biofilm development – from adhesion to maturation. Stp2 is a transcription factor that activates the expression of amino acid permease genes and is required for environmental alkalinization and hyphal growth in vitro and during macrophage phagocytosis. While it is well established that Stp2 is activated in response to external amino acids, its role in biofilm formation remains unknown. In addition to widely used techniques, we applied newly developed approaches for automated image analysis to quantify Stp2-regulated filamentation and biofilm growth. Our results show that in the stp2Δ deletion mutant adherence to abiotic surfaces and initial germ tube formation were strongly impaired, but formed mature biofilms with cell density and morphological structures comparable to the control strains. Stp2-dependent nutrient adaptation appeared to play an important role in biofilm development: stp2Δ biofilms formed under continuous nutrient flow displayed an overall reduction in biofilm formation, whereas under steady conditions the mutant strain formed biofilms with lower metabolic activity, resulting in increased cell survival and biofilm longevity. A deletion of STP2 led to increased rapamycin susceptibility and transcriptional activation of GCN4, the transcriptional regulator of the general amino acid control pathway, demonstrating a connection of Stp2 to other nutrient-responsive pathways. In summary, the transcription factor Stp2 is important for C. albicans biofilm formation, where it contributes to adherence and induction of morphogenesis, and mediates nutrient adaption and cell longevity in mature biofilms.
Collapse
Affiliation(s)
- Bettina Böttcher
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Bianca Hoffmann
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Enrico Garbe
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | | | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Philipp Brandt
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Stefanie Dietrich
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | | | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany.,Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Germany
| | - Slavena Vylkova
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| |
Collapse
|
26
|
Shinobu-Mesquita CS, Martins E, Júnior JB, de Souza Bonfim-Mendonça P, Felipe MSS, Kioshima ÉS, Svidzinski TIE. In vitro and in vivo activity of a possible novel antifungal small molecule against Candida albicans. J Mycol Med 2020; 30:100939. [PMID: 32111506 DOI: 10.1016/j.mycmed.2020.100939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 10/09/2019] [Accepted: 02/06/2020] [Indexed: 01/09/2023]
Abstract
Nosocomial infections by fungi are important causes of morbidity and mortality, and the adhesion capacity of yeast on abiotic and biotic surfaces has been considered an important step in this process. Als3 proteins are widely studied for their ability to allow Candida albicans to bind to various surfaces. The objective of the present study was to verify, with more details, the action of F2768-0318 in relation to its antifungal activity as well as its ability to act on C. albicans virulence factors related to adhesion and biofilm formation in vitro and in vivo by inhibiting the Als3 protein. F2768-0318 was assessed in tests of biofilm formation and adhesion on abiotic surfaces (polystyrene plates) and adherence on biotic surfaces, including human endocervical (HeLa) cells, human umbilical vein endothelial cells (HUVECs), and fresh buccal epithelial cells (BEC). Our results showed F2768-0318 was useful in reducing the adhesion and biofilm formation of C. albicans on abiotic surfaces, indicating the possibility of treating hospital materials and preventing biofilm formation on these types of equipment. Further studies are still needed, including optimization of the molecule to allow this molecule to be effective on other types of surfaces, such as human cells.
Collapse
Affiliation(s)
- C S Shinobu-Mesquita
- Universidade Estadual de Maringá, Departamento Análises Clínicas e Biomedicina, Av. Colombo n(o) 5790, bloco T20, 87020-900, Maringá PR, Brazil.
| | - E Martins
- Universidade Estadual de Maringá, Departamento Análises Clínicas e Biomedicina, Av. Colombo n(o) 5790, bloco T20, 87020-900, Maringá PR, Brazil
| | - J B Júnior
- Universidade Estadual de Maringá, Departamento Análises Clínicas e Biomedicina, Av. Colombo n(o) 5790, bloco T20, 87020-900, Maringá PR, Brazil
| | - P de Souza Bonfim-Mendonça
- Universidade Estadual de Maringá, Departamento Análises Clínicas e Biomedicina, Av. Colombo n(o) 5790, bloco T20, 87020-900, Maringá PR, Brazil
| | - M S S Felipe
- Universidade de Brasília, Campus Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório Biologia Molecular, Bloco K, 2 pavimento, 70910-900 Brasília, DF, Brazil
| | - É S Kioshima
- Universidade Estadual de Maringá, Departamento Análises Clínicas e Biomedicina, Av. Colombo n(o) 5790, bloco T20, 87020-900, Maringá PR, Brazil
| | - T I E Svidzinski
- Universidade Estadual de Maringá, Departamento Análises Clínicas e Biomedicina, Av. Colombo n(o) 5790, bloco T20, 87020-900, Maringá PR, Brazil
| |
Collapse
|
27
|
Selection of potential anti-adhesion drugs by in silico approaches targeted to ALS3 from Candida albicans. Biotechnol Lett 2019; 41:1391-1401. [DOI: 10.1007/s10529-019-02747-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/18/2019] [Indexed: 12/25/2022]
|
28
|
The NDV-3A vaccine protects mice from multidrug resistant Candida auris infection. PLoS Pathog 2019; 15:e1007460. [PMID: 31381597 PMCID: PMC6695204 DOI: 10.1371/journal.ppat.1007460] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 08/15/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Candida auris is an emerging, multi-drug resistant, health care-associated fungal pathogen. Its predominant prevalence in hospitals and nursing homes indicates its ability to adhere to and colonize the skin, or persist in an environment outside the host—a trait unique from other Candida species. Besides being associated globally with life-threatening disseminated infections, C. auris also poses significant clinical challenges due to its ability to adhere to polymeric surfaces and form highly drug-resistant biofilms. Here, we performed bioinformatic studies to identify the presence of adhesin proteins in C. auris, with sequence as well as 3-D structural homologies to the major adhesin/invasin of C. albicans, Als3. Anti-Als3p antibodies generated by vaccinating mice with NDV-3A (a vaccine based on the N-terminus of Als3 protein formulated with alum) recognized C. auris in vitro, blocked its ability to form biofilms and enhanced macrophage-mediated killing of the fungus. Furthermore, NDV-3A vaccination induced significant levels of C. auris cross-reactive humoral and cellular immune responses, and protected immunosuppressed mice from lethal C. auris disseminated infection, compared to the control alum-vaccinated mice. The mechanism of protection is attributed to anti-Als3p antibodies and CD4+ T helper cells activating tissue macrophages. Finally, NDV-3A potentiated the protective efficacy of the antifungal drug micafungin, against C. auris candidemia. Identification of Als3-like adhesins in C. auris makes it a target for immunotherapeutic strategies using NDV-3A, a vaccine with known efficacy against other Candida species and safety as well as efficacy in clinical trials. Considering that C. auris can be resistant to almost all classes of antifungal drugs, such an approach has profound clinical relevance. Candida auris has emerged as a major health concern to hospitalized patients and nursing home subjects. C. auris strains display multidrug resistance to current antifungal therapy and cause lethal infections. We have determined that C. auris harbors homologs of C. albicans Als cell surface proteins. The C. albicans NDV-3A vaccine, harboring the N-terminus of Als3p formulated with alum, generates cross-reactive antibodies against C. auris clinical isolates and protects neutropenic mice from hematogenously disseminated C. auris infection. Importantly, the NDV-3A vaccine displays an additive protective effect in neutropenic mice when combined with micafungin. Due to its proven safety and efficacy in humans against C. albicans infection, our studies support the expedited testing of the NDV-3A vaccine against C. auris in future clinical trials.
Collapse
|
29
|
Contributions of Candida albicans Dimorphism, Adhesive Interactions, and Extracellular Matrix to the Formation of Dual-Species Biofilms with Streptococcus gordonii. mBio 2019; 10:mBio.01179-19. [PMID: 31213561 PMCID: PMC6581863 DOI: 10.1128/mbio.01179-19] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial communities have a great impact in health and disease. C. albicans interacts with multiple microorganisms in the oral cavity, frequently forming polymicrobial biofilms. We report on the synergistic interactions between C. albicans and the Gram-positive bacterium S. gordonii, for which we have examined the different contributions of adhesive interactions, filamentation, and the extracellular matrix to the formation of dual-species biofilms. Our results demonstrate that growth in the presence of the bacterium can restore the biofilm-forming ability of different C. albicans mutant strains with defects in adhesion and filamentation. The mixed-species biofilms also show high levels of resistance to antibacterial and antifungal antibiotics, and our results indicate that the fungal biofilm matrix protects bacterial cells within these mixed-species biofilms. Our observations add to a growing body of evidence indicating a high level of complexity in the reciprocal interactions and consortial behavior of fungal/bacterial biofilms. Fungal and bacterial populations coexist in the oral cavity, frequently forming mixed-species biofilms that complicate treatment against polymicrobial infections. However, despite relevance to oral health, the bidirectional interactions between these microbial populations are poorly understood. In this study, we aimed to elucidate the mechanisms underlying the interactions between the fungal species Candida albicans and the bacterial species Streptococcus gordonii as they coexist in mixed-species biofilms. Specifically, the interactions of different C. albicans mutant strains deficient in filamentation (efg1Δ/Δ and brg1Δ/Δ), adhesive interactions (als3Δ/Δ and bcr1Δ/Δ), and production of matrix exopolymeric substances (EPS) (kre5Δ/Δ, mnn9Δ/Δ, rlm1Δ/Δ, and zap1Δ/Δ) were evaluated with S. gordonii under different conditions mimicking the environment in the oral cavity. Interestingly, our results revealed that growth of the biofilm-deficient C. albicansals3Δ/Δ and bcr1Δ/Δ mutant strains in synthetic saliva or with S. gordonii restored their biofilm-forming ability. Moreover, challenging previous observations indicating an important role of morphogenetic conversions in the interactions between these two species, our results indicated a highly synergistic interaction between S. gordonii and the C. albicans filamentation-deficient efg1Δ/Δ and brg1Δ/Δ deletion mutants, which was particularly noticeable when the mixed biofilms were grown in synthetic saliva. Importantly, dual-species biofilms were found to exhibit increase in antimicrobial resistance, indicating that components of the fungal exopolymeric material confer protection to streptococcal cells against antibacterial treatment. Collectively, these findings unravel a high degree of complexity in the interactions between C. albicans and S. gordonii in mixed-species biofilms, which may impact homeostasis in the oral cavity.
Collapse
|
30
|
de Jong AW, Hagen F. Attack, Defend and Persist: How the Fungal Pathogen Candida auris was Able to Emerge Globally in Healthcare Environments. Mycopathologia 2019; 184:353-365. [PMID: 31209693 DOI: 10.1007/s11046-019-00351-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022]
Abstract
Within a decade after its first description, the multidrug-resistant yeast Candida auris has emerged globally as a nosocomial pathogen causing difficult to control outbreaks. This, together with the alarmingly high mortality rate of up to 66% associated with C. auris candidemia, calls for a better understanding of its virulence traits and routes of transmission. Unlike other clinically relevant Candida species, C. auris seems to have the unique ability to be easily transmitted between patients. Although initially thought to express fewer virulence traits than Candida albicans, recent genomic insights suggest C. auris to possess these traits to a much more similar extent. This review highlights the virulence traits C. auris expresses to attack the host, defend itself against antimicrobial agents and to persist within the healthcare environment.
Collapse
Affiliation(s)
- Auke W de Jong
- Department of Medical Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Ferry Hagen
- Department of Medical Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, Shandong, People's Republic of China.
| |
Collapse
|
31
|
Candida spp. and phagocytosis: multiple evasion mechanisms. Antonie van Leeuwenhoek 2019; 112:1409-1423. [PMID: 31079344 DOI: 10.1007/s10482-019-01271-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/02/2019] [Indexed: 01/01/2023]
Abstract
Invasive fungal infections are a global health problem, mainly in hospitals, where year by year hundreds of patients die because of these infections. Commensal yeasts may become pathogenic to human beings, affecting mainly immunocompromised patients. During infectious processes, the immune system uses phagocytes to eliminate invader microorganisms. In order to prevent or neutralize phagocyte attacks, pathogenic yeasts can use virulence factors to survive, as well as to colonize and infect the host. In this review, we describe how Candida spp., mainly Candida albicans, interact with phagocytes and use several factors that contribute to immune evasion. Polymorphism, biofilm formation, gene expression and enzyme production mediate distinct functions such as adhesion, invasion, oxidative stress response, proteolysis and escape from phagocytes. Fungal and human cells have similar structures and mechanisms that decrease the number of potential targets for antifungal drugs. Therefore, research on host-pathogen interaction may aid in the discovery of new targets and in the development of new drugs or treatments for these diseases and thus to save lives.
Collapse
|
32
|
NDV-3A vaccination prevents C. albicans colonization of jugular vein catheters in mice. Sci Rep 2019; 9:6194. [PMID: 30996274 PMCID: PMC6470131 DOI: 10.1038/s41598-019-42517-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
NDV-3A, a novel fungal vaccine undergoing clinical trials, contains a recombinant version of the Candida albicans rAls3 N-terminus protein (rAls3p-N) in aluminum hydroxide. In a Phase 1b/2a clinical trial, NDV-3A protected women from recurrent vulvovaginal candidiasis. Here, we reveal that active immunization in mice with NDV-3A induces high titers of anti-rAls3p-N antibodies that interfere with C. albicans ability to adhere to and invade endothelial cells, and form biofilm in vitro. Anti-rAls3p-N antibodies also significantly inhibit yeast dispersal from the hyphal layers of biofilms. Compared to placebo, NDV-3A vaccination inhibited C. albicans dissemination to kidneys and prevented colonization of central venous catheters in mice. Overall, these preclinical studies suggest that NDV-3A may serve as an immunotherapeutic strategy for prevention of infections on indwelling medical devices.
Collapse
|
33
|
|
34
|
Barbosa AH, Damasceno JL, Casemiro LA, Martins CHG, Pires RH, Candido RC. Susceptibility to Oral Antiseptics and Virulence Factors Ex Vivo Associated with Candida spp. Isolated from Dental Prostheses. J Prosthodont 2019; 28:398-408. [PMID: 30768738 DOI: 10.1111/jopr.13037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2019] [Indexed: 12/16/2022] Open
Abstract
PURPOSE To isolate Candida spp. from dental prosthesis users' saliva and to evaluate the isolates for the presence of several virulence factors. This research also aimed to investigate the antifungal activity of 3 commercial mouthwashes/oral antiseptic formulations containing 0.12% chlorhexidine, 0.07% cetylpyridinium, or 0.075% cetylpyridinium against planktonic and sessile (biofilm mode) yeast cells. MATERIALS AND METHODS Forty-three Candida yeasts were isolated from 32 of 70 selected patients, and the virulence factors of C. albicans, C. krusei, C. glabrata, C. tropicalis, and C. parapsilosis species were investigated by polymerase chain reaction (PCR) and proteinase in plates. Minimum inhibitory concentration (MIC), and in vitro biofilm assay evaluated the antifungal activity of antiseptics. RESULTS C. albicans, C. krusei, C. glabrata, C. tropicalis, and C. parapsilosis were detected in mono and mixed cultures. Only C. albicans displayed genes related to adhesion and proteinases (ALS2, ALS3, SAP1, and SAP3). The aspartate proteinase activity was found in 60.46% of isolates. The tested antiseptic formulations exhibited a MIC less than 1.25% toward yeasts in the planktonic mode. According to XTT ((2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) assay results, most Candida isolates and all mixed cultures formed biofilms within 24 hours. The evaluated antiseptic formulations were also active against biofilms. CONCLUSION Most virulence factors investigated here (ALS2, ALS3, SAP1, and SAP3) occurred in the majority of the Candida spp. isolates, especially in C. albicans. The tested mouthwash formulations were effective against all the yeast isolates in both the planktonic and sessile growth modes. Developing alternative therapies that can avoid or control biofilm formation is necessary to prevent oral candidiasis and other Candida spp. infections.
Collapse
Affiliation(s)
| | | | | | | | - Regina Helena Pires
- Laboratory of Research in Applied Microbiology (LAPEMA), University of Franca, São Paulo, Brazil
| | - Regina Célia Candido
- School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
35
|
Kean R, Delaney C, Sherry L, Borman A, Johnson EM, Richardson MD, Rautemaa-Richardson R, Williams C, Ramage G. Transcriptome Assembly and Profiling of Candida auris Reveals Novel Insights into Biofilm-Mediated Resistance. mSphere 2018; 3:e00334-18. [PMID: 29997121 PMCID: PMC6041501 DOI: 10.1128/msphere.00334-18] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022] Open
Abstract
Candida auris has emerged as a significant global nosocomial pathogen. This is primarily due to its antifungal resistance profile but also its capacity to form adherent biofilm communities on a range of clinically important substrates. While we have a comprehensive understanding of how other Candida species resist and respond to antifungal challenge within the sessile phenotype, our current understanding of C. auris biofilm-mediated resistance is lacking. In this study, we are the first to perform transcriptomic analysis of temporally developing C. auris biofilms, which were shown to exhibit phase- and antifungal class-dependent resistance profiles. A de novo transcriptome assembly was performed, where sequenced sample reads were assembled into an ~11.5-Mb transcriptome consisting of 5,848 genes. Differential expression (DE) analysis demonstrated that 791 and 464 genes were upregulated in biofilm formation and planktonic cells, respectively, with a minimum 2-fold change. Adhesin-related glycosylphosphatidylinositol (GPI)-anchored cell wall genes were upregulated at all time points of biofilm formation. As the biofilm developed into intermediate and mature stages, a number of genes encoding efflux pumps were upregulated, including ATP-binding cassette (ABC) and major facilitator superfamily (MFS) transporters. When we assessed efflux pump activity biochemically, biofilm efflux was greater than that of planktonic cells at 12 and 24 h. When these were inhibited, fluconazole sensitivity was enhanced 4- to 16-fold. This study demonstrates the importance of efflux-mediated resistance within complex C. auris communities and may explain the resistance of C. auris to a range of antimicrobial agents within the hospital environment.IMPORTANCE Fungal infections represent an important cause of human morbidity and mortality, particularly if the fungi adhere to and grow on both biological and inanimate surfaces as communities of cells (biofilms). Recently, a previously unrecognized yeast, Candida auris, has emerged globally that has led to widespread concern due to the difficulty in treating it with existing antifungal agents. Alarmingly, it is also able to grow as a biofilm that is highly resistant to antifungal agents, yet we are unclear about how it does this. Here, we used a molecular approach to investigate the genes that are important in causing the cells to be resistant within the biofilm. The work provides significant insights into the importance of efflux pumps, which actively pump out toxic antifungal drugs and therefore enhance fungal survival within a variety of harsh environments.
Collapse
Affiliation(s)
- Ryan Kean
- Oral Sciences Research Group, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Institute of Healthcare, Policy and Practise, University of the West of Scotland, Paisley, United Kingdom
| | - Christopher Delaney
- Oral Sciences Research Group, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Leighann Sherry
- Oral Sciences Research Group, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrew Borman
- National Mycology Reference Laboratory, Public Health England South-West, Bristol, United Kingdom
| | - Elizabeth M Johnson
- National Mycology Reference Laboratory, Public Health England South-West, Bristol, United Kingdom
| | - Malcolm D Richardson
- Mycology Reference Centre Manchester, University Hospital of South Manchester & University of Manchester, Manchester Academic Health Sciences Centre, Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, Manchester, United Kingdom
| | - Riina Rautemaa-Richardson
- Mycology Reference Centre Manchester, University Hospital of South Manchester & University of Manchester, Manchester Academic Health Sciences Centre, Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, Manchester, United Kingdom
| | - Craig Williams
- Institute of Healthcare, Policy and Practise, University of the West of Scotland, Paisley, United Kingdom
- ESCMID Study Group for Biofilms (ESGB)‡
| | - Gordon Ramage
- Oral Sciences Research Group, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- ESCMID Study Group for Biofilms (ESGB)‡
| |
Collapse
|
36
|
Uppuluri P, Singh S, Alqarihi A, Schmidt CS, Hennessey JP, Yeaman MR, Filler SG, Edwards JE, Ibrahim AS. Human Anti-Als3p Antibodies Are Surrogate Markers of NDV-3A Vaccine Efficacy Against Recurrent Vulvovaginal Candidiasis. Front Immunol 2018; 9:1349. [PMID: 29963049 PMCID: PMC6013566 DOI: 10.3389/fimmu.2018.01349] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/31/2018] [Indexed: 12/14/2022] Open
Abstract
A Phase 1b/2a clinical trial of NDV-3A vaccine containing a Candida albicans recombinant Als3 protein formulated with alum protected women <40 years old from recurrent vulvovaginal candidiasis (RVVC). We investigated the potential use of anti-Als3p sera as surrogate marker of NDV-3A efficacy. Pre- and post-vaccination sera from subjects who experienced recurrence of vulvovaginal candidiasis (R) vs. those who were recurrence-free [non-recurrent (NR)] were evaluated. Anti-Als3p antisera obtained were evaluated for (1) titer and subclass profile and (2) their ability to influence C. albicans virulence traits including hyphal elongation, adherence to plastic, invasion of vaginal epithelial cells, biofilm formation on plastic and catheter material, and susceptibility to neutrophil killing in vitro. Serum IgG titers in NR patients were consistently higher than in R patients, particularly for anti-Als3 subclass IgG2. Sera from vaccinated NR patients reduced hyphal elongation, adhesion to plastic, invasion of vaginal epithelial cells, and biofilm formation significantly more than pre-immune sera, or sera from R- or placebo-group subjects. Pre-adsorption of sera with C. albicans germ tubes eliminated these effects, while heat inactivation did not. Finally, sera from NR subjects enhanced neutrophil-mediated killing of C. albicans relative to pre-immune sera or sera from R patients. Our results suggest that higher Als3p antibody titers are associated with protection from RVVC, attenuate C. albicans virulence, and augment immune clearance of the fungus in vitro. Thus, Als3p serum IgG antibodies are likely useful markers of efficacy in RVVC patients vaccinated with NDV-3A.
Collapse
Affiliation(s)
- Priya Uppuluri
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Shakti Singh
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Abdullah Alqarihi
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States
| | | | | | - Michael R Yeaman
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Scott G Filler
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - John E Edwards
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Ashraf S Ibrahim
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
37
|
Community Development between Porphyromonas gingivalis and Candida albicans Mediated by InlJ and Als3. mBio 2018; 9:mBio.00202-18. [PMID: 29691333 PMCID: PMC5915736 DOI: 10.1128/mbio.00202-18] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pleiomorphic yeast Candida albicans is a significant pathogen in immunocompromised individuals. In the oral cavity, C. albicans is an inhabitant of polymicrobial communities, and interspecies interactions promote hyphal formation and biofilm formation. C. albicans colonizes the subgingival area, and the frequency of colonization increases in periodontal disease. In this study, we investigated the interactions between C. albicans and the periodontal pathogen Porphyromonas gingivalisC. albicans and P. gingivalis were found to coadhere in both the planktonic and sessile phases. Loss of the internalin-family protein InlJ abrogated adhesion of P. gingivalis to C. albicans, and recombinant InlJ protein competitively inhibited interspecies binding. A mutant of C. albicans deficient in expression of major hyphal protein Als3 showed diminished binding to P. gingivalis, and InlJ interacted with Als3 heterologously expressed in Saccharomyces cerevisiae Transcriptional profiling by RNA sequencing (RNA-Seq) established that 57 genes were uniquely upregulated in an InlJ-dependent manner in P. gingivalis-C. albicans communities, with overrepresentation of those corresponding to 31 gene ontology terms, including those associated with growth and division. Of potential relevance to the disease process, C. albicans induced upregulation of components of the type IX secretion apparatus. Collectively, these findings indicate that InlJ-Als3-dependent binding facilitates interdomain community development between C. albicans and P. gingivalis and that P. gingivalis has the potential for increased virulence within such communities.IMPORTANCE Many diseases involve the concerted actions of microorganisms assembled in polymicrobial communities. Inflammatory periodontal diseases are among the most common infections of humans and result in destruction of gum tissue and, ultimately, in loss of teeth. In periodontal disease, pathogenic communities can include the fungus Candida albicans; however, the contribution of C. albicans to the synergistic virulence of the community is poorly understood. Here we characterize the interactions between C. albicans and the keystone bacterial pathogen Porphyromonas gingivalis and show that coadhesion mediated by specific proteins results in major changes in gene expression by P. gingivalis, which could serve to increase pathogenic potential. The work provides significant insights into interdomain interactions that can enhance our understanding of diseases involving a multiplicity of microbial pathogens.
Collapse
|
38
|
Manfiolli AO, Dos Reis TF, de Assis LJ, de Castro PA, Silva LP, Hori JI, Walker LA, Munro CA, Rajendran R, Ramage G, Goldman GH. Mitogen activated protein kinases (MAPK) and protein phosphatases are involved in Aspergillus fumigatus adhesion and biofilm formation. Cell Surf 2018; 1:43-56. [PMID: 32743127 PMCID: PMC7389341 DOI: 10.1016/j.tcsw.2018.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/08/2018] [Accepted: 03/14/2018] [Indexed: 12/28/2022] Open
Abstract
The main characteristic of biofilm formation is extracellular matrix (ECM) production. The cells within the biofilm are surrounded by ECM which provides structural integrity and protection. During an infection, this protection is mainly against cells of the immune system and antifungal drugs. A. fumigatus forms biofilms during static growth on a solid substratum and in chronic aspergillosis infections. It is important to understand how, and which, A. fumigatus signal transduction pathways are important for the adhesion and biofilm formation in a host during infection. Here we investigated the role of MAP kinases and protein phosphatases in biofilm formation. The loss of the MAP kinases MpkA, MpkC and SakA had an impact on the cell surface and the ECM during biofilm formation and reduced the adherence of A. fumigatus to polystyrene and fibronectin-coated plates. The phosphatase null mutants ΔsitA and ΔptcB, involved in regulation of MpkA and SakA phosphorylation, influenced cell wall carbohydrate exposure. Moreover, we characterized the A. fumigatus protein phosphatase PphA. The ΔpphA strain was more sensitive to cell wall-damaging agents, had increased β-(1,3)-glucan and reduced chitin, decreased conidia phagocytosis by Dictyostelium discoideum and reduced adhesion and biofilm formation. Finally, ΔpphA strain was avirulent in a murine model of invasive pulmonary aspergillosis and increased the released of tumor necrosis factor alpha (TNF-α) from bone marrow derived macrophages (BMDMs). These results show that MAP kinases and phosphatases play an important role in signaling pathways that regulate the composition of the cell wall, extracellular matrix production as well as adhesion and biofilm formation in A. fumigatus.
Collapse
Affiliation(s)
- Adriana Oliveira Manfiolli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Thaila Fernanda Dos Reis
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Leandro José de Assis
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Patrícia Alves de Castro
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Lilian Pereira Silva
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Juliana I Hori
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Louise A Walker
- School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Carol A Munro
- School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Ranjith Rajendran
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, The University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK
| | - Gordon Ramage
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, The University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK
| | - Gustavo H Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
39
|
Sui X, Yan L, Jiang YY. The vaccines and antibodies associated with Als3p for treatment of Candida albicans infections. Vaccine 2017; 35:5786-5793. [DOI: 10.1016/j.vaccine.2017.08.082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/14/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022]
|
40
|
Candida albicans fatty acyl-CoA synthetase, CaFaa4p, is involved in the uptake of exogenous long-chain fatty acids and cell activity in the biofilm. Curr Genet 2017; 64:429-441. [PMID: 28942495 DOI: 10.1007/s00294-017-0751-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/09/2017] [Accepted: 09/12/2017] [Indexed: 01/25/2023]
Abstract
Fatty acyl-CoA synthetase (Faa) activates fatty acid (FA) by converting the FA into the CoA ester in the cell. In the present study, we characterized a FAA homologue (CaFAA4) from the opportunistic pathogen Candida albicans. Most organisms can not only synthesize long-chain fatty acyl-CoAs (LCFA-CoAs) endogenously using a fatty acid synthase (Fas) activity but also can uptake long-chain fatty acids (LCFAs) from the extracellular environment and convert them into LCFA-CoAs via a vectorial acylation system. The budding yeast Saccharomyces cerevisiae possesses two LCFA-CoA synthetases, ScFaa1p and ScFaa4p. The disruption of ScFAA1 and ScFAA4 leads to synthetic lethality in the presence of a fatty acid synthesis inhibitor-cerulenin. The homologue-CaFAA4-rescued the lethality of an S. cerevisiae Scfaa1-Scfaa4 double mutant in the presence of cerulenin. On the other hand, a C. albicans faa4 mutant was unable to grow in the presence of cerulenin even if LCFAs were provided exogenously. Moreover, a biofilm analysis showed that the metabolic activity of the Cafaa4 mutant was approximately 40% lower than that of the wild-type parent, even though there was no significant difference in cell number or cell morphology between these strains. Notably, the Cafaa4 mutant showed increased susceptibility to micafungin during biofilm formation, a phenotype that presumably can be attributed to the impaired metabolism of the mutant strain. These results indicated that CaFaa4p is the unique C. albicans Faa protein responsible for activating LCFAs and is involved in the metabolism of biofilms.
Collapse
|
41
|
Protocol for Identifying Natural Agents That Selectively Affect Adhesion, Thickness, Architecture, Cellular Phenotypes, Extracellular Matrix, and Human White Blood Cell Impenetrability of Candida albicans Biofilms. Antimicrob Agents Chemother 2017; 61:AAC.01319-17. [PMID: 28893778 DOI: 10.1128/aac.01319-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/04/2017] [Indexed: 11/20/2022] Open
Abstract
In the screening of natural plant extracts for antifungal activity, assessment of their effects on the growth of cells in suspension or in the wells of microtiter plates is expedient. However, microorganisms, including Candida albicans, grow in nature as biofilms, which are organized cellular communities with a complex architecture capable of conditioning their microenvironment, communicating, and excluding low- and high-molecular-weight molecules and white blood cells. Here, a confocal laser scanning microscopy (CLSM) protocol for testing the effects of large numbers of agents on biofilm development is described. The protocol assessed nine parameters from a single z-stack series of CLSM scans for each individual biofilm analyzed. The parameters included adhesion, thickness, formation of a basal yeast cell polylayer, hypha formation, the vertical orientation of hyphae, the hyphal bend point, pseudohypha formation, calcofluor white staining of the extracellular matrix (ECM), and human white blood cell impenetrability. The protocol was applied first to five plant extracts and derivative compounds and then to a collection of 88 previously untested plant extracts. They were found to cause a variety of phenotypic profiles, as was the case for 64 of the 88 extracts (73%). Half of the 46 extracts that did not affect biofilm thickness affected other biofilm parameters. Correlations between specific effects were revealed. The protocol will be useful not only in the screening of chemical libraries but also in the analysis of compounds with known effects and mutations.
Collapse
|
42
|
Abstract
We focus this article on turning a biofilm inside out. The "inside" of the biofilm comprises the individual biofilm-related phenotypes, their environmental drivers and genetic determinants, and the coordination of gene functions through transcriptional regulators. Investigators have viewed the inside of the biofilm through diverse approaches, and this article will attempt to capture the essence of many. The ultimate goal is to connect the inside to the "outside," which we view as biofilm structure, development, pharmacological attributes, and medical impact.
Collapse
|
43
|
Hwang G, Liu Y, Kim D, Li Y, Krysan DJ, Koo H. Candida albicans mannans mediate Streptococcus mutans exoenzyme GtfB binding to modulate cross-kingdom biofilm development in vivo. PLoS Pathog 2017; 13:e1006407. [PMID: 28617874 PMCID: PMC5472321 DOI: 10.1371/journal.ppat.1006407] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/09/2017] [Indexed: 01/08/2023] Open
Abstract
Candida albicans is frequently detected with heavy infection by Streptococcus mutans in plaque-biofilms from children with early-childhood caries (ECC). This cross-kingdom biofilm contains an extensive matrix of extracellular α-glucans that is produced by an exoenzyme (GtfB) secreted by S. mutans. Here, we report that mannans located on the outer surface of C. albicans cell-wall mediates GtfB binding, enhancing glucan-matrix production and modulating bacterial-fungal association within biofilms formed in vivo. Using single-molecule atomic force microscopy, we determined that GtfB binds with remarkable affinity to mannans and to the C. albicans surface, forming a highly stable and strong bond (1-2 nN). However, GtfB binding properties to C. albicans was compromised in strains defective in O-mannan (pmt4ΔΔ) or N-mannan outer chain (och1ΔΔ). In particular, the binding strength of GtfB on och1ΔΔ strain was severely disrupted (>3-fold reduction vs. parental strain). In turn, the GtfB amount on the fungal surface was significantly reduced, and the ability of C. albicans mutant strains to develop mixed-species biofilms with S. mutans was impaired. This phenotype was independent of hyphae or established fungal-biofilm regulators (EFG1, BCR1). Notably, the mechanical stability of the defective biofilms was weakened, resulting in near complete biomass removal by shear forces. In addition, these in vitro findings were confirmed in vivo using a rodent biofilm model. Specifically, we observed that C. albicans och1ΔΔ was unable to form cross-kingdom biofilms on the tooth surface of rats co-infected with S. mutans. Likewise, co-infection with S. mutans defective in GtfB was also incapable of forming mixed-species biofilms. Taken together, the data support a mechanism whereby S. mutans-secreted GtfB binds to the mannan layer of C. albicans to promote extracellular matrix formation and their co-existence within biofilms. Enhanced understanding of GtfB-Candida interactions may provide new perspectives for devising effective therapies to disrupt this cross-kingdom relationship associated with an important childhood oral disease.
Collapse
Affiliation(s)
- Geelsu Hwang
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Yuan Liu
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Dongyeop Kim
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Yong Li
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Damian J. Krysan
- Department of Pediatrics, Infectious Diseases and Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Hyun Koo
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
44
|
The Emerging Pathogen Candida auris: Growth Phenotype, Virulence Factors, Activity of Antifungals, and Effect of SCY-078, a Novel Glucan Synthesis Inhibitor, on Growth Morphology and Biofilm Formation. Antimicrob Agents Chemother 2017; 61:AAC.02396-16. [PMID: 28223375 PMCID: PMC5404565 DOI: 10.1128/aac.02396-16] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/05/2017] [Indexed: 12/23/2022] Open
Abstract
Candidaauris, a new multidrug-resistant Candida spp. which is associated with invasive infection and high rates of mortality, has recently emerged. Here, we determined the virulence factors (germination, adherence, biofilm formation, phospholipase and proteinase production) of 16 C. auris isolates and their susceptibilities to 11 drugs belonging to different antifungal classes, including a novel orally bioavailable 1,3-β-d-glucan synthesis inhibitor (SCY-078). We also examined the effect of SCY-078 on the growth, ultrastructure, and biofilm-forming abilities of C. auris. Our data showed that while the tested strains did not germinate, they did produce phospholipase and proteinase in a strain-dependent manner and had a significantly reduced ability to adhere and form biofilms compared to that of Candida albicans (P = 0.01). C. auris isolates demonstrated reduced susceptibility to fluconazole and amphotericin B, while, in general, they were susceptible to the remaining drugs tested. SCY-078 had an MIC90 of 1 mg/liter against C. auris and caused complete inhibition of the growth of C. auris and C. albicans. Scanning electron microscopy analysis showed that SCY-078 interrupted C. auris cell division, with the organism forming abnormal fused fungal cells. Additionally, SCY-078 possessed potent antibiofilm activity, wherein treated biofilms demonstrated significantly reduced metabolic activity and a significantly reduced thickness compared to the untreated control (P < 0.05 for both comparisons). Our study shows that C. auris expresses several virulence determinants (albeit to a lesser extent than C. albicans) and is resistant to fluconazole and amphotericin B. SCY-078, the new orally bioavailable antifungal, had potent antifungal/antibiofilm activity against C. auris, indicating that further evaluation of this antifungal is warranted.
Collapse
|
45
|
Penicillenols from a deep-sea fungus Aspergillus restrictus inhibit Candida albicans biofilm formation and hyphal growth. J Antibiot (Tokyo) 2017. [DOI: 10.1038/ja.2017.45 pmid: 283776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
46
|
Wang J, Yao QF, Amin M, Nong XH, Zhang XY, Qi SH. Penicillenols from a deep-sea fungus Aspergillus restrictus inhibit Candida albicans biofilm formation and hyphal growth. J Antibiot (Tokyo) 2017; 70:763-770. [PMID: 28377634 DOI: 10.1038/ja.2017.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 02/13/2017] [Accepted: 03/03/2017] [Indexed: 12/19/2022]
Abstract
Penicillenols (A1, A2, B1, B2, C1 and C2) were isolated from Aspergillus restrictus DFFSCS006, and could differentially inhibit biofilm formation and eradicate pre-developed biofilms of Candida albicans. Their structure-bioactivity relationships suggested that the saturation of hydrocarbon chain at C-8, R-configuration of C-5 and trans-configuration of the double bond between C-5 and C-6 of pyrrolidine-2,4-dione unit were important for their anti-biofilm activities. Penicillenols A2 and B1 slowed the hyphal growth and suppressed the transcripts of hypha specific genes HWP1, ALS1, ALS3, ECE1 and SAP4. Moreover, penicillenols A2 and B1 were found to act synergistically with amphotericin B against C. albicans biofilm formation.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Qi-Feng Yao
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Muhammad Amin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Xu-Hua Nong
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Xiao-Yong Zhang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Shu-Hua Qi
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou, China
| |
Collapse
|
47
|
Abstract
We focus this article on turning a biofilm inside out. The "inside" of the biofilm comprises the individual biofilm-related phenotypes, their environmental drivers and genetic determinants, and the coordination of gene functions through transcriptional regulators. Investigators have viewed the inside of the biofilm through diverse approaches, and this article will attempt to capture the essence of many. The ultimate goal is to connect the inside to the "outside," which we view as biofilm structure, development, pharmacological attributes, and medical impact.
Collapse
Affiliation(s)
- Katherine Lagree
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Aaron P Mitchell
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
48
|
Real-Time Approach to Flow Cell Imaging of Candida albicans Biofilm Development. J Fungi (Basel) 2017; 3:jof3010013. [PMID: 29371532 PMCID: PMC5715965 DOI: 10.3390/jof3010013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/17/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
The ability of Candida albicans to form biofilms is a virulence factor that allows tissue attachment and subsequent infection of host tissues. Fungal biofilms have been particularly well studied, however the vast majority of these studies have been conducted under static conditions. Oral biofilms form in the presence of salivary flow, therefore we developed a novel flow system used for real-time imaging of fungal biofilm development. C. albicans wild-type (WT) cells readily attached to the substrate surface during the 2 h attachment phase, then formed heterogeneous biofilms after 18 h flow. Quantitative values for biomass, rates of attachment and detachment, and cell-cell adhesion events were obtained for C. albicans WT cells and for a hyperfilamentous mutant Δhog1. Attachment rates of C. albicans WT cells were nearly 2-fold higher than C. albicans Δhog1 cells, although Δhog1 cells formed 4-fold higher biomass. The reduced normalized detachment rate was the primary factor responsible for the increased biomass of Δhog1 biofilm, showing that cell detachment rates are an important predictor for ultimate biofilm mass under flow. Unlike static biofilms, C. albicans cells under constant laminar flow undergo continuous detachment and seeding that may be more representative of the development of in vivo biofilms.
Collapse
|
49
|
The Emerging Pathogen Candida auris: Growth Phenotype, Virulence Factors, Activity of Antifungals, and Effect of SCY-078, a Novel Glucan Synthesis Inhibitor, on Growth Morphology and Biofilm Formation. Antimicrob Agents Chemother 2017. [PMID: 28223375 DOI: 10.1128/aac.02396-16.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candidaauris, a new multidrug-resistant Candida spp. which is associated with invasive infection and high rates of mortality, has recently emerged. Here, we determined the virulence factors (germination, adherence, biofilm formation, phospholipase and proteinase production) of 16 C. auris isolates and their susceptibilities to 11 drugs belonging to different antifungal classes, including a novel orally bioavailable 1,3-β-d-glucan synthesis inhibitor (SCY-078). We also examined the effect of SCY-078 on the growth, ultrastructure, and biofilm-forming abilities of C. auris Our data showed that while the tested strains did not germinate, they did produce phospholipase and proteinase in a strain-dependent manner and had a significantly reduced ability to adhere and form biofilms compared to that of Candida albicans (P = 0.01). C. auris isolates demonstrated reduced susceptibility to fluconazole and amphotericin B, while, in general, they were susceptible to the remaining drugs tested. SCY-078 had an MIC90 of 1 mg/liter against C. auris and caused complete inhibition of the growth of C. auris and C. albicans Scanning electron microscopy analysis showed that SCY-078 interrupted C. auris cell division, with the organism forming abnormal fused fungal cells. Additionally, SCY-078 possessed potent antibiofilm activity, wherein treated biofilms demonstrated significantly reduced metabolic activity and a significantly reduced thickness compared to the untreated control (P < 0.05 for both comparisons). Our study shows that C. auris expresses several virulence determinants (albeit to a lesser extent than C. albicans) and is resistant to fluconazole and amphotericin B. SCY-078, the new orally bioavailable antifungal, had potent antifungal/antibiofilm activity against C. auris, indicating that further evaluation of this antifungal is warranted.
Collapse
|
50
|
Abstract
Candida albicans, the most pervasive fungal pathogen that colonizes humans, forms biofilms that are architecturally complex. They consist of a basal yeast cell polylayer and an upper region of hyphae encapsulated in extracellular matrix. However, biofilms formed in vitro vary as a result of the different conditions employed in models, the methods used to assess biofilm formation, strain differences, and, in a most dramatic fashion, the configuration of the mating type locus (MTL). Therefore, integrating data from different studies can lead to problems of interpretation if such variability is not taken into account. Here we review the conditions and factors that cause biofilm variation, with the goal of engendering awareness that more attention must be paid to the strains employed, the methods used to assess biofilm development, every aspect of the model employed, and the configuration of the MTL locus. We end by posing a set of questions that may be asked in comparing the results of different studies and developing protocols for new ones. This review should engender the notion that not all biofilms are created equal.
Collapse
Affiliation(s)
- David R Soll
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| | - Karla J Daniels
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|