51
|
Wang D, Zeng N, Li C, Li Z, Zhang N, Li B. Fungal biofilm formation and its regulatory mechanism. Heliyon 2024; 10:e32766. [PMID: 38988529 PMCID: PMC11233959 DOI: 10.1016/j.heliyon.2024.e32766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 07/12/2024] Open
Abstract
Fungal biofilm is a microbial community composed of fungal cells and extracellular polymeric substances (EPS). In recent years, fungal biofilms have played an increasingly important role in many fields. However, there are few studies on fungal biofilms and their related applications and development are still far from enough. Therefore, this review summarizes the composition and function of EPS in fungal biofilms, and improves and refines the formation process of fungal biofilms according to the latest viewpoints. Moreover, based on the study of Saccharomyces cerevisiae and Candida albicans, this review summarizes the gene regulation network of fungal biofilm synthesis, which is crucial for systematically understanding the molecular mechanism of fungal biofilm formation. It is of great significance to further develop effective methods at the molecular level to control harmful biofilms or enhance and regulate the formation of beneficial biofilms. Finally, the quorum sensing factors and mixed biofilms formed by fungi in the current research of fungal biofilms are summarized. These results will help to deepen the understanding of the formation process and internal regulation mechanism of fungal biofilm, provide reference for the study of EPS composition and structure, formation, regulation, group behavior and mixed biofilm formation of other fungal biofilms, and provide strategies and theoretical basis for the control, development and utilization of fungal biofilms.
Collapse
Affiliation(s)
- Dandan Wang
- College of Land and Environment, Shenyang Agricultural University, Shenyang, 110866, PR China
| | - Nan Zeng
- College of Land and Environment, Shenyang Agricultural University, Shenyang, 110866, PR China
| | - Chunji Li
- Innovative Institute for Plant Health, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, PR China
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China, Ministry of Agriculture and Rural Affairs, Guangzhou, 510225, PR China
- College of Agriculture and Biology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, PR China
| | - Zijing Li
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, PR China
| | - Ning Zhang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, PR China
| | - Bingxue Li
- College of Land and Environment, Shenyang Agricultural University, Shenyang, 110866, PR China
| |
Collapse
|
52
|
Barbosa PF, Gonçalves DS, Ramos LS, Mello TP, Braga-Silva LA, Pinto MR, Taborda CP, Branquinha MH, Santos ALS. Saps1-3 Antigens in Candida albicans: Differential Modulation Following Exposure to Soluble Proteins, Mammalian Cells, and Infection in Mice. Infect Dis Rep 2024; 16:572-586. [PMID: 39051243 PMCID: PMC11270244 DOI: 10.3390/idr16040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The secreted aspartic peptidases (Saps) of Candida albicans play crucial roles in various steps of fungal-host interactions. Using a flow cytometry approach, this study investigated the expression of Saps1-3 antigens after (i) incubation with soluble proteins, (ii) interaction with mammalian cells, and (iii) infection in immunosuppressed BALB/c mice. Supplementation strategies involving increasing concentrations of bovine serum albumin (BSA) added to yeast carbon base (YCB) medium as the sole nitrogenous source revealed a positive and significant correlation between BSA concentration and both the growth rate and the percentage of fluorescent cells (%FC) labeled with anti-Saps1-3 antibodies. Supplementing the YCB medium with various soluble proteins significantly modulated the expression of Saps1-3 antigens in C. albicans. Specifically, immunoglobulin G, gelatin, and total bovine/human sera significantly reduced the %FC, while laminin, human serum albumin, fibrinogen, hemoglobin, and mucin considerably increased the %FC compared to BSA. Furthermore, co-cultivating C. albicans yeasts with either live epithelial or macrophage cells induced the expression of Saps1-3 antigens in 78% (mean fluorescence intensity [MFI] = 152.1) and 82.7% (MFI = 178.2) of the yeast cells, respectively, compared to BSA, which resulted in 29.3% fluorescent cells (MFI = 50.9). Lastly, the yeasts recovered from the kidneys of infected immunosuppressed mice demonstrated a 4.8-fold increase in the production of Saps1-3 antigens (MFI = 246.6) compared to BSA, with 95.5% of yeasts labeled with anti-Saps1-3 antibodies. Altogether, these results demonstrated the positive modulation of Saps' expression in C. albicans by various key host proteinaceous components, as well as by in vitro and in vivo host challenges.
Collapse
Affiliation(s)
- Pedro F. Barbosa
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Diego S. Gonçalves
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Thaís P. Mello
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Lys A. Braga-Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Marcia R. Pinto
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense (UFF), Niterói 24210-130, Brazil;
| | - Carlos P. Taborda
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo 05508-060, Brazil;
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-901, Brazil
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-901, Brazil
| |
Collapse
|
53
|
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
|
54
|
Kurnia D, Lestari S, Mayanti T, Gartika M, Nurdin D. Anti-Infection of Oral Microorganisms from Herbal Medicine of Piper crocatum Ruiz & Pav. Drug Des Devel Ther 2024; 18:2531-2553. [PMID: 38952486 PMCID: PMC11215520 DOI: 10.2147/dddt.s453375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/20/2024] [Indexed: 07/03/2024] Open
Abstract
The WHO Global Status Report on Oral Health 2022 reveals that oral diseases caused by infection with oral pathogenic microorganisms affect nearly 3.5 billion people worldwide. Oral health problems are caused by the presence of S. mutans, S. sanguinis, E. faecalis and C. albicans in the oral cavity. Synthetic anti-infective drugs have been widely used to treat oral infections, but have been reported to cause side effects and resistance. Various strategies have been implemented to overcome this problem. Synthetic anti-infective drugs have been widely used to treat oral infections, but they have been reported to cause side effects and resistance. Therefore, it is important to look for safe anti-infective alternatives. Ethnobotanical and ethnopharmacological studies suggest that Red Betel leaf (Piper crocatum Ruiz & Pav) could be a potential source of oral anti-infectives. This review aims to discuss the pathogenesis mechanism of several microorganisms that play an important role in causing health problems, the mechanism of action of synthetic oral anti-infective drugs in inhibiting microbial growth in the oral cavity, and the potential of red betel leaf (Piper crocatum Ruiz & Pav) as an herbal oral anti-infective drug. This study emphasises the importance of researching natural components as an alternative treatment for oral infections that is more effective and can meet global needs.
Collapse
Affiliation(s)
- Dikdik Kurnia
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Seftiana Lestari
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Tri Mayanti
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Meirina Gartika
- Department of Pediatric Dentistry, Faculty of Medicine, University of Padjadjaran, Bandung, Indonesia
| | - Denny Nurdin
- Departement of Conservative Dentistry, Faculty of Dentistry, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
55
|
Vasconcelos PGS, Abuna GF, Raimundo e Silva JP, Tavares JF, Costa EMMDB, Murata RM. Syzygium aromaticum essential oil and its major constituents: Assessment of activity against Candida spp. and toxicity. PLoS One 2024; 19:e0305405. [PMID: 38889118 PMCID: PMC11185461 DOI: 10.1371/journal.pone.0305405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/28/2024] [Indexed: 06/20/2024] Open
Abstract
Syzigium aromaticum essential oil (EO), eugenol, and β-caryophyllene were evaluated regarding antifungal, antibiofilm, and in vitro toxicity. Additionally, in vivo toxicity of EO was observed. Anti-Candida activity was assessed through broth microdilution assay for all compounds. Time-kill assay (0, 1, 10, 30 min, 1, 2, and 4 h) was used to determine the influence of EO and eugenol on Candida Growth kinetics. Thereafter, both compounds were evaluated regarding their capacity to act on a biofilm formation and on mature biofilm, based on CFU/ml/g of dry weight. Cell Titer Blue Viability Assay was used for in vitro cytotoxicity, using oral epithelial cells (TR146) and human monocytes (THP-1). Lastly, Galleria mellonella model defined the EO in vivo acute toxicity. All compounds, except β-cariofilene (MIC > 8000 μg/ml), presented antifungal activity against Candida strains (MIC 500-1000 μg/ml). The growth kinetics of Candida was affected by the EO (5xMIC 30 min onward; 10xMIC 10 min onward) and eugenol (5xMIC 10 min onward; 10xMIC 1 min onward). Fungal viability was also affected by 5xMIC and 10xMIC of both compounds during biofilm formation and upon mature biofilms. LD50 was defined for TR146 and THP1 cells at, respectively, 59.37 and 79.54 μg/ml for the EO and 55.35 and 84.16 μg/ml for eugenol. No sign of toxicity was seen in vivo up to 10mg/ml (20 x MIC) for the EO. S. aromaticum and eugenol presented antifungal and antibiofilm activity, with action on cell growth kinetics. In vivo acute toxicity showed a safe parameter for the EO up to 10 mg/ml.
Collapse
Affiliation(s)
| | - Gabriel Flores Abuna
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | | | - Josean Fechine Tavares
- Multi-User Laboratory for Characterization and Analysis, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Edja Maria Melo de Brito Costa
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Paraíba, Brazil
| | - Ramiro Mendonça Murata
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, North Carolina, United States of America
| |
Collapse
|
56
|
Wu S, Jia W, Lu Y, Jiang H, Huang C, Tang S, Du L. Mechanism and bioinformatics analysis of the effect of berberine-enhanced fluconazole against drug-resistant Candida albicans. BMC Microbiol 2024; 24:196. [PMID: 38849761 PMCID: PMC11157861 DOI: 10.1186/s12866-024-03334-0] [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: 08/19/2023] [Accepted: 05/16/2024] [Indexed: 06/09/2024] Open
Abstract
Biofilms produced by Candida albicans present a challenge in treatment with antifungal drug. Enhancing the sensitivity to fluconazole (FLC) is a reasonable method for treating FLC-resistant species. Moreover, several lines of evidence have demonstrated that berberine (BBR) can have antimicrobial effects. The aim of this study was to clarify the underlying mechanism of these effects. We conducted a comparative study of the inhibition of FLC-resistant strain growth by FLC treatment alone, BBR treatment alone, and the synergistic effect of combined FLC and BBR treatment. Twenty-four isolated strains showed distinct biofilm formation capabilities. The antifungal effect of combined FLC and BBR treatment in terms of the growth and biofilm formation of Candida albicans species was determined via checkerboard, time-kill, and fluorescence microscopy assays. The synergistic effect of BBR and FLC downregulated the expression of the efflux pump genes CDR1 and MDR, the hyphal gene HWP1, and the adhesion gene ALS3; however, the gene expression of the transcriptional repressor TUP1 was upregulated following treatment with this drug combination. Furthermore, the addition of BBR led to a marked reduction in cell surface hydrophobicity. To identify resistance-related genes and virulence factors through genome-wide sequencing analysis, we investigated the inhibition of related resistance gene expression by the combination of BBR and FLC, as well as the associated signaling pathways and metabolic pathways. The KEGG metabolic map showed that the metabolic genes in this strain are mainly involved in amino acid and carbon metabolism. The metabolic pathway map showed that several ergosterol (ERG) genes were involved in the synthesis of cell membrane sterols, which may be related to drug resistance. In this study, BBR + FLC combination treatment upregulated the expression of the ERG1, ERG3, ERG4, ERG5, ERG24, and ERG25 genes and downregulated the expression of the ERG6 and ERG9 genes compared with fluconazole treatment alone (p < 0.05).
Collapse
Affiliation(s)
- Sitong Wu
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital, Liu Zhou, 545006, China
| | - Wei Jia
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yu Lu
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital, Liu Zhou, 545006, China
| | - Hongkun Jiang
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital, Liu Zhou, 545006, China
| | - Chunlan Huang
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital, Liu Zhou, 545006, China
| | - Shifu Tang
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital, Liu Zhou, 545006, China
| | - Le Du
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital, Liu Zhou, 545006, China.
| |
Collapse
|
57
|
Kechi EL, Ubah CB, Runde M, Owen AE, Godfrey OC, Agurokpon DC, Odey MO, Edet UO, Ekpong BO, Iyam SO, Benjamin I, Sampathkumar G. Elucidating the structural basis for the enhanced antifungal activity of amide derivative against Candida albicans: a comprehensive computational investigation. In Silico Pharmacol 2024; 12:48. [PMID: 38828443 PMCID: PMC11139824 DOI: 10.1007/s40203-024-00222-3] [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: 03/22/2024] [Accepted: 05/18/2024] [Indexed: 06/05/2024] Open
Abstract
The continuous search for more effective options against well-known pathogens such as Candida albicans remains the rationale for the search for novel lead compounds from various sources. This study aims to investigate the chemical structure, chemical properties, of 5-(2-((5-(((1S,3R) -3-(5-acetamido-1,3,4-thiadiazolidin-2-yl) cyclopentyl) methyl)-1,3,4-thiadiazolidin-2-yl)amino)-2-oxoethyl)-2-methyl-2,3-dihydro-1H-pyrazol-3-ide designated ATCTP using DFT method ωB97XD/-311 + + g(2d, 2p) and the biological potential of compound ATCTP against Candida albicans using molecular docking and ADMET studies. Geometry optimization was carried out in DMSO, ethanol. gas and water revealing minute discrepancies in bond length and wider differences in bond angles. Frontier molecular orbital investigations reveal HOMO-LUMO energy gap magnitude in decreasing order of ATCTP_Gas > ATCTP_Water > ATCTP_ethanol > ATCTP_DMSO inferring that water influences chemical stability of the compound the most compared to ethanol and DMSO. Density of state investigations have revealed electron density contributions at corresponding energy peaks. In silico pharmacokinetic predicts ATCTP not to be cytotoxic, hepatotoxic, immunotoxic or mutagenic but probable mutagen. Molecular docking investigation of ATCTP against aspartic proteinase of Candida albicans (ID: 2QZX) in comparison with standard drug Fluconazole. Compound ATCTP had higher binding affinity (- 8.1 kcal/mol) compared to that of the standard drug fluconazole (- 5.6 kcal/mol) which records 4 conventional hydrogen interactions compared to 2 formed in the interaction of ATCTP + 2QZX. ATCTP also reports binding affinity of - 7.2 kcal/mol which reportedly surpassed that of 2QZX interaction with fluconazole (- 5.7 kcal/mol). ATCTP binds with lanosterol14-α-demethylase (5v5z) with binding affinity of - 9.7 kcal/mol binding to active site amino acid residues of the protein compared to fluconazole + 5v5z (- 8.0 kcal/mol). ATCTP is therefore recommended to be a lead compound for the possible design of a new and more effective anti-candida therapeutic compound.
Collapse
Affiliation(s)
- Eban L. Kechi
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
- Department of Pharmacology, University of Calabar, Calabar, Nigeria
| | - Chioma B. Ubah
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| | - Musa Runde
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
- Department of Chemistry, National Open University of Nigeria, Abuja, Nigeria
| | - Aniekan E. Owen
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
- Department of Chemistry, Akwa Ibom State University, Uyo, Nigeria
| | - Obinna C. Godfrey
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
- Department of Biochemistry, University of Calabar, Calabar, Nigeria
| | - Daniel C. Agurokpon
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| | - Michael O. Odey
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
- Department of Biochemistry, University of Calabar, Calabar, Nigeria
| | - Uwem O. Edet
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| | - Bassey O. Ekpong
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| | - Solomon O. Iyam
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| | - Innocent Benjamin
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, University of Calabar, Calabar, Nigeria
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| | - Gopinath Sampathkumar
- Department of Chemistry, Chettinad College of Engineering and Technology, Karur, Tamilnadu India
- Department of Genetics and Biotechnology, University of Calabar, Calabar, Nigeria
| |
Collapse
|
58
|
Liu Y, Wang R, Liu J, Fan M, Ye Z, Hao Y, Xie F, Wang T, Jiang Y, Liu N, Cui X, Lv Q, Yan L. The vacuolar fusion regulated by HOPS complex promotes hyphal initiation and penetration in Candida albicans. Nat Commun 2024; 15:4131. [PMID: 38755250 PMCID: PMC11099166 DOI: 10.1038/s41467-024-48525-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
The transition between yeast and hyphae is crucial for regulating the commensalism and pathogenicity in Candida albicans. The mechanisms that affect the invasion of hyphae in solid media, whose deficiency is more related to the pathogenicity of C. albicans, have not been elucidated. Here, we found that the disruption of VAM6 or VPS41 which are components of the homotypic vacuolar fusion and protein sorting (HOPS) complex, or the Rab GTPase YPT72, all responsible for vacuole fusion, led to defects in hyphal growth in both liquid and solid media, but more pronounced on solid agar. The phenotypes of vac8Δ/Δ and GTR1OE-vam6Δ/Δ mutants indicated that these deficiencies are mainly caused by the reduced mechanical forces that drive agar and organs penetration, and confirmed that large vacuoles are required for hyphal mechanical penetration. In summary, our study revealed that large vacuoles generated by vacuolar fusion support hyphal penetration and provided a perspective to refocus attention on the role of solid agar in evaluating C. albicans invasion.
Collapse
Affiliation(s)
- Yu Liu
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Ruina Wang
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Jiacun Liu
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Mengting Fan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, PR China
| | - Zi Ye
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Yumeng Hao
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Fei Xie
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Ting Wang
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China
| | - Yuanying Jiang
- School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Ningning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| | - Xiaoyan Cui
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, PR China.
| | - Quanzhen Lv
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China.
| | - Lan Yan
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, 200433, PR China.
| |
Collapse
|
59
|
Hu QL, Zhong H, Wang XR, Han L, Ma SS, Li L, Wang Y. Mitochondrial phosphate carrier plays an important role in virulence of Candida albicans. Mycology 2024; 16:369-381. [PMID: 40083413 PMCID: PMC11899212 DOI: 10.1080/21501203.2024.2354876] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/08/2024] [Indexed: 03/16/2025] Open
Abstract
Candida albicans is a common fungal pathogen that can cause life-threatening infections. MIR1 is considered to be a mitochondrial phosphate carrier of C. albicans, while its role in virulence has not been fully elucidated. In this study, we found that mir1Δ/Δ mutant exhibited severe virulence defect in both nematode and murine models. Further mechanism studies revealed that the mir1Δ/Δ mutant grew more slowly than the wild-type strain and showed severe filamentation defects on the hypha-inducing agar media, including YPD + serum, Lee, Spider + glucose, SLAD, SLD, and YPS. Furthermore, the loss of MIR1 resulted in unfermentable carbon utilisation defect, ATP decrease, and reactive oxygen species (ROS) accumulation in C. albicans. Antioxidant proanthocyanidins, vitamin E, and N-acetyl cysteine (NAC) could reduce intracellular ROS levels and partially rescue the filamentation defects of the mir1Δ/Δ mutant. Accordingly, hypha-specific genes, as well as CEK1 and RIM101 were down-regulated in the mir1Δ/Δ mutant, and this down-regulation could be partially rescued by the addition of the antioxidant NAC. Collectively, MIR1 plays an important role in C. albicans mitochondrial function, filamentation and virulence, and would be a promising antifungal target.
Collapse
Affiliation(s)
- Qiao-Ling Hu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Hua Zhong
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Xin-Rong Wang
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Lei Han
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shan-Shan Ma
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Ling Li
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yan Wang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, China
- The Center for Fungal Infectious Diseases Basic Research and Innovation of Medicine and Pharmacy, Ministry of Education, Shanghai, China
| |
Collapse
|
60
|
Xiong L, Goerlich K, Mitchell AP. Regulatory features of Candida albicans hemin-induced filamentation. G3 (BETHESDA, MD.) 2024; 14:jkae053. [PMID: 38470537 PMCID: PMC11075532 DOI: 10.1093/g3journal/jkae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
Candida albicans is a prominent fungal pathogen that can infect the bloodstream and deep tissues. One key pathogenicity trait is the ability to transition between yeast and hyphal growth. Hyphae are critical for the formation of biofilms, which in turn enable device-associated infection. Among signals that drive hypha formation is the presence of hemin, an oxidized Fe(III)-containing heme derivative found in blood. In this study, we asked 4 questions. First, how uniform is the filamentation response to hemin among C. albicans strains? We tested 26 diverse isolates and found that the strength of a strain's filamentation response to hemin reflected its filamentation level in the absence of hemin. Second, does hemin induce biofilm formation? Hemin biofilm induction was evident in 5 out of 10 isolates tested, including most of the weaker biofilm formers tested. Third, what is the gene expression response to hemin? We compared RNA-seq data for type strain SC5314 grown in pH 5.5 minimal media with or without hemin. We also compared that response to SC5314 grown in pH 7.0 minimal media, where it undergoes well-studied pH-dependent filamentation. We found a common set of 72 genes with upregulated RNA levels in response to both signals, including many known hypha-associated genes. Surprisingly, overlap among those 72 genes with 2 recent consensus definitions of hypha-associated genes was limited to only 16 genes. Fourth, which regulators govern hemin-induced filamentation? A mutant survey indicated that the response depends upon filamentation regulators Efg1, Brg1, and Rim101, but not upon heme acquisition regulator Hap1 or its target genes HMX1, RBT5, PGA10, PGA7, and CSA2. These findings argue that hemin induces hypha formation independently of its utilization.
Collapse
Affiliation(s)
- Liping Xiong
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Katharina Goerlich
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Aaron P Mitchell
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
61
|
Basrani ST, Gavandi TC, Patil SB, Kadam NS, Yadav DV, Chougule SA, Karuppayil SM, Jadhav AK. Hydroxychloroquine an Antimalarial Drug, Exhibits Potent Antifungal Efficacy Against Candida albicans Through Multitargeting. J Microbiol 2024; 62:381-391. [PMID: 38587590 DOI: 10.1007/s12275-024-00111-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 04/09/2024]
Abstract
Candida albicans is the primary etiological agent associated with candidiasis in humans. Unrestricted growth of C. albicans can progress to systemic infections in the worst situation. This study investigates the antifungal activity of Hydroxychloroquine (HCQ) and mode of action against C. albicans. HCQ inhibited the planktonic growth and yeast to hyphal form morphogenesis of C. albicans significantly at 0.5 mg/ml concentration. The minimum inhibitory concentrations (MIC50) of HCQ for C. albicans adhesion and biofilm formation on the polystyrene surface was at 2 mg/ml and 4 mg/ml respectively. Various methods, such as scanning electron microscopy, exploration of the ergosterol biosynthesis pathway, cell cycle analysis, and assessment of S oxygen species (ROS) generation, were employed to investigate HCQ exerting its antifungal effects. HCQ was observed to reduce ergosterol levels in the cell membranes of C. albicans in a dose-dependent manner. Furthermore, HCQ treatment caused a substantial arrest of the C. albicans cell cycle at the G0/G1 phase, which impeded normal cell growth. Gene expression analysis revealed upregulation of SOD2, SOD1, and CAT1 genes after HCQ treatment, while genes like HWP1, RAS1, TEC1, and CDC 35 were downregulated. The study also assessed the in vivo efficacy of HCQ in a mice model, revealing a reduction in the pathogenicity of C. albicans after HCQ treatment. These results indicate that HCQ holds for the development of novel antifungal therapies.
Collapse
Affiliation(s)
- Sargun Tushar Basrani
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
| | - Tanjila Chandsaheb Gavandi
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
| | - Shivani Balasaheb Patil
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
| | - Nandkumar Subhash Kadam
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
- iSERA Biological Pvt Ltd., MIDC Shirala, Dist., Sangli, Maharashtra, 41540, India
| | - Dhairyasheel Vasantrao Yadav
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
- iSERA Biological Pvt Ltd., MIDC Shirala, Dist., Sangli, Maharashtra, 41540, India
| | - Sayali Ashok Chougule
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
| | - Sankunny Mohan Karuppayil
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India
| | - Ashwini Khanderao Jadhav
- Department of Stem Cell and Regenerative Medicine and Medical Biotechnology, Centre for Interdisciplinary Research, DY Patil Education Society (Deemed to be University), Kadamwadi, Kolhapur, Maharashtra, 416003, India.
| |
Collapse
|
62
|
Ahmady L, Gothwal M, Mukkoli MM, Bari VK. Antifungal drug resistance in Candida: a special emphasis on amphotericin B. APMIS 2024; 132:291-316. [PMID: 38465406 DOI: 10.1111/apm.13389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
Invasive fungal infections in humans caused by several Candida species, increased considerably in immunocompromised or critically ill patients, resulting in substantial morbidity and mortality. Candida albicans is the most prevalent species, although the frequency of these organisms varies greatly according to geographic region. Infections with C. albicans and non-albicans Candida species have become more common, especially in the past 20 years, as a result of aging, immunosuppressive medication use, endocrine disorders, malnourishment, extended use of medical equipment, and an increase in immunogenic diseases. Despite C. albicans being the species most frequently associated with human infections, C. glabrata, C. parapsilosis, C. tropicalis, and C. krusei also have been identified. Several antifungal drugs with different modes of action are approved for use in clinical settings to treat fungal infections. However, due to the common eukaryotic structure of humans and fungi, only a limited number of antifungal drugs are available for therapeutic use. Furthermore, drug resistance in Candida species has emerged as a result of the growing use of currently available antifungal drugs against fungal infections. Amphotericin B (AmB), a polyene class of antifungal drugs, is mainly used for the treatment of serious systemic fungal infections. AmB interacts with fungal plasma membrane ergosterol, triggering cellular ion leakage via pore formation, or extracting the ergosterol from the plasma membrane inducing cellular death. AmB resistance is primarily caused by changes in the content or structure of ergosterol. This review summarizes the antifungal drug resistance exhibited by Candida species, with a special focus on AmB.
Collapse
Affiliation(s)
- Lailema Ahmady
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | - Manisha Gothwal
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | | | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
63
|
Grayton QE, Conlon IL, Broberg CA, Schoenfisch MH. Impact of Nitric Oxide-Release Kinetics on Antifungal Activity. J Fungi (Basel) 2024; 10:308. [PMID: 38786663 PMCID: PMC11121837 DOI: 10.3390/jof10050308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Pathogenic fungi are an increasing health threat due to the rise in drug resistance. The limited number of antifungals currently available and growing incidence of multi-drug-resistant fungi has caused rising healthcare costs and a decreased quality of life for patients with fungal infections. Nitric oxide (NO) has previously been shown to act as an antimicrobial agent, albeit with a limited understanding of the effects of the NO-release kinetics against pathogenic fungi. Herein, the antifungal effects of four nitric oxide-releasing small molecules were studied against the pathogenic fungi Candida albicans, Candida auris, Cryptococcus neoformans, and Aspergillus fumigatus, to demonstrate the broad-spectrum antifungal activity of NO. A bolus dose of NO was found to eradicate fungi after 24 h, where nitric oxide donors with shorter half-lives achieved antifungal activity at lower concentrations and thus had wider selectivity indexes. Each NO donor was found to cause a severe surface destruction of fungi, and all NO donors exhibited compatibility with currently prescribed antifungals against several different fungi species.
Collapse
Affiliation(s)
- Quincy E. Grayton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
| | - Ivie L. Conlon
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
| | - Christopher A. Broberg
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
64
|
Hang S, Lu H, Jiang Y. Marine-Derived Metabolites Act as Promising Antifungal Agents. Mar Drugs 2024; 22:180. [PMID: 38667797 PMCID: PMC11051449 DOI: 10.3390/md22040180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
The incidence of invasive fungal diseases (IFDs) is on the rise globally, particularly among immunocompromised patients, leading to significant morbidity and mortality. Current clinical antifungal agents, such as polyenes, azoles, and echinocandins, face increasing resistance from pathogenic fungi. Therefore, there is a pressing need for the development of novel antifungal drugs. Marine-derived secondary metabolites represent valuable resources that are characterized by varied chemical structures and pharmacological activities. While numerous compounds exhibiting promising antifungal activity have been identified, a comprehensive review elucidating their specific underlying mechanisms remains lacking. In this review, we have compiled a summary of antifungal compounds derived from marine organisms, highlighting their diverse mechanisms of action targeting various fungal cellular components, including the cell wall, cell membrane, mitochondria, chromosomes, drug efflux pumps, and several biological processes, including vesicular trafficking and the growth of hyphae and biofilms. This review is helpful for the subsequent development of antifungal drugs due to its summary of the antifungal mechanisms of secondary metabolites from marine organisms.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 200092 Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 200092 Shanghai, China
| |
Collapse
|
65
|
An JX, Zhang BQ, Liang HJ, Zhang ZJ, Liu YQ, Zhang SY. Antifungal Activity and Putative Mechanism of HWY-289, a Semisynthetic Protoberberine Derivative, against Botrytis cinerea. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7716-7726. [PMID: 38536397 DOI: 10.1021/acs.jafc.3c08858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
The emergence of resistant pathogens has increased the demand for alternative fungicides. The use of natural products as chemical scaffolds is a potential method for developing fungicides. HWY-289, a semisynthetic protoberberine derivative, demonstrated broad-spectrum and potent activities against phytopathogenic fungi, particularly Botrytis cinerea (with EC50 values of 1.34 μg/mL). SEM and TEM imaging indicated that HWY-289 altered the morphology of the mycelium and the internal structure of cells. Transcriptomics revealed that it could break down cellular walls through amino acid sugar and nucleotide sugar metabolism. In addition, it substantially decreased chitinase activity and chitin synthase gene (BcCHSV) expression by 53.03 and 82.18% at 1.5 μg/mL, respectively. Moreover, this impacted the permeability and integrity of cell membranes. Finally, HWY-289 also hindered energy metabolism, resulting in a significant reduction of ATP content, ATPase activities, and key enzyme activities in the TCA cycle. Therefore, HWY-289 may be a potential candidate for the development of plant fungicides.
Collapse
Affiliation(s)
- Jun-Xia An
- School of Pharmacy, Lanzhou University, Lanzhou 730000, People's Republic of China
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Bao-Qi Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Hong-Jie Liang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Zhi-Jun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, People's Republic of China
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou 313000, China
| | - Shao-Yong Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou 313000, China
| |
Collapse
|
66
|
Sumlu E, Aydin M, Korucu EN, Alyar S, Nsangou AM. Artemisinin May Disrupt Hyphae Formation by Suppressing Biofilm-Related Genes of Candida albicans: In Vitro and In Silico Approaches. Antibiotics (Basel) 2024; 13:310. [PMID: 38666986 PMCID: PMC11047306 DOI: 10.3390/antibiotics13040310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/29/2024] Open
Abstract
This study aimed to assess the antifungal and antibiofilm efficacy of artemisinin against Candida (C.) species, analyze its impact on gene expression levels within C. albicans biofilms, and investigate the molecular interactions through molecular docking. The antifungal efficacy of artemisinin on a variety of Candida species, including fluconazole-resistant and -susceptible species, was evaluated by the microdilution method. The effect of artemisinin on C. albicans biofilm formation was investigated by MTT and FESEM. The mRNA expression of the genes related to biofilm was analyzed by qRT-PCR. In addition, molecular docking analysis was used to understand the interaction between artemisinin and C. albicans at the molecular level with RAS1-cAMP-EFG1 and EFG1-regulated genes. Artemisinin showed higher sensitivity against non-albicans Candida strains. Furthermore, artemisinin was strongly inhibitory against C. albicans biofilms at 640 µg/mL. Artemisinin downregulated adhesion-related genes ALS3, HWP1, and ECE1, hyphal development genes UME6 and HGC1, and hyphal CAMP-dependent protein kinase regulators CYR1, RAS1, and EFG1. Furthermore, molecular docking analysis revealed that artemisinin and EFG1 had the highest affinity, followed by UME6. FESEM analysis showed that the fluconazole- and artemisinin-treated groups exhibited a reduced hyphal network, unusual surface bulges, and the formation of pores on the cell surfaces. Our study suggests that artemisinin may have antifungal potential and showed a remarkable antibiofilm activity by significantly suppressing adhesion and hyphal development through interaction with key proteins involved in biofilm formation, such as EFG1.
Collapse
Affiliation(s)
- Esra Sumlu
- Department of Medical Pharmacology, Faculty of Medicine, KTO Karatay University, 42020 Konya, Turkey;
| | - Merve Aydin
- Department of Medical Microbiology, Faculty of Medicine, KTO Karatay University, 42020 Konya, Turkey
| | - Emine Nedime Korucu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, 42090 Konya, Turkey;
| | - Saliha Alyar
- Department of Chemistry, Faculty of Science, Karatekin University, 18100 Çankırı, Turkey;
| | - Ahmed Moustapha Nsangou
- Department of Medical Microbiology, Faculty of Medicine, Selçuk University, 42130 Konya, Turkey;
| |
Collapse
|
67
|
Lass-Flörl C, Kanj SS, Govender NP, Thompson GR, Ostrosky-Zeichner L, Govrins MA. Invasive candidiasis. Nat Rev Dis Primers 2024; 10:20. [PMID: 38514673 DOI: 10.1038/s41572-024-00503-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/15/2024] [Indexed: 03/23/2024]
Abstract
Invasive candidiasis is an important fungal disease caused by Candida albicans and, increasingly, non-albicans Candida pathogens. Invasive Candida infections originate most frequently from endogenous human reservoirs and are triggered by impaired host defences. Signs and symptoms of invasive candidiasis are non-specific; candidaemia is the most diagnosed manifestation, with disseminated candidiasis affecting single or multiple organs. Diagnosis poses many challenges, and conventional culture techniques are frequently supplemented by non-culture-based assays. The attributable mortality from candidaemia and disseminated infections is ~30%. Fluconazole resistance is a concern for Nakaseomyces glabratus, Candida parapsilosis, and Candida auris and less so in Candida tropicalis infection; acquired echinocandin resistance remains uncommon. The epidemiology of invasive candidiasis varies in different geographical areas and within various patient populations. Risk factors include intensive care unit stay, central venous catheter use, broad-spectrum antibiotics use, abdominal surgery and immune suppression. Early antifungal treatment and central venous catheter removal form the cornerstones to decrease mortality. The landscape of novel therapeutics is growing; however, the application of new drugs requires careful selection of eligible patients as the spectrum of activity is limited to a few fungal species. Unanswered questions and knowledge gaps define future research priorities and a personalized approach to diagnosis and treatment of invasive candidiasis is of paramount importance.
Collapse
Affiliation(s)
- Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, ECMM Excellence Centres of Medical Mycology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Souha S Kanj
- Infectious Diseases Division, and Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nelesh P Govender
- Faculty of Health Sciences, University of the Witwatersrand and National Institute for Communicable Diseases, Johannesburg, South Africa
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - George R Thompson
- UC Davis Health Medical Center, Division of Infectious Diseases, Sacramento, CA, USA
| | | | - Miriam Alisa Govrins
- Institute of Hygiene and Medical Microbiology, ECMM Excellence Centres of Medical Mycology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
68
|
Pattini VC, Polaquini CR, Lemes TH, Brizzotti-Mazuchi NS, Sardi JDCO, Paziani MH, Kress MRVZ, de Almeida MTG, Regasini LO. Antifungal activity of 3,3'-dimethoxycurcumin (DMC) against dermatophytes and Candida species. Lett Appl Microbiol 2024; 77:ovae019. [PMID: 38499446 DOI: 10.1093/lambio/ovae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/24/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Dermatomycosis is an infection with global impacts caused especially by dermatophytes and Candida species. Current antifungal therapies involve drugs that face fungal resistance barriers. This clinical context emphasizes the need to discover new antifungal agents. Herein, the antifungal potential of 10 curcumin analogs was evaluated against four Candida and four dermatophyte species. The most active compound, 3,3'-dimethoxycurcumin, exhibited minimum inhibitory concentration values ranging from 1.9‒62.5 to 15.6‒62.5 µg ml-1 against dermatophytes and Candida species, respectively. According to the checkerboard method, the association between DMC and terbinafine demonstrated a synergistic effect against Trichophyton mentagrophytes and Epidermophyton floccosum. Ergosterol binding test indicated DMC forms a complex with ergosterol of Candida albicans, C. krusei, and C. tropicalis. However, results from the sorbitol protection assay indicated that DMC had no effect on the cell walls of Candida species. The in vivo toxicity, using Galleria mellonella larvae, indicated no toxic effect of DMC. Altogether, curcumin analog DMC was a promising antifungal agent with a promising ability to act against Candida and dermatophyte species.
Collapse
Affiliation(s)
- Veridianna Camilo Pattini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Carlos Roberto Polaquini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Thiago Henrique Lemes
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| | | | | | - Mário Henrique Paziani
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 05508-000, Brazil
| | - Marcia Regina von Zeska Kress
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 05508-000, Brazil
| | | | - Luis Octávio Regasini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| |
Collapse
|
69
|
Das S, Konwar BK. Inhibiting pathogenicity of vaginal Candida albicans by lactic acid bacteria and MS analysis of their extracellular compounds. APMIS 2024; 132:161-186. [PMID: 38168754 DOI: 10.1111/apm.13365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
Maintaining healthy vaginal microflora post-puberty is critical. In this study we explore the potential of vaginal lactic acid bacteria (LAB) and their extracellular metabolites against the pathogenicity of Candida albicans. The probiotic culture free supernatant (PCFS) from Lactobacillus crispatus, L. gasseri, and L. vaginalis exhibit an inhibitory effect on budding, hyphae, and biofilm formation of C. albicans. LGPCFS manifested the best potential among the LAB PCFS, inhibiting budding for 24 h and restricting hyphae formation post-stimulation. LGPCFS also pre-eminently inhibited biofilm formation. Furthermore, L. gasseri itself grew under RPMI 1640 stimulation suppressing the biofilm formation of C. albicans. The PCFS from the LAB downregulated the hyphal genes of C. albicans, inhibiting the yeast transformation to fungi. Hyphal cell wall proteins HWP1, ALS3, ECE1, and HYR1 and transcription factors BCR1 and CPH1 were downregulated by the metabolites from LAB. Finally, the extracellular metabolome of the LAB was studied by LC-MS/MS analysis. L.gasseri produced the highest antifungal compounds and antibiotics, supporting its best activity against C. albicans. Vaginal LAB and their extracellular metabolites perpetuate C. albicans at an avirulent state. The metabolites produced by these LAB in vitro have been identified, and can be further exploited as a preventive measure against vaginal candidiasis.
Collapse
Affiliation(s)
- Shreaya Das
- Department of MBBT, Tezpur University, Napaam, Assam, India
| | | |
Collapse
|
70
|
Silva VBD, Almeida-Bezerra JW, Novais MHG, Farias NS, Coelho JJ, Ribeiro PRV, Canuto KM, Coutinho HDM, Morais-Braga MFB, Oliveira AFMD. Chemical composition, antifungal, and anti-virulence action of the stem bark of Hancornia speciosa Gomes (Apocynaceae) against Candida spp. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117506. [PMID: 38012976 DOI: 10.1016/j.jep.2023.117506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hancornia speciosa Gomes is a fruit and medicinal species used for treating infectious diseases of the genitourinary system. However, its mechanism of action against microbes is still not fully understood. Infections in the genitourinary system caused by Candida spp. are associated with its fungal resistance and pathogenicity. New plant-derived compounds are an alternative to fight these Candida infections. AIM OF THE STUDY The objective of this study was to evaluate the anti-Candida effects of extracts of the stem bark of H. speciosa. This research investigated the chemical composition of sulfuric ether (EEHS) and methanolic (MEHS) extracts, their drug-modifying action on fluconazole, and their anti-virulence action on the morphological transition of Candida species. MATERIALS AND METHODS The extracts (EEHS and MEHS) of the stem bark of H. speciosa were chemically characterized via qualitative phytochemical screening and by liquid chromatography coupled with mass spectrometry (UPLC-MS-ESI-QTOF). The extracts were evaluated regarding their antifungal effects and fluconazole-modifying activity against Candida albicans, Candida krusei, and Candida tropicalis using the broth microdilution method. Additionally, the study evaluated the inhibition of fungal virulence in Candida species through morphological transition assays. RESULTS The phytochemical screening revealed the presence of anthocyanidins, anthocyanins, aurones, catechins, chalcones, flavones, flavonols, flavanones, leucoanthocyanidins, tannins (condensed and pyrogallic), and xanthones in both extracts of the stem bark of H. speciosa. The UPLC-MS-ESI-QTOF analysis identified the same compounds in both extracts, predominating phenolic compounds. Some compounds were first time recorded in this species: gluconic acid, cinchonain IIb, cinchonain Ib isomer, and lariciresinol hexoside isomers. Most of the intrinsic antifungal activity was observed for the MEHS against C. krusei (IC50: 58.41 μg/mL). At subinhibitory concentrations (MC/8), the EEHS enhanced the action of fluconazole against all Candida strains. The MEHS exhibited greater efficacy than fluconazole inhibiting C. krusei growth. The EEHS completely inhibited hyphae appearance and reduced pseudohyphae formation in C. albicans. CONCLUSION The stem bark of H. speciosa is a rich source of bioactive compounds, especially phenolic. Phenolic compounds can have important roles in fighting infectious diseases of the genitourinary system, such as candidiasis. The extracts of H. speciosa improved the action of the drug fluconazole against Candida species, inhibited hyphae appearance, and reduced pseudohyphae formation. The results of this study can support the development of new therapeutics against resistant strains of Candida.
Collapse
Affiliation(s)
- Viviane Bezerra da Silva
- Department of Botany, Universidade Federal de Pernambuco - UFPE, Rua Professor Moraes Rego, s/n, Recife, Pernambuco, 50.670-901, Brazil.
| | - José Weverton Almeida-Bezerra
- Department of Botany, Universidade Federal de Pernambuco - UFPE, Rua Professor Moraes Rego, s/n, Recife, Pernambuco, 50.670-901, Brazil
| | - Maria Hellena Garcia Novais
- Department of Biological Sciences, Universidade Regional do Cariri - URCA, Rua Cel. Antônio Luiz, 1161, Crato, Ceará, 63.105-000, Brazil
| | - Naiza Saraiva Farias
- Department of Biological Sciences, Universidade Regional do Cariri - URCA, Rua Cel. Antônio Luiz, 1161, Crato, Ceará, 63.105-000, Brazil
| | - Janerson José Coelho
- Animal Science Department, Universidade Estadual do Maranhão - UEMA, São Luís, Maranhão, Brazil
| | - Paulo Riceli Vasconcelos Ribeiro
- Multi-User Natural Products Chemistry Laboratory - LMQPN, Embrapa Agroindustria Tropical, Rua Dra. Sara Mesquita, 2270, Fortaleza, Ceará, 60511-110, Brazil
| | - Kirley Marques Canuto
- Multi-User Natural Products Chemistry Laboratory - LMQPN, Embrapa Agroindustria Tropical, Rua Dra. Sara Mesquita, 2270, Fortaleza, Ceará, 60511-110, Brazil
| | - Henrique Douglas Melo Coutinho
- Department of Biological Chemistry, Universidade Regional do Cariri - URCA, Rua Cel. Antônio Luiz, 1161, Crato, Ceará, 63.105-000, Brazil
| | | | | |
Collapse
|
71
|
Wang J, Shen J, Chen D, Liao B, Chen X, Zong Y, Wei Y, Shi Y, Liu Y, Gou L, Zhou X, Cheng L, Ren B. Secretory IgA reduced the ergosterol contents of Candida albicans to repress its hyphal growth and virulence. Appl Microbiol Biotechnol 2024; 108:244. [PMID: 38421461 PMCID: PMC10904422 DOI: 10.1007/s00253-024-13063-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
Candida albicans, one of the most prevalent conditional pathogenic fungi, can cause local superficial infections and lethal systemic infections, especially in the immunocompromised population. Secretory immunoglobulin A (sIgA) is an important immune protein regulating the pathogenicity of C. albicans. However, the actions and mechanisms that sIgA exerts directly against C. albicans are still unclear. Here, we investigated that sIgA directs against C. albicans hyphal growth and virulence to oral epithelial cells. Our results indicated that sIgA significantly inhibited C. albicans hyphal growth, adhesion, and damage to oral epithelial cells compared with IgG. According to the transcriptome and RT-PCR analysis, sIgA significantly affected the ergosterol biosynthesis pathway. Furthermore, sIgA significantly reduced the ergosterol levels, while the addition of exogenous ergosterol restored C. albicans hyphal growth and adhesion to oral epithelial cells, indicating that sIgA suppressed the growth of hyphae and the pathogenicity of C. albicans by reducing its ergosterol levels. By employing the key genes mutants (erg11Δ/Δ, erg3Δ/Δ, and erg3Δ/Δ erg11Δ/Δ) from the ergosterol pathway, sIgA lost the hyphal inhibition on these mutants, while sIgA also reduced the inhibitory effects of erg11Δ/Δ and erg3Δ/Δ and lost the inhibition of erg3Δ/Δ erg11Δ/Δ on the adhesion to oral epithelial cells, further proving the hyphal repression of sIgA through the ergosterol pathway. We demonstrated for the first time that sIgA inhibited C. albicans hyphal development and virulence by affecting ergosterol biosynthesis and suggest that ergosterol is a crucial regulator of C. albicans-host cell interactions. KEY POINTS: • sIgA repressed C. albicans hyphal growth • sIgA inhibited C. albicans virulence to host cells • sIgA affected C. albicans hyphae and virulence by reducing its ergosterol levels.
Collapse
Affiliation(s)
- Jiannan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xi Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yawen Zong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
72
|
Yang J, Ma Y, Li B, Xi Z, Zhang L, Wang Y, Feng W. Roles of Nucleolar Factor RCL1 in Itraconazole Resistance of Clinical Candida albicans Under Different Stress Conditions. Infect Drug Resist 2024; 17:769-777. [PMID: 38433785 PMCID: PMC10908289 DOI: 10.2147/idr.s431024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Purpose RNA terminal phosphate cyclase like 1 (RCL1) undergoes overexpression during the immune response of Candida albicans following drug treatment. This study aims to investigate the expression levels of RCL1 in C. albicans under various stress conditions. Methods Fifteen itraconazole (ITR)-resistant strains of clinical C. albicans, and one standard strain were employed for RCL1 sequencing, and mutations in RCL1 were analyzed. Subsequently, 14 out of the 15 ITR-resistant clinical strains and 14 clinical strains sensitive to ITR, fluconazole (FCA) as well as voriconazole (VRC) were cultured under diverse conditions. The expression of RCL1 ITR-resistant and sensitive C. albicans was then assessed using real-time quantitative PCR (RT-qPCR) assays. Results Compared to the standard strain, three missense mutations (C6A, G10A, and A11T) were identified in the RCL1 gene of ITR-resistant C. albicans through successful forward sequencing. Additionally, using successful reverse sequencing, one synonymous mutation (C1T) and four missense mutations (C1T, A3T, A7G, and T8G) were found in the RCL1 gene of ITR-resistant C. albicans. RCL1 expression was significantly higher in ITR-resistant C. albicans than in sensitive strains under standard conditions (37°C, 0.03% CO2, pH 4.0). Low temperature (25°C) increased RCL1 expression in sensitive C. albicans while decreasing it in ITR-resistant strains. Elevated CO2 concentrations (5% CO2) had a negligible effect on RCL1 expression in sensitive C. albicans, but effectively reduced RCL1 level in ITR-resistant strains. Furthermore, a medium with a pH of 7 decreased the expression of RCL1 in both resistant and sensitive C. albicans. Conclusion This study demonstrated that RCL1 mutations in ITR-resistant C. albicans, and variations in culture conditions significantly influence RCL1 expression in both ITR-resistant and sensitive C. albicans, thereby inducing alterations in the dimorphism of C. albicans.
Collapse
Affiliation(s)
- Jing Yang
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Yan Ma
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Bo Li
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Zhiqin Xi
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Li Zhang
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Yuxi Wang
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Wenli Feng
- Department of Dermatovenereology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| |
Collapse
|
73
|
Arbon D, Mach J, Čadková A, Sipkova A, Stursa J, Klanicová K, Machado M, Ganter M, Levytska V, Sojka D, Truksa J, Werner L, Sutak R. Chelation of Mitochondrial Iron as an Antiparasitic Strategy. ACS Infect Dis 2024; 10:676-687. [PMID: 38287902 PMCID: PMC10862539 DOI: 10.1021/acsinfecdis.3c00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
Iron, as an essential micronutrient, plays a crucial role in host-pathogen interactions. In order to limit the growth of the pathogen, a common strategy of innate immunity includes withdrawing available iron to interfere with the cellular processes of the microorganism. Against that, unicellular parasites have developed powerful strategies to scavenge iron, despite the effort of the host. Iron-sequestering compounds, such as the approved and potent chelator deferoxamine (DFO), are considered a viable option for therapeutic intervention. Since iron is heavily utilized in the mitochondrion, targeting iron chelators in this organelle could constitute an effective therapeutic strategy. This work presents mitochondrially targeted DFO, mitoDFO, as a candidate against a range of unicellular parasites with promising in vitro efficiency. Intracellular Leishmania infection can be cleared by this compound, and experimentation with Trypanosoma brucei 427 elucidates its possible mode of action. The compound not only affects iron homeostasis but also alters the physiochemical properties of the inner mitochondrial membrane, resulting in a loss of function. Furthermore, investigating the virulence factors of pathogenic yeasts confirms that mitoDFO is a viable candidate for therapeutic intervention against a wide spectrum of microbe-associated diseases.
Collapse
Affiliation(s)
- Dominik Arbon
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Jan Mach
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Aneta Čadková
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Anna Sipkova
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Jan Stursa
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
- Laboratory
of Clinical Pathophysiology, Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech
Republic
| | - Kristýna Klanicová
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Prague 128 00, Czech Republic
| | - Marta Machado
- Graduate
Program in Areas of Basic and Applied Biology, Instituto de Ciências
Biomédicas Abel Salazar, Universidade
do Porto, Porto 4050-313, Portugal
- Centre for
Infectious Diseases, Parasitology, Heidelberg
University Hospital, Heidelberg 69120, Germany
| | - Markus Ganter
- Centre for
Infectious Diseases, Parasitology, Heidelberg
University Hospital, Heidelberg 69120, Germany
| | - Viktoriya Levytska
- Institute
of Parasitology, Biology Centre, Academy
of Sciences of the Czech Republic, Branišovská 1160/31, České Budějovice 37005, Czech Republic
| | - Daniel Sojka
- Institute
of Parasitology, Biology Centre, Academy
of Sciences of the Czech Republic, Branišovská 1160/31, České Budějovice 37005, Czech Republic
| | - Jaroslav Truksa
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
| | - Lukáš Werner
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
- Laboratory
of Clinical Pathophysiology, Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech
Republic
| | - Robert Sutak
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| |
Collapse
|
74
|
Dong Y, Du J, Deng Y, Cheng M, Shi Z, Zhu H, Sun H, Yu Q, Li M. Reduction of histone proteins dosages increases CFW sensitivity and attenuates virulence of Candida albicans. Microbiol Res 2024; 279:127552. [PMID: 38000336 DOI: 10.1016/j.micres.2023.127552] [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: 08/31/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023]
Abstract
Histone proteins are important components of nucleosomes, which play an important role in regulating the accessibility of DNA and the function of genomes. However, the effect of histone proteins dosages on physiological processes is not clear in the human fungal pathogen Candida albicans. In this study, we found that the deletion of the histone protein H3 coding gene HHT21 and the histone protein H4 coding gene HHF1 resulted in a significant decrease in the expression dosage of the histone proteins H3 and H4, which had a significant impact on the localization of the histone protein H2A and plasmid maintenance. Stress sensitivity experiments showed that the mutants hht21Δ/Δ, hhf1Δ/Δ and hht21Δ/Δhhf1Δ/Δ were more sensitive to cell wall stress induced by Calcofluor White (CFW) than the wild-type strain. Further studies showed that the decrease in the dosage of the histone proteins H3 and H4 led to the change of cell wall components, increased chitin contents, and down-regulated expression of the SAP9, KAR2, and CRH11 genes involved in the cell wall integrity (CWI) pathway. Overexpression of SAP9 could rescue the sensitivity of the mutants to CFW. Moreover, the decrease in the histone protein s dosages affected the FAD-catalyzed oxidation of Ero1 protein, resulting in the obstruction of protein folding in the ER, and thus reduced resistance to CFW. It was also found that CFW induced a large amount of ROS accumulation in the mutants, and the addition of ROS scavengers could restore the growth of the mutants under CFW treatment. In addition, the reduction of the histone proteins dosages greatly weakened systemic infection and kidney fungal burden in mice, and hyphal development was significantly impaired in the mutants under macrophage treatment, indicating that the histone proteins dosages is very important for the virulence of C. albicans. This study revealed that histone proteins dosages play a key role in the cell wall stress response and pathogenicity in C. albicans.
Collapse
Affiliation(s)
- Yixuan Dong
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jiawen Du
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ying Deng
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mengjuan Cheng
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhishang Shi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hao Sun
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
75
|
Kovács-Simon A, Fones HN. Use of chitin:DNA ratio to assess growth form in fungal cells. BMC Biol 2024; 22:10. [PMID: 38233847 PMCID: PMC10795418 DOI: 10.1186/s12915-024-01815-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Dimorphism, the ability to switch between a 'yeast-like' and a hyphal growth form, is an important feature of certain fungi, including important plant and human pathogens. The switch to hyphal growth is often associated with virulence, pathogenicity, biofilm formation and stress resistance. Thus, the ability to accurately and efficiently measure fungal growth form is key to research into these fungi, especially for discovery of potential drug targets. To date, fungal growth form has been assessed microscopically, a process that is both labour intensive and costly. RESULTS Here, we unite quantification of the chitin in fungal cell walls and the DNA in nuclei to produce a methodology that allows fungal cell shape to be estimated by calculation of the ratio between cell wall quantity and number of nuclei present in a sample of fungus or infected host tissue. Using the wheat pathogen Zymoseptoria tritici as a test case, with confirmation in the distantly related Fusarium oxysporum, we demonstrate a close, linear relationship between the chitin:DNA ratio and the average polarity index (length/width) of fungal cells. We show the utility of the method for estimating growth form in infected wheat leaves, differentiating between the timing of germination in two different Z. tritici isolates using this ratio. We also show that the method is robust to the occurrence of thick-walled chlamydospores, which show a chitin:DNA ratio that is distinct from either 'yeast-like' blastospores or hyphae. CONCLUSIONS The chitin:DNA ratio provides a simple methodology for determining fungal growth form in bulk tissue samples, reducing the need for labour-intensive microscopic studies requiring specific staining or GFP-tags to visualise the fungus within host tissues. It is applicable to a range of dimorphic fungi under various experimental conditions.
Collapse
|
76
|
Janeczko M, Kochanowicz E. Biochanin A Inhibits the Growth and Biofilm of Candida Species. Pharmaceuticals (Basel) 2024; 17:89. [PMID: 38256922 PMCID: PMC10818846 DOI: 10.3390/ph17010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The aim of this study was to investigate the antifungal activity of biochanin A (BCA) against planktonic growth and biofilms of six Candida species, including C. albicans, C. parapsilosis, C. glabrata, C. tropicalis, C. auris, and C. krusei. We applied various assays that determined (a) the antimicrobial effect on growth of Candida species, (b) the effect on formation of hyphae and biofilm, (c) the effect on the expression of genes related to hyphal growth and biofilm formation, (d) the influence on cell wall structure, and (e) the effect on cell membrane integrity and permeability. Moreover, disk diffusion tests were used to investigate the effect of a combination of BCA with fluconazole to assess their possible synergistic effect on drug-resistant C. albicans, C. glabrata, and C. auris. Our results showed that the BCA MIC50 values against Candida species ranged between 125 µg/mL and 500 µg/mL, and the MIC90 values were in a concentration range from 250 µg/mL to 1000 µg/mL. The treatment with BCA inhibited adhesion of cells, cell surface hydrophobicity (CSH), and biofilm formation and reduced hyphal growth in all the analyzed Candida species. Real-time qRT-PCR revealed that BCA down-regulated the expression of biofilm-specific genes in C. albicans. Furthermore, physical destruction of C. albicans cell membranes and cell walls as a result of the treatment with BCA was observed. The combination of BCA and fluconazole did not exert synergistic effects against fluconazole-resistant Candida.
Collapse
Affiliation(s)
- Monika Janeczko
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland;
| | | |
Collapse
|
77
|
King WR, Singer J, Warman M, Wilson D, Hube B, Lager I, Patton-Vogt J. The glycerophosphocholine acyltransferase Gpc1 contributes to phosphatidylcholine biosynthesis, long-term viability, and embedded hyphal growth in Candida albicans. J Biol Chem 2024; 300:105543. [PMID: 38072057 PMCID: PMC10790099 DOI: 10.1016/j.jbc.2023.105543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023] Open
Abstract
Candida albicans is a commensal fungus, opportunistic pathogen, and the most common cause of fungal infection in humans. The biosynthesis of phosphatidylcholine (PC), a major eukaryotic glycerophospholipid, occurs through two primary pathways. In Saccharomyces cerevisiae and some plants, a third PC synthesis pathway, the PC deacylation/reacylation pathway (PC-DRP), has been characterized. PC-DRP begins with the acylation of the lipid turnover product, glycerophosphocholine (GPC), by the GPC acyltransferase, Gpc1, to form Lyso-PC. Lyso-PC is then acylated by lysolipid acyltransferase, Lpt1, to produce PC. Importantly, GPC, the substrate for Gpc1, is a ubiquitous metabolite available within the host. GPC is imported by C. albicans, and deletion of the major GPC transporter, Git3, leads to decreased virulence in a murine model. Here we report that GPC can be directly acylated in C. albicans by the protein product of orf19.988, a homolog of ScGpc1. Through lipidomic studies, we show loss of Gpc1 leads to a decrease in PC levels. This decrease occurs in the absence of exogenous GPC, indicating that the impact on PC levels may be greater in the human host where GPC is available. A gpc1Δ/Δ strain exhibits several sensitivities to antifungals that target lipid metabolism. Furthermore, loss of Gpc1 results in both a hyphal growth defect in embedded conditions and a decrease in long-term cell viability. These results demonstrate for the first time the importance of Gpc1 and this alternative PC biosynthesis route (PC-DRP) to the physiology of a pathogenic fungus.
Collapse
Affiliation(s)
- William R King
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Justin Singer
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Mitchell Warman
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Duncan Wilson
- Department of Biosciences, University of Exeter, Exeter, England
| | - Bernard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Products and Infection Biology Hans Knöll Institute, Jena, Germany
| | - Ida Lager
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Jana Patton-Vogt
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
78
|
Li X, Kong B, Sun Y, Sun F, Yang H, Zheng S. Synergistic potential of teriflunomide with fluconazole against resistant Candida albicans in vitro and in vivo. Front Cell Infect Microbiol 2023; 13:1282320. [PMID: 38169891 PMCID: PMC10758495 DOI: 10.3389/fcimb.2023.1282320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction Candida albicans is the primary cause of systemic candidiasis, which is involved in high morbidity and mortality. Drug resistance exacerbates these problems. In addition, there are limited antifungal drugs available. In order to solve these problems, combination therapy has aroused great interest. Teriflunomide is an immunosuppressant. In the present work, we aimed to identify whether teriflunomide can reverse the resistance of Candida albicans in the presence of sub-inhibitory concentrations of fluconazole in vitro and in vivo. Methods Seven Candida albicans isolates were used in this study. Susceptibility of Candida albicans in vitro to the drugs was determined using a checkerboard microdilution assay in accordance with the recommendations of the Clinical and Laboratory Standards Institute. The effects of drugs on biofilm biomass of Candida albicans were determined by crystal violet staining. The development ability of Candida albicans hyphae was performed using a modified broth microdilution method. Galleria mellonella was used for testing the in vivo efficacy of the combination therapies. Results We found that the combination of teriflunomide (64 µg/mL) and fluconazole (0.5-1 µg/mL) has a significant synergistic effect in all resistant Candida albicans isolates (n=4). Also, this drug combination could inhibit the immature biofilm biomass and hyphae formation of resistant Candida albicans. Galleria mellonella was used for testing the in vivo efficacy of this combination therapies. As for the Galleria mellonella larvae infected by resistant Candida albicans, teriflunomide (1.6 µg/larvae) combined with fluconazole (1.6 µg/larvae) significantly increased their survival rates, and reduced the fungal burden, as well as damage of tissue in comparison to that in the control group or drug monotherapy group. Conclusion These results expand our knowledge about the antifungal potential of teriflunomide as an adjuvant of existing antifungal drugs, and also open new perspectives in the treatment of resistant Candida albicans based on repurposing clinically available nonantifungal drugs.
Collapse
Affiliation(s)
- Xiuyun Li
- Maternal and Child Health Development Research Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bing Kong
- Department of Critical Care Medicine, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Yaqiong Sun
- Obstetrics Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Fenghua Sun
- Radiology Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Huijun Yang
- Reproductive Medicine Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Shicun Zheng
- Maternal and Child Health Development Research Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
79
|
Wu M, Xu X, Hu R, Chen Q, Chen L, Yuan Y, Li J, Zhou L, Feng S, Wang L, Chen S, Gu M. A Membrane-Targeted Photosensitizer Prevents Drug Resistance and Induces Immune Response in Treating Candidiasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207736. [PMID: 37875397 PMCID: PMC10724446 DOI: 10.1002/advs.202207736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Candida albicans (C. albicans), a ubiquitous polymorphic fungus in humans, causes different types of candidiasis, including oral candidiasis (OC) and vulvovaginal candidiasis (VVC), which are physically and mentally concerning and financially costly. Thus, developing alternative antifungals that prevent drug resistance and induce immunity to eliminate Candida biofilms is crucial. Herein, a novel membrane-targeted aggregation-induced emission (AIE) photosensitizer (PS), TBTCP-QY, is developed for highly efficient photodynamic therapy (PDT) of candidiasis. TBTCP-QY has a high molar absorption coefficient and an excellent ability to generate 1 O2 and •OH, entering the interior of biofilms due to its high permeability. Furthermore, TBTCP-QY can efficiently inhibit biofilm formation by suppressing the expression of genes related to the adhesion (ALS3, EAP1, and HWP1), invasion (SAP1 and SAP2), and drug resistance (MDR1) of C. albicans, which is also advantageous for eliminating potential fungal resistance to treat clinical infectious diseases. TBTCP-QY-mediated PDT efficiently targets OC and VVC in vivo in a mouse model, induces immune response, relieves inflammation, and accelerates the healing of mucosal defects to combat infections caused by clinically isolated fluconazole-resistant strains. Moreover, TBTCP-QY demonstrates excellent biocompatibility, suggesting its potential applications in the clinical treatment of OC and VVC.
Collapse
Affiliation(s)
- Ming‐Yu Wu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Xiaoyu Xu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Rui Hu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Qingrong Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Luojia Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Yuncong Yuan
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Jie Li
- Department of Medical Intensive Care UnitMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430070China
| | - Li Zhou
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Lianrong Wang
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Shi Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Meijia Gu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| |
Collapse
|
80
|
Lee Y, Park E, Jang B, Hwang J, Lee J, Oh ES. Antifungal Activity of Bulgarian Rose Damascena Oil against Vaginitis-Causing Opportunistic Fungi. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:5054865. [PMID: 38074845 PMCID: PMC10708955 DOI: 10.1155/2023/5054865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 10/16/2024]
Abstract
Since Bulgarian rose damascena oil is known for its anti-inflammatory, antioxidant, and antimicrobial properties, we investigated its antifungal activity against the species of Candida, which are among the most common opportunistic fungal pathogens. Our disk-diffusion assay revealed that Bulgarian rose damascena oil effectively inhibited the growth of Candida albicans along with various bacteria. The minimum inhibitory and fungicidal concentrations against Candida albicans and Candida glabrata were all 0.25%. Under our experimental conditions, Bulgarian rose damascena oil showed better inhibitory effects on Candida glabrata and Candida albicans than several popular essential oils reported to have antifungal activity other than Origanum vulgare oil. Interestingly, Bulgarian rose damascena oil showed better antifungal activity against Candida species at acidic pH and induced cell death of Candida species in the culture medium, with cell death seen in 25-35% of the cells exposed to 0.05% Bulgarian rose damascena oil. Furthermore, Bulgarian rose damascena oil inhibited the hyphal growth of Candida albicans cultured in the RPMI medium with fetal bovine serum. These findings collectively suggest that Bulgarian rose damascena oil has antifungal activity against Candida species and thus could potentially be developed in novel therapies for vaginitis-causing pathogenic fungi.
Collapse
Affiliation(s)
- Yejin Lee
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Eunhye Park
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Bohee Jang
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jisun Hwang
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinmin Lee
- Jayeonin Inc., Seoul 04995, Republic of Korea
| | - Eok-Soo Oh
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
81
|
Ke CL, Lew SQ, Hsieh Y, Chang SC, Lin CH. Convergent and divergent roles of the glucose-responsive kinase SNF4 in Candida tropicalis. Virulence 2023; 14:2175914. [PMID: 36745535 PMCID: PMC9928470 DOI: 10.1080/21505594.2023.2175914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The sucrose non-fermenting 1 (SNF1) complex is a heterotrimeric protein kinase complex that is an ortholog of the mammalian AMPK complex and is evolutionally conserved in most eukaryotes. This complex contains a catalytic subunit (Snf1), a regulatory subunit (Snf4) and a scaffolding subunit (Sip1/Sip2/Gal73) in budding yeast. Although the function of AMPK has been well studied in Saccharomyces cerevisiae and Candida albicans, the role of AMPK in Candida tropicalis has never been investigated. In this study, we focused on SNF4 in C. tropicalis as this fungus cannot produce a snf1Δ mutant. We demonstrated that C. tropicalis SNF4 shares similar roles in glucose derepression and is necessary for cell wall integrity and virulence. The expression of both SNF1 and SNF4 was significantly induced when glucose was limited. Furthermore, snf4Δ strains exhibited high sensitivity to many surface-perturbing agents because the strains contained lower levels of glucan, chitin and mannan. Interestingly, in contrast to C. albicans sak1Δ and snf4Δ, C. tropicalis snf4Δ exhibited phenotypes for cell aggregation and pseudohypha production. These data indicate that SNF4 performs convergent and divergent roles in C. tropicalis and possibly other unknown roles in the C. tropicalis SNF1-SNF4 AMPK pathway.
Collapse
Affiliation(s)
- Cai-Ling Ke
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Shi Qian Lew
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi Hsieh
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Szu-Cheng Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsuan Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan,CONTACT Ching-Hsuan Lin
| |
Collapse
|
82
|
Marcut L, Mohan AG, Corneschi I, Grosu E, Paltanea G, Avram I, Badaluta AV, Vasilievici G, Nicolae CA, Ditu LM. Improving the Hydrophobicity of Plasticized Polyvinyl Chloride for Use in an Endotracheal Tube. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7089. [PMID: 38005019 PMCID: PMC10672304 DOI: 10.3390/ma16227089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023]
Abstract
An endotracheal tube (ETT) is a greatly appreciated medical device at the global level with widespread application in the treatment of respiratory diseases, such as bronchitis and asthma, and in general anesthesia, to provide narcotic gases. Since an important quantitative request for cuffed ETTs was recorded during the COVID-19 pandemic, concerns about infection have risen. The plasticized polyvinyl chloride (PVC) material used to manufacture ETTs favors the attachment of microorganisms from the human biological environment and the migration of plasticizer from the polymer that feeds the microorganisms and promotes the growth of biofilms. This leads to developing infections, which means additional suffering, discomfort for patients, and increased hospital costs. In this work, we propose to modify the surfaces of some samples taken from commercial ETTs in order to develop their hydrophobic character using surface fluorination by a plasma treatment in SF6 discharge and magnetron sputtering physical evaporation from the PTFE target. Samples with surfaces thus modified were subsequently tested using XPS, ATR-FTIR, CA, SEM + EDAX, profilometry, density, Shore A hardness, TGA-DSC, and biological antimicrobial and biocompatibility properties. The obtained results demonstrate a successful increase in the hydrophobic character of the plasticized PVC samples and biocompatibility properties.
Collapse
Affiliation(s)
- Lavinia Marcut
- Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (L.M.); (A.G.M.)
- Intensive Care Unit, Clinical Emergency Hospital Oradea, 65 Gheorghe Doja Street, RO-410169 Oradea, Romania
| | - Aurel George Mohan
- Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (L.M.); (A.G.M.)
- Department of Neurosurgery, Clinical Emergency Hospital Oradea, 65 Gheorghe Doja Street, RO-410169 Oradea, Romania
| | - Iuliana Corneschi
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Elena Grosu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Ionela Avram
- Faculty of Biology, Botanic and Microbiology Department, University of Bucharest, 3, Aleea Portocalelor, District 5, Grădina Botanică, RO-050095 Bucharest, Romania; (I.A.); (A.V.B.); (L.M.D.)
| | - Alexandra Valentina Badaluta
- Faculty of Biology, Botanic and Microbiology Department, University of Bucharest, 3, Aleea Portocalelor, District 5, Grădina Botanică, RO-050095 Bucharest, Romania; (I.A.); (A.V.B.); (L.M.D.)
| | - Gabriel Vasilievici
- National Institute for Research & Development in Chemistry and Petrochemistry ICECHIM, 202 Splaiul Independenței, District 6, RO-060021 Bucharest, Romania; (G.V.); (C.-A.N.)
| | - Cristian-Andi Nicolae
- National Institute for Research & Development in Chemistry and Petrochemistry ICECHIM, 202 Splaiul Independenței, District 6, RO-060021 Bucharest, Romania; (G.V.); (C.-A.N.)
| | - Lia Mara Ditu
- Faculty of Biology, Botanic and Microbiology Department, University of Bucharest, 3, Aleea Portocalelor, District 5, Grădina Botanică, RO-050095 Bucharest, Romania; (I.A.); (A.V.B.); (L.M.D.)
| |
Collapse
|
83
|
Zheng D, Yue D, Shen J, Li D, Song Z, Huang Y, Yong J, Li Y. Berberine inhibits Candida albicans growth by disrupting mitochondrial function through the reduction of iron absorption. J Appl Microbiol 2023; 134:lxad276. [PMID: 37994672 DOI: 10.1093/jambio/lxad276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/04/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
AIMS This study aimed to investigate whether berberine (BBR) can inhibit the iron reduction mechanism of Candida albicans, lowering the iron uptake of the yeast and perhaps having antimicrobial effects. METHODS AND RESULTS We determined that BBR may cause extensive transcriptional remodeling in C. albicans and that iron permease Ftr1 played a crucial role in this process through eukaryotic transcriptome sequencing. Mechanistic research showed that BBR might selectively inhibit the iron reduction pathway to lower the uptake of exogenous iron ions, inhibiting C. albicans from growing and metabolizing. Subsequent research revealed that BBR caused significant mitochondrial dysfunction, which triggered the process of mitochondrial autophagy. Moreover, we discovered that C. albicans redox homeostasis, susceptibility to antifungal drugs, and hyphal growth are all impacted by the suppression of this mechanism by BBR. CONCLUSIONS The iron reduction mechanism in C. albicans is disrupted by BBR, which disrupts mitochondrial function and inhibits fungal growth. These findings highlight the potential promise of BBR in antifungal applications.
Collapse
Affiliation(s)
- Dongming Zheng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Daifan Yue
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Jinyang Shen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Dongmei Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Zhen Song
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Yifu Huang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Jiangyan Yong
- Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan 610075, China
| | - Yan Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| |
Collapse
|
84
|
Vandermeulen MD, Cullen PJ. Ecological inducers of the yeast filamentous growth pathway reveal environment-dependent roles for pathway components. mSphere 2023; 8:e0028423. [PMID: 37732804 PMCID: PMC10597418 DOI: 10.1128/msphere.00284-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Signaling modules, such as mitogen-activated protein kinase (MAPK) pathways, are evolutionarily conserved drivers of cell differentiation and stress responses. In many fungal species including pathogens, MAPK pathways control filamentous growth, where cells differentiate into an elongated cell type. The convenient model budding yeast Saccharomyces cerevisiae undergoes filamentous growth by the filamentous growth (fMAPK) pathway; however, the inducers of the pathway remain unclear, perhaps because pathway activity has been mainly studied in laboratory conditions. To address this knowledge gap, an ecological framework was used, which uncovered new fMAPK pathway inducers, including pectin, a material found in plants, and the metabolic byproduct ethanol. We also show that induction by a known inducer of the pathway, the non-preferred carbon source galactose, required galactose metabolism and induced the pathway differently than glucose limitation or other non-preferred carbon sources. By exploring fMAPK pathway function in fruit, we found that induction of the pathway led to visible digestion of fruit rind through a known target, PGU1, which encodes a pectolytic enzyme. Combinations of inducers (galactose and ethanol) stimulated the pathway to near-maximal levels, which showed dispensability of several fMAPK pathway components (e.g., mucin sensor, p21-activated kinase), but not others (e.g., adaptor, MAPKKK) and required the Ras2-protein kinase A pathway. This included a difference between the transcription factor binding partners for the pathway, as Tec1p, but not Ste12p, was partly dispensable for fMAPK pathway activity. Thus, by exploring ecologically relevant stimuli, new modes of MAPK pathway signaling were uncovered, perhaps revealing how a pathway can respond differently to specific environments. IMPORTANCE Filamentous growth is a cell differentiation response and important aspect of fungal biology. In plant and animal fungal pathogens, filamentous growth contributes to virulence. One signaling pathway that regulates filamentous growth is an evolutionarily conserved MAPK pathway. The yeast Saccharomyces cerevisiae is a convenient model to study MAPK-dependent regulation of filamentous growth, although the inducers of the pathway are not clear. Here, we exposed yeast cells to ecologically relevant compounds (e.g., plant compounds), which identified new inducers of the MAPK pathway. In combination, the inducers activated the pathway to near-maximal levels but did not cause detrimental phenotypes associated with previously identified hyperactive alleles. This context allowed us to identify conditional bypass for multiple pathway components. Thus, near-maximal induction of a MAPK pathway by ecologically relevant inducers provides a powerful tool to assess cellular signaling during a fungal differentiation response.
Collapse
Affiliation(s)
| | - Paul J. Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
85
|
Montoya C, Kurylec J, Ossa A, Orrego S. Cyclic strain of poly (methyl methacrylate) surfaces triggered the pathogenicity of Candida albicans. Acta Biomater 2023; 170:415-426. [PMID: 37625677 PMCID: PMC10705016 DOI: 10.1016/j.actbio.2023.08.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/21/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023]
Abstract
Candida albicans is an opportunistic yeast and the primary etiological factor in oral candidiasis and denture stomatitis. The pathogenesis of C. albicans could be triggered by several variables, including environmental, nutritional, and biomaterial surface cues. Specifically, biomaterial interactions are driven by different surface properties, including wettability, stiffness, and roughness. Dental biomaterials experience repetitive (cyclic) stresses from chewing and biomechanical movements. Pathogenic biofilms are formed over these biomaterial surfaces under cyclic strain. This study investigated the effect of the cyclic strain (deformation) of biomaterial surfaces on the virulence of Candida albicans. Candida biofilms were grown over Poly (methyl methacrylate) (PMMA) surfaces subjected to static (no strain) and cyclic strain with different levels (ε˜x=0.1 and 0.2%). To evaluate the biomaterial-biofilm interactions, the biofilm characteristics, yeast-to-hyphae transition, and the expression of virulent genes were measured. Results showed the biofilm biomass and metabolic activity to be significantly higher when Candida adhered to surfaces subjected to cyclic strain compared to static surfaces. Examination of the yeast-to-hyphae transition showed pseudo-hyphae cells (pathogenic) in cyclically strained biomaterial surfaces, whereas static surfaces showed spherical yeast cells (commensal). RNA sequencing was used to determine and compare the transcriptome profiles of cyclically strained and static surfaces. Genes and transcription factors associated with cell adhesion (CSH1, PGA10, and RBT5), biofilm formation (EFG1), and secretion of extracellular matrix (ECM) (CRH1, ADH5, GCA1, and GCA2) were significantly upregulated in the cyclically strained biomaterial surfaces compared to static ones. Genes and transcription factors associated with virulence (UME6 and HGC1) and the secretion of extracellular enzymes (LIP, PLB, and SAP families) were also significantly upregulated in the cyclically strained biomaterial surfaces compared to static. For the first time, this study reveals a biomaterial surface factor triggering the pathogenesis of Candida albicans, which is essential for understanding, controlling, and preventing oral infections. STATEMENT OF SIGNIFICANCE: Fungal infections produced by Candida albicans are a significant contributor to various health conditions. Candida becomes pathogenic when certain environmental conditions change, including temperature, pH, nutrients, and CO2 levels. In addition, surface properties, including wettability, stiffness, and roughness, drive the interactions between Candida and biomaterials. Clinically, Candida adheres to biomaterials that are under repetitive deformation due to body movements. In this work, we revealed that when Candida adhered to biomaterial surfaces subjected to repetitive deformation, the microorganism becomes pathogenic by increasing the formation of biofilms and the expression of virulent factors related to hyphae formation and secretion of enzymes. Findings from this work could aid the development of new strategies for treating fungal infections in medical devices or implanted biomaterials.
Collapse
Affiliation(s)
- Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Julia Kurylec
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Alex Ossa
- Production Engineering Department, School of Engineering, Universidad EAFIT, Medellín, Colombia
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States; Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
86
|
Sztafrowski D, Muraszko J, Jasiura A, Bryk P, Urbanek AK, Krasowska A. The alternating 50 Hz magnetic field depending on the hydrophobicity of the strain affects the viability, filamentation and sensitivity to drugs of Candida albicans. PLoS One 2023; 18:e0291438. [PMID: 37796949 PMCID: PMC10553255 DOI: 10.1371/journal.pone.0291438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/26/2023] [Indexed: 10/07/2023] Open
Abstract
In recent decades, Candida albicans have been the main etiological agent of life-threatening invasive infections, characterized by various mechanisms of resistance to commonly used antifungals. One of the strategies to fight Candida infections may be the use of an electromagnetic field. In this study, we examined the influence of the alternating magnetic field of 50 Hz on the cells of C. albicans. We checked the impact of the alternating magnetic field of 50 Hz on the viability, filamentation and sensitivity to fluconazole and amphotericin B of two, differing in hydrophobicity, strains of C. albicans, CAF2-1 and CAF 4-2. Our results indicate that using the alternating magnetic field of 50 Hz reduces the growth of C. albicans. Interestingly, it presents a stronger effect on the hydrophobic strain CAF4-2 than on the hydrophilic CAF2-1. The applied electromagnetic field also affects the permeabilization of the cell membrane. However, it does not inhibit the transformation from yeast to hyphal forms. AMF is more effective in combination with fluconazole rather than amphotericin B. Our findings confirm the hypothesis that the application of the alternating magnetic field of 50 Hz in antifungal therapy may arise as a new option to support the treatment of Candida infections.
Collapse
Affiliation(s)
- Dariusz Sztafrowski
- Faculty of Electrical Engineering, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Jakub Muraszko
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Adam Jasiura
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Patrycja Bryk
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Aneta K. Urbanek
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Anna Krasowska
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
87
|
Domi OKB, Darmani H. Syzygium aromaticum extracts debilitate Candida albicans by radically inhibiting its morphological plasticity and biofilm formation. JOURNAL OF HERBS, SPICES & MEDICINAL PLANTS 2023; 29:392-404. [DOI: 10.1080/10496475.2023.2196463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 01/03/2025]
Affiliation(s)
- Omar K.H. Bani Domi
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Homa Darmani
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
88
|
San Martín Andrade D, Cárdenas Amendaño CA, Solórzano Cuenca AB, Ulloa Pacheco JM, Medina-Sotomayor P. [Candida albicans: risk factor in covid-19 patients]. REVISTA CIENTÍFICA ODONTOLÓGICA 2023; 10:e132. [PMID: 38390607 PMCID: PMC10880693 DOI: 10.21142/2523-2754-1004-2022-132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/02/2022] [Indexed: 02/24/2024] Open
Abstract
The Candida Albicans fungus in our body can cause various conditions and will depend directly on the systemic condition of the host. Patients with COVID-19 who have previously presented this fungus can increase the likelihood of morbidity and mortality, since this microorganism can be located in areas that correspond to the respiratory system, generating a functional deficit. If not treated timely, it will proliferate into the blood and digestive system. Many patients with respiratory difficulties on account of this condition require mechanical ventilation to combat it. Objective To relate the presence of Candida Albicans as an aggravating factor in patients with COVID-19. Materials and methods: A literature review took place using the Redalyc, Scielo, PubMed, Research gate, Science direct, Google Scholar databases. The inclusion criteria used were: articles in English and Spanish, along with articles published from 2020 up to date. There were 65 scientific articles that met the search criteria and were analyzed. The analysis determined that oral candidiasis negatively affects patients with COVID-19 infection, increasing the risk of admission to the ICU with the use of artificial ventilators.
Collapse
Affiliation(s)
- Daniela San Martín Andrade
- Carrera de Odontologia de la Universidad Catolica de Cuenca. Campus Universitario Azogues, Ecuador. , Universidad Católica de Cuenca Carrera de Odontologia Universidad Catolica de Cuenca Campus Universitario Azogues Ecuador
| | - Cristopher Andrés Cárdenas Amendaño
- Universidad Catolica de Cuenca. Campus Universitario Azogues, Ecuador. , , Universidad Católica de Cuenca Universidad Catolica de Cuenca Campus Universitario Azogues Ecuador
| | - Allison Brigitte Solórzano Cuenca
- Universidad Catolica de Cuenca. Campus Universitario Azogues, Ecuador. , , Universidad Católica de Cuenca Universidad Catolica de Cuenca Campus Universitario Azogues Ecuador
| | - Johanna Maribel Ulloa Pacheco
- Universidad Catolica de Cuenca. Campus Universitario Azogues, Ecuador. , , Universidad Católica de Cuenca Universidad Catolica de Cuenca Campus Universitario Azogues Ecuador
| | - Priscilla Medina-Sotomayor
- Carrera de Odontologia de la Universidad Catolica de Cuenca. Campus Universitario Azogues, Ecuador. , Universidad Católica de Cuenca Carrera de Odontologia Universidad Catolica de Cuenca Campus Universitario Azogues Ecuador
| |
Collapse
|
89
|
Thompson GR, Jenks JD, Baddley JW, Lewis JS, Egger M, Schwartz IS, Boyer J, Patterson TF, Chen SCA, Pappas PG, Hoenigl M. Fungal Endocarditis: Pathophysiology, Epidemiology, Clinical Presentation, Diagnosis, and Management. Clin Microbiol Rev 2023; 36:e0001923. [PMID: 37439685 PMCID: PMC10512793 DOI: 10.1128/cmr.00019-23] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023] Open
Abstract
Fungal endocarditis accounts for 1% to 3% of all infective endocarditis cases, is associated with high morbidity and mortality (>70%), and presents numerous challenges during clinical care. Candida spp. are the most common causes of fungal endocarditis, implicated in over 50% of cases, followed by Aspergillus and Histoplasma spp. Important risk factors for fungal endocarditis include prosthetic valves, prior heart surgery, and injection drug use. The signs and symptoms of fungal endocarditis are nonspecific, and a high degree of clinical suspicion coupled with the judicious use of diagnostic tests is required for diagnosis. In addition to microbiological diagnostics (e.g., blood culture for Candida spp. or galactomannan testing and PCR for Aspergillus spp.), echocardiography remains critical for evaluation of potential infective endocarditis, although radionuclide imaging modalities such as 18F-fluorodeoxyglucose positron emission tomography/computed tomography are increasingly being used. A multimodal treatment approach is necessary: surgery is usually required and should be accompanied by long-term systemic antifungal therapy, such as echinocandin therapy for Candida endocarditis or voriconazole therapy for Aspergillus endocarditis.
Collapse
Affiliation(s)
- George R. Thompson
- Department of Internal Medicine, Division of Infectious Diseases, University of California-Davis Medical Center, Sacramento, California, USA
- Department of Medical Microbiology and Immunology, University of California-Davis, Davis, California, USA
| | - Jeffrey D. Jenks
- Durham County Department of Public Health, Durham, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - John W. Baddley
- Department of Medicine, Division of Infectious Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James S. Lewis
- Department of Pharmacy, Oregon Health & Science University, Portland, Oregon, USA
| | - Matthias Egger
- Division of Infectious Diseases, ECMM Excellence Center for Medical Mycology, Department of Medicine, Medical University of Graz, Graz, Austria
| | - Ilan S. Schwartz
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Johannes Boyer
- Division of Infectious Diseases, ECMM Excellence Center for Medical Mycology, Department of Medicine, Medical University of Graz, Graz, Austria
| | - Thomas F. Patterson
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center, San Antonio, Texas, USA
| | - Sharon C.-A. Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, New South Wales Health Pathology, Sydney, New South Wales, Australia
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, The University of Sydney, Sydney, New South Wales, Australia
| | - Peter G. Pappas
- Department of Medicine Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Martin Hoenigl
- Division of Infectious Diseases, ECMM Excellence Center for Medical Mycology, Department of Medicine, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
90
|
Abstract
Candida auris is a multidrug-resistant fungal pathogen that presents a serious threat to global human health. Since the first reported case in 2009 in Japan, C. auris infections have been reported in more than 40 countries, with mortality rates between 30% and 60%. In addition, C. auris has the potential to cause outbreaks in health care settings, especially in nursing homes for elderly patients, owing to its efficient transmission via skin-to-skin contact. Most importantly, C. auris is the first fungal pathogen to show pronounced and sometimes untreatable clinical drug resistance to all known antifungal classes, including azoles, amphotericin B, and echinocandins. In this review, we explore the causes of the rapid spread of C. auris. We also highlight its genome organization and drug resistance mechanisms and propose future research directions that should be undertaken to curb the spread of this multidrug-resistant pathogen.
Collapse
Affiliation(s)
- Anuradha Chowdhary
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India;
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Kusum Jain
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India;
| | - Neeraj Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
91
|
Puerner C, Vellanki S, Strauch JL, Cramer RA. Recent Advances in Understanding the Human Fungal Pathogen Hypoxia Response in Disease Progression. Annu Rev Microbiol 2023; 77:403-425. [PMID: 37713457 PMCID: PMC11034785 DOI: 10.1146/annurev-micro-032521-021745] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Fungal-mediated disease progression and antifungal drug efficacy are significantly impacted by the dynamic infection microenvironment. At the site of infection, oxygen often becomes limiting and induces a hypoxia response in both the fungal pathogen and host cells. The fungal hypoxia response impacts several important aspects of fungal biology that contribute to pathogenesis, virulence, antifungal drug susceptibility, and ultimately infection outcomes. In this review, we summarize recent advances in understanding the molecular mechanisms of the hypoxia response in the most common human fungal pathogens, discuss potential therapeutic opportunities, and highlight important areas for future research.
Collapse
Affiliation(s)
- Charles Puerner
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA;
| | - Sandeep Vellanki
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA;
| | - Julianne L Strauch
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA;
- Department of Biology, Dartmouth College, Hanover, New Hampshire, USA
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA;
| |
Collapse
|
92
|
Liang X, Chen D, Wang J, Liao B, Shen J, Ye X, Wang Z, Zhu C, Gou L, Zhou X, Cheng L, Ren B, Zhou X. Artemisinins inhibit oral candidiasis caused by Candida albicans through the repression on its hyphal development. Int J Oral Sci 2023; 15:40. [PMID: 37699886 PMCID: PMC10497628 DOI: 10.1038/s41368-023-00245-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Candida albicans is the most abundant fungal species in oral cavity. As a smart opportunistic pathogen, it increases the virulence by switching its forms from yeasts to hyphae and becomes the major pathogenic agent for oral candidiasis. However, the overuse of current clinical antifungals and lack of new types of drugs highlight the challenges in the antifungal treatments because of the drug resistance and side effects. Anti-virulence strategy is proved as a practical way to develop new types of anti-infective drugs. Here, seven artemisinins, including artemisinin, dihydroartemisinin, artemisinic acid, dihydroartemisinic acid, artesunate, artemether and arteether, were employed to target at the hyphal development, the most important virulence factor of C. albicans. Artemisinins failed to affect the growth, but significantly inhibited the hyphal development of C. albicans, including the clinical azole resistant isolates, and reduced their damage to oral epithelial cells, while arteether showed the strongest activities. The transcriptome suggested that arteether could affect the energy metabolism of C. albicans. Seven artemisinins were then proved to significantly inhibit the productions of ATP and cAMP, while reduced the hyphal inhibition on RAS1 overexpression strain indicating that artemisinins regulated the Ras1-cAMP-Efg1 pathway to inhibit the hyphal development. Importantly, arteether significantly inhibited the fungal burden and infections with no systemic toxicity in the murine oropharyngeal candidiasis models in vivo caused by both fluconazole sensitive and resistant strains. Our results for the first time indicated that artemisinins can be potential antifungal compounds against C. albicans infections by targeting at its hyphal development.
Collapse
Affiliation(s)
- Xiaoyue Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingchen Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengguang Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
93
|
Matsumoto Y, Sugiyama Y, Nagamachi T, Yoshikawa A, Sugita T. Hog1-mediated stress tolerance in the pathogenic fungus Trichosporon asahii. Sci Rep 2023; 13:13539. [PMID: 37598230 PMCID: PMC10439922 DOI: 10.1038/s41598-023-40825-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023] Open
Abstract
Trichosporon asahii is an opportunistic pathogenic fungus that causes severe and sometimes fatal infections in immunocompromised patients. Hog1, a mitogen-activated protein kinase, regulates the stress resistance of some pathogenic fungi, however its role in T. asahii has not been investigated. Here, we demonstrated that the hog1 gene-deficient T. asahii mutant is sensitive to high temperature, cell membrane stress, oxidative stress, and antifungal drugs. Growth of the hog1 gene-deficient T. asahii mutant was delayed at 40 °C. The hog1 gene-deficient T. asahii mutant also exhibited sensitivity to sodium dodecyl sulfate, hydrogen peroxide, menadione, methyl methanesulfonate, UV exposure, and antifungal drugs such as amphotericin B under a glucose-rich condition. Under a glucose-restricted condition, the hog1 gene-deficient mutant exhibited sensitivity to NaCl and KCl. The virulence of the hog1 gene-deficient mutant against silkworms was attenuated. Moreover, the viability of the hog1 gene-deficient mutant decreased in the silkworm hemolymph. These phenotypes were restored by re-introducing the hog1 gene into the gene-deficient mutant. Our findings suggest that Hog1 plays a critical role in regulating cellular stress responses in T. asahii.
Collapse
Affiliation(s)
- Yasuhiko Matsumoto
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan.
| | - Yu Sugiyama
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Tae Nagamachi
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Asami Yoshikawa
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| |
Collapse
|
94
|
Rapala-Kozik M, Surowiec M, Juszczak M, Wronowska E, Kulig K, Bednarek A, Gonzalez-Gonzalez M, Karkowska-Kuleta J, Zawrotniak M, Satała D, Kozik A. Living together: The role of Candida albicans in the formation of polymicrobial biofilms in the oral cavity. Yeast 2023; 40:303-317. [PMID: 37190878 DOI: 10.1002/yea.3855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
The oral cavity of humans is colonized by diversity of microbial community, although dominated by bacteria, it is also constituted by a low number of fungi, often represented by Candida albicans. Although in the vast minority, this usually commensal fungus under certain conditions of the host (e.g., immunosuppression or antibiotic therapy), can transform into an invasive pathogen that adheres to mucous membranes and also to medical or dental devices, causing mucosal infections. This transformation is correlated with changes in cell morphology from yeast-like cells to hyphae and is supported by numerous virulence factors exposed by C. albicans cells at the site of infection, such as multifunctional adhesins, degradative enzymes, or toxin. All of them affect the surrounding host cells or proteins, leading to their destruction. However, at the site of infection, C. albicans can interact with different bacterial species and in its filamentous form may produce biofilms-the elaborated consortia of microorganisms, that present increased ability to host colonization and resistance to antimicrobial agents. In this review, we highlight the modification of the infectious potential of C. albicans in contact with different bacterial species, and also consider the mutual bacterial-fungal relationships, involving cooperation, competition, or antagonism, that lead to an increase in the propagation of oral infection. The mycofilm of C. albicans is an excellent hiding place for bacteria, especially those that prefer low oxygen availability, where microbial cells during mutual co-existence can avoid host recognition or elimination by antimicrobial action. However, these microbial relationships, identified mainly in in vitro studies, are modified depending on the complexity of host conditions and microbial dominance in vivo.
Collapse
Affiliation(s)
- Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Magdalena Surowiec
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Aneta Bednarek
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Miriam Gonzalez-Gonzalez
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satała
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
95
|
Zhang K, Sun IG, Liao B, Yang Y, Ma H, Jiang A, Chen S, Guo Q, Ren B. Streptococcus mutans sigX-inducing peptide inhibits the virulence of Candida albicans and oral candidiasis through the Ras1-cAMP-Efg1 pathway. Int J Antimicrob Agents 2023; 62:106855. [PMID: 37211262 DOI: 10.1016/j.ijantimicag.2023.106855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/23/2023]
Abstract
Oral candidiasis is the most common fungal infectious disease in the human oral cavity, and Candida albicans is the major pathogenic agent. Increasing drug resistance and the lack of new types of antifungals greatly increase the challenges for treating fungal infections. Targeting hyphal transition provides a promising strategy to inhibit the virulence of C. albicans and overcome drug resistance. This study aimed to investigate the effects and mechanisms of sigX-inducing peptide (XIP), a quorum-sensing signal peptide secreted by Streptococcus mutans, on C. albicans hyphal development and biofilm formation in vitro and oropharyngeal candidiasis in vivo. XIP significantly inhibited C. albicans yeast-to-hypha transition and biofilm formation in a dose-dependent manner from 0.01 to 0.1 µM. XIP significantly downregulated expression of genes from the Ras1-cAMP-Efg1 pathway (RAS1, CYR1, TPK2, EFG1 and UME6), a key pathway to regulate C. albicans hyphal development. Importantly, XIP reduced the levels of key molecules cAMP and ATP from this pathway, while the addition of exogenous cAMP and overexpression of RAS1 restored the hyphal development inhibited by XIP. XIP also lost its hyphal inhibitory effects on ras1Δ/Δ and efg1Δ/Δ strains. These results further confirmed that XIP inhibited hyphal development through downregulation of the Ras1-cAMP-Efg1 pathway. A murine oropharyngeal candidiasis model was employed to evaluate the therapeutic effects of XIP on oral candidiasis. XIP effectively reduced the infected epithelial area, fungal burden, hyphal invasion and inflammatory infiltrates. These results revealed the antifungal effects of XIP, and highlighted that XIP can be a potential antifungal peptide against C. albicans infection.
Collapse
Affiliation(s)
- Kaiwen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Orthodontics Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ivy Guofang Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yichun Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huangshui Ma
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Aiming Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Orthodontics Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
96
|
Chen HT, Li JS, Li J, Li L, Xu ZC, Zhang Y, Wang RR. Lactobacillus murinus: A key factor in suppression of enterogenous Candida albicans infections in Compound Agrimony enteritis capsules-treated mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116361. [PMID: 36963475 DOI: 10.1016/j.jep.2023.116361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/04/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Compound Agrimony (FuFangXianHeCao, FFXHC) Enteritis Capsules is an ethnomedicine that is derived from Yi Nationality Herbal Medicine for enteritis treatment. We found that FFXHC reduced the mortality outcomes in enterogenic Candida albicans infected mice models and increased the abundance of Lactobacillus murinus in the intestines. Lactobacillus murinus exhibited comparable therapeutic effects to those of FFXHC in enterogenic Candida albicans infected mice. This study provides novel perspectives into the pharmacological mechanisms of FFXHC. AIM OF THE STUDY We investigated the mechanisms via which FFXHC inhibits C. albicans infections and its effects on L. murinus. MATERIALS AND METHODS Enterogenous C. albicans infection mice models were established and various parameters, including survival rate, weight change, number of colonies, treatment effects on intestinal mucosa, microecology, and immune cytokines evaluated. Susceptibility of C. albicans to L. murinus was evaluated in vitro. RESULTS Treatment with FFXHC reduced the number of colonies, improved the health status, enhanced the survival rates, increased the abundance of L. murinus, reduced damage to the intestinal mucosa, and elevated occludin as well as claudin-1 levels. Interestingly, TNF-α, IFN-γ, IL-10, IL-22, and IL-17A levels were increased while IL-1β levels were suppressed in the intestinal mucosa without any change in peripheral blood cytokine levels. Moreover, FFXHC promoted L. murinus proliferation. This study also confirmed the incubation-dependent anti-C. albicans effects exerted by the metabolic supernatants of L. murinus. CONCLUSIONS FFXHC effectively alleviated intestinal infections of C. albicans in mice and increased the abundance of L. murinus. Supplementation of L. murinus in food can achieve the effects that are comparable to those of FFXHC. Thus, L. murinus maybe essential in FFXHC-based treatment of intestinal C. albicans infections.
Collapse
Affiliation(s)
- Hui-Ting Chen
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Jia-Sheng Li
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Jun Li
- Yunnan University of Chinese Medicine, Kunming, 650500, China; Hospital of Traditional Chinese Medicine and Western Medicine of Panzhihua, Panzhihua, 617099, China
| | - Li Li
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Zhi-Chang Xu
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Yi Zhang
- Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Rui-Rui Wang
- Yunnan University of Chinese Medicine, Kunming, 650500, China.
| |
Collapse
|
97
|
Zeng H, Stadler M, Abraham WR, Müsken M, Schrey H. Inhibitory Effects of the Fungal Pigment Rubiginosin C on Hyphal and Biofilm Formation in Candida albicans and Candida auris. J Fungi (Basel) 2023; 9:726. [PMID: 37504715 PMCID: PMC10381533 DOI: 10.3390/jof9070726] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023] Open
Abstract
The two fungal human pathogens, Candida auris and Candida albicans, possess a variety of virulence mechanisms. Among them are the formation of biofilms to protect yeast against harsh conditions through the development of (pseudo)hyphae whilst also facilitating the invasion of host tissues. In recent years, increased rates of antifungal resistance have been associated with C. albicans and C. auris, posing a significant challenge for the effective treatment of fungal infections. In the course of our ongoing search for novel anti-infectives, six selected azaphilones were tested for their cytotoxicity and antimicrobial effects as well as for their inhibitory activity against biofilm and hyphal formation. This study revealed that rubiginosin C, derived from stromata of the ascomycete Hypoxylon rubiginosum, effectively inhibited the formation of biofilms, pseudohyphae, and hyphae in both C. auris and C. albicans without lethal effects. Crystal violet staining assays were utilized to assess the inhibition of biofilm formation, while complementary microscopic techniques, such as confocal laser scanning microscopy, scanning electron microscopy, and optical microscopy, were used to investigate the underlying mechanisms. Rubiginosin C is one of the few substances known to effectively target both biofilm formation and the yeast-to-hyphae transition of C. albicans and C. auris within a concentration range not affecting host cells, making it a promising candidate for therapeutic intervention in the future.
Collapse
Affiliation(s)
- Haoxuan Zeng
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Marc Stadler
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Wolf-Rainer Abraham
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Hedda Schrey
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| |
Collapse
|
98
|
Li Y, Chen C, Cong L, Mao S, Shan M, Han Z, Mao J, Xie Z, Zhu Z. Inhibitory Effects of a Maleimide Compound on the Virulence Factors of Candida albicans. Virulence 2023:2230009. [PMID: 37367101 DOI: 10.1080/21505594.2023.2230009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/26/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
Candidiasis caused by Candida albicans infection has long been a serious human health problem. The pathogenicity of C. albicans is mainly due to its virulence factors, which are the novel targets of antifungal drugs for low risk of resistance development. In this study, we identified a maleimide compound [1-(4-methoxyphenyl)-1hydro-pyrrole-2,5-dione, MPD] that exerts effective anti-virulence activity. It could inhibit the process of adhesion, filamentation, and biofilm formation in C. albicans. In addition, it exhibited low cytotoxicity, hemolytic activity and drug resistance development. Moreover, in Galleria mellonella-C. albicans (in vivo) infection model, the survival time of infected larvae was significantly prolonged under the treatment of MPD. Further, mechanism research revealed that MPD increased farnesol secretion by upregulating the expression of Dpp3. The increased farnesol inhibited the activity of Cdc35, which then decreased the intracellular cAMP content resulting in the inhibition of virulence factors via the Ras1-cAMP-Efg1 pathway. In all, this study evaluated the inhibitory effect of MPD on various virulence factors of C. albicans and identified the underlying mechanisms. This suggests a potential application of MPD to overcome fungal infections in clinic.
Collapse
Affiliation(s)
- Ying Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Chaoqun Chen
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Liu Cong
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Shanshan Mao
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Mingzhu Shan
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
- Clinical laboratory, The Central Hospital of Xuzhou City, Xuzhou, China
| | - Zibing Han
- Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Jiayi Mao
- Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Zhiyu Xie
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, China
| | - Zuobin Zhu
- Department of Genetics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
99
|
Lin L, Zhuo Y, Dong Q, Yang C, Cheng C, Liu T. Plasma activated Ezhangfeng Cuji as innovative antifungal agent and its inactivation mechanism. AMB Express 2023; 13:65. [PMID: 37368076 DOI: 10.1186/s13568-023-01571-6] [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: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Candida albicans is a highly drug-resistant fungus for which new treatments are urgently needed due to the lack of clinically effective options. In this study, we evaluated the antifungal activity and mechanism of plasma-activated Ezhangfeng Cuji (PAEC) against Candida albicans and compared it with physiological saline (PS), plasma-activated physiological saline (PAPS) and Ezhangfeng Cuji (EC). After dielectric barrier discharge (DBD) plasma treatment with EC for 20 min followed by a 10 min immersion of Candida albicans, the fungus was reduced by approximately 3 orders of magnitude. High performance liquid chromatography (HPLC) results showed an increase of 41.18% and 129.88% in the concentration of oxymatrine and rhein, respectively, after plasma-treated EC. The concentrations of reactive species (RS), such as H2O2, [Formula: see text], and O3, were found to be higher and the pH value was getting lower in PS after plasma treatment. Detailed analysis of intracellular material leakage, reactive oxygen species (ROS), apoptosis for Candida albicans and observation by transmission electron microscopy (TEM) and scanning electron microscopy (SEM) demonstrated that PAPS, EC and PAEC disrupt the morphological structure of Candida albicans to varying degrees.Additionally, specific analyses on Candida albicans virulence factors, such as adhesion to tissue surfaces, cell surface hydrophobicity (CSH), the transition of yeast-phase cells to mycelium-phase cells, and the secretion of hydrolytic enzymes for Candida albicans were conducted and found to be inhibited after PAPS/EC/PAEC treatment. In our investigation, the inhibitory effects on Candida albicans were ranked from strong to weak as follows: PAEC, EC, PAPS, and PS.
Collapse
Affiliation(s)
- Lin Lin
- The Postgraduate School of Anhui, University of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Yue Zhuo
- Department of Dermatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
- Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qiran Dong
- Department of Dermatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Chunjun Yang
- Department of Dermatology, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230601, People's Republic of China
| | - Cheng Cheng
- Institute of Plasma Physics, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Taofeng Liu
- Department of Dermatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China.
| |
Collapse
|
100
|
Tarrés-Solé A, Battistini F, Gerhold JM, Piétrement O, Martínez-García B, Ruiz-López E, Lyonnais S, Bernadó P, Roca J, Orozco M, Le Cam E, Sedman J, Solà M. Structural analysis of the Candida albicans mitochondrial DNA maintenance factor Gcf1p reveals a dynamic DNA-bridging mechanism. Nucleic Acids Res 2023; 51:5864-5882. [PMID: 37207342 PMCID: PMC10287934 DOI: 10.1093/nar/gkad397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 04/01/2023] [Accepted: 05/03/2023] [Indexed: 05/21/2023] Open
Abstract
The compaction of mitochondrial DNA (mtDNA) is regulated by architectural HMG-box proteins whose limited cross-species similarity suggests diverse underlying mechanisms. Viability of Candida albicans, a human antibiotic-resistant mucosal pathogen, is compromised by altering mtDNA regulators. Among them, there is the mtDNA maintenance factor Gcf1p, which differs in sequence and structure from its human and Saccharomyces cerevisiae counterparts, TFAM and Abf2p. Our crystallographic, biophysical, biochemical and computational analysis showed that Gcf1p forms dynamic protein/DNA multimers by a combined action of an N-terminal unstructured tail and a long helix. Furthermore, an HMG-box domain canonically binds the minor groove and dramatically bends the DNA while, unprecedentedly, a second HMG-box binds the major groove without imposing distortions. This architectural protein thus uses its multiple domains to bridge co-aligned DNA segments without altering the DNA topology, revealing a new mechanism of mtDNA condensation.
Collapse
Affiliation(s)
- Aleix Tarrés-Solé
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | - Federica Battistini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Department of Biochemistry, University of Barcelona, Barcelona 08028, Spain
| | - Joachim M Gerhold
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Olivier Piétrement
- Genome Integrity and Cancer UMR 9019 CNRS, Université Paris Saclay, Gustave Roussy Campus, 114 rue Edouard Vaillant 94805VillejuifCedex, France
| | | | - Elena Ruiz-López
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | - Sébastien Lyonnais
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | - Pau Bernadó
- Centre de Biologie Structurale (CBS), Inserm, CNRS and Université de Montpellier, France, Sébastien Lyonnais, UAR 3725 CNRS, Université de Montpellier, 34000 Montpellier, France
| | - Joaquim Roca
- Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Department of Biochemistry, University of Barcelona, Barcelona 08028, Spain
| | - Eric Le Cam
- Genome Integrity and Cancer UMR 9019 CNRS, Université Paris Saclay, Gustave Roussy Campus, 114 rue Edouard Vaillant 94805VillejuifCedex, France
| | - Juhan Sedman
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Maria Solà
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| |
Collapse
|