1
|
Zhu S, Sun J, Bi X, Wang Z, Yang F, Ta R, Su C, Wu X, Han K, Lan M, Hu H, Li H, Li Y. Chemical composition of the traditional Chinese medicine compound (ICAM), its antifungal effects against Candida albicans, and the underlying Mechanisms: Therapeutic potential and safety evaluation for vulvovaginal candidiasis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119844. [PMID: 40254107 DOI: 10.1016/j.jep.2025.119844] [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: 03/12/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) compound preparations play a significant role in the clinical treatment of vulvovaginal candidiasis (VVC). AIM OF THE STUDY Candida albicans (CA) is an opportunistic fungal pathogen responsible for various human diseases, including vulvovaginal candidiasis (VVC). Hyphal growth and biofilm formation are critical virulence factors contributing to CA's pathogenicity and drug resistance. ICAM, a topical TCM compound preparation developed by our laboratory, was investigated for its chemical component, antifungal mechanisms against CA and therapeutic efficacy against VVC. MATERIALS AND METHODS The main components of ICAM were analyzed using the Gas Chromatography-Mass Spectrometry (GC-MS) method. To elucidate the mechanisms underlying ICAM's antifungal activity, we combined phenotypic assays, transcriptomic and proteomic analyses. The therapeutic potential of ICAM for VVC and its irritancy to vaginal tissue were evaluated using cavity model experiments. RESULTS ICAM contained a diverse range of phenolic compounds, such as phenol, 2-methoxyphenol, and 4-ethyl-2-methoxyphenol, among others. The minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) of ICAM against CA were 2.50 % and 10.00 % for the standard strain, and 5.00 % and 20.00 % for the clinical strain, respectively. At 1.25 %, ICAM significantly inhibited CA adherence, hyphal growth, and biofilm formation, while also reducing surface hydrophobicity and exopolysaccharide production. Treatment with 10.00 % ICAM completely disrupted CA membrane integrity. Transcriptome analysis revealed that multiple genes associated with biofilm and hyphal formation, including five MAPK signaling pathway genes (Ras1, Cdc24, Ste11, Cek1, Hst7), four hyphae-specific genes (Hgc1, Hwp1, Ece1, Als3), and three additional genes (Tec1, Csh1, Pmt1), were significantly downregulated. Additionally, proteins associated with the MAPK signaling pathway, including the 14-3-3 domain-containing protein, cell wall protein RTB1, Msb2p, Ras family protein, and RhoGAP domain family protein, were significantly downregulated. These findings suggest that the MAPK signaling pathway plays a crucial role in mediating ICAM's inhibition of hyphal growth and biofilm formation in CA. In vivo, 10.00 % ICAM completely eliminated the symptoms of CA infection. The vaginal fungal burden in the 20.00 % and 40.00 % ICAM groups was reduced to zero after 12 days of treatment. Furthermore, 40.00 % ICAM significantly reduced lactate dehydrogenase and inflammatory cytokine levels, demonstrating efficacy comparable to the positive control. ICAM demonstrated excellent mucosal compatibility in the cavity experiment. CONCLUSIONS These findings highlight the potential of ICAM as a novel antifungal agent for the treatment of VVC.
Collapse
Affiliation(s)
- Shuang Zhu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, Guizhou, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, China
| | - Jianfang Sun
- Department of Obstetrics and Gynecology, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China
| | - Xueling Bi
- Obstetrics and Gynecology Outpatient Department, Yan'an People's Hospital, Yan'an, Shaanxi, 716000, China
| | - Ziyi Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, Guizhou, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, China
| | - Fenge Yang
- Department of Obstetrics and Gynecology, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China; Outpatient Department of Pediatrics, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China
| | - Rongrong Ta
- Department of Obstetrics and Gynecology, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China; Outpatient Department of Pediatrics, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China
| | - Chengli Su
- Department of Obstetrics and Gynecology, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China; Outpatient Department of Pediatrics, Yan'an Municipal Traditional Chinese Medicine Hospital, Yan'an, Shaanxi, 716000, China
| | - Xiaomei Wu
- Department of Medicine, Yan'an Vocational & Technical College, Yanan Shaanxi, 716000, China
| | - Kezhan Han
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, Guizhou, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, China
| | - Meng Lan
- Yan'an Consumer Rights Protection Center, Yanan Shaanxi, 716000, China
| | - Huijun Hu
- Department of Criminal Investigation, Yan'an Pollice Office, Yanan Shaanxi, 716000, China
| | - Hui Li
- Obstetrics and Gynecology Outpatient Department, Yan'an People's Hospital, Yan'an, Shaanxi, 716000, China.
| | - Yan Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, Guizhou, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, China
| |
Collapse
|
2
|
Zhang M, Yang W, Liu N, Tu J, Lin J, Dong G, Zhao D, Sheng C. Lanosterol 14α-Demethylase (CYP51)/Heat Shock Protein 90 (Hsp90) Dual Inhibitors for the Treatment of Invasive Candidiasis. J Med Chem 2025; 68:1668-1681. [PMID: 39754582 DOI: 10.1021/acs.jmedchem.4c02305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Invasive candidiasis has attracted global attention with a high incidence and mortality. Current antifungal drugs are limited by unfavorable therapeutic efficacy, significant hepatorenal toxicity, and the development of drug resistance. Herein, we designed the first generation of lanosterol 14α-demethylase (CYP51)/heat shock protein 90 (Hsp90) dual inhibitors on the basis of antifungal synergism. Among them, dual inhibitor MM4 exhibited potent in vitro and in vivo antifungal activity against Candida albicans and effectively inhibited important fungal virulence factors (e.g., hyphae, biofilm). Therefore, CYP51/Hsp90 dual inhibitors show great promise in the development of novel antifungal drugs to combat invasive candidiasis.
Collapse
Affiliation(s)
- Mingming Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| | - Wanzhen Yang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| | - Na Liu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| | - Jie Tu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| | - Jingsheng Lin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Guoqiang Dong
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| | - Dongmei Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
3
|
Gharieb MM, Rizk A, Elfeky N. Anticandidal activity of a wild Bacillus subtilis NAM against clinical isolates of pathogenic Candida albicans. ANN MICROBIOL 2024; 74:23. [DOI: 10.1186/s13213-024-01764-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/13/2024] [Indexed: 01/03/2025] Open
Abstract
Abstract
Background
Resistance to antifungal medications poses a significant obstacle in combating fungal infections. The development of novel therapeutics for Candida albicans is necessary due to the increasing resistance of candidiasis to the existing medications. The utilization of biological control is seen as a more advantageous and less hazardous strategy therefore the objective of this study is to identify the antifungal properties of Bacillus subtilis against pathogenic C. albicans.
Results
We conducted a study to evaluate the antifungal properties of three bacterial isolates against the human pathogen Candida albicans. One of the bacterial isolates exhibited a potent antifungal activity against this fungal pathogen. This bacterium was identified as Bacillus subtilis based on the 16Sr RNA gene sequence. It exhibited inhibitory efficacy ranging from 33.5 to 44.4% against 15 Candida isolates. The optimal incubation duration for achieving the maximum antifungal activity was determined to be 48 h, resulting in a mean inhibition zone diameter of 29 ± 0.39 mm. The Potato Dextrose agar (PDA) medium was the best medium for the most effective antifungal activity. Incubation temperature of 25oC and medium pH value of 8.0 were the most favorable conditions for maximum antagonistic activity that resulted fungal growth inhibition of 40 ± 0.16 and 36 ± 0.94 mm respectively. Furthermore, the addition of 10.5 mg/ml of bacterial filtrate to C. albicans colonies resulted in 86.51%. decrease in the number of germinated cells. The fungal cell ultrastructural responses due to exposure to B. subtilis filtrate after 48 h were investigated using transmission electron microscopy (TEM). It revealed primary a drastic abnormality that lead to cellular disintegration including folding and lysis of the cell wall, total collapse of the yeast cells, and malformed germ tube following the exposure to the filtrate. However, the control culture treatment had a characteristic morphology of the normal fungal cells featuring a consistently dense central region, a well-organized nucleus, and a cytoplasm containing several components of the endomembrane system. The cells were surrounded by a uniform and intact cell wall.
Conclusion
The current study demonstrates a notable antifungal properties of B. subtilis against C. albicans as a result of production of bioactive components of the bacterial exudate. This finding could be a promising natural antifungal agent that could be utilized to combat C. albicans.
Collapse
|
4
|
do Nascimento Dias J, Hurtado Erazo FA, Bessa LJ, Eaton P, Leite JRDSDA, Paes HC, Nicola AM, Silva-Pereira I, Albuquerque P. Synergic Effect of the Antimicrobial Peptide ToAP2 and Fluconazole on Candida albicans Biofilms. Int J Mol Sci 2024; 25:7769. [PMID: 39063009 PMCID: PMC11276877 DOI: 10.3390/ijms25147769] [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: 05/29/2024] [Revised: 06/29/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Candida albicans is one of the agents of invasive candidiasis, a life-threatening disease strongly associated with hospitalization, particularly among patients in intensive care units with central venous catheters. This study aimed to evaluate the synergistic activity of the antifungal peptide ToAP2 combined with fluconazole against C. albicans biofilms grown on various materials. We tested combinations of different concentrations of the peptide ToAP2 with fluconazole on C. albicans biofilms. These biofilms were generated on 96-well plates, intravenous catheters, and infusion tubes in RPMI medium at two maturation stages. Scanning electron microscopy and atomic force microscopy were employed to assess the biofilm structure. We also evaluated the expression of genes previously proven to be involved in C. albicans biofilm formation in planktonic and biofilm cells after treatment with the peptide ToAP2 using qPCR. ToAP2 demonstrated a synergistic effect with fluconazole at concentrations up to 25 µM during both the early and mature stages of biofilm formation in 96-well plates and on medical devices. Combinations of 50, 25, and 12.5 µM of ToAP2 with 52 µM of fluconazole significantly reduced the biofilm viability compared to individual treatments and untreated controls. These results were supported by substantial structural changes in the biofilms observed through both scanning and atomic force microscopy. The gene expression analysis of C. albicans cells treated with 25 µM of ToAP2 revealed a decrease in the expression of genes associated with membrane synthesis, along with an increase in the expression of genes involved in efflux pumps, adhesins, and filamentation. Our results highlight the efficacy of the combined ToAP2 and fluconazole treatment against C. albicans biofilms. This combination not only shows therapeutic potential but also suggests its utility in developing preventive biofilm tools for intravenous catheters.
Collapse
Affiliation(s)
- Jhones do Nascimento Dias
- Laboratory of Molecular Biology of Fungi, University of Brasilia, Brasilia 70910-900, Brazil; (J.d.N.D.); (F.A.H.E.)
| | - Fabián Andrés Hurtado Erazo
- Laboratory of Molecular Biology of Fungi, University of Brasilia, Brasilia 70910-900, Brazil; (J.d.N.D.); (F.A.H.E.)
| | - Lucinda J. Bessa
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal; (L.J.B.); (P.E.)
| | - Peter Eaton
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal; (L.J.B.); (P.E.)
- The Bridge, School of Chemistry, University of Lincoln, Lincoln LN6 7TS, UK
| | | | - Hugo Costa Paes
- Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil; (H.C.P.); (A.M.N.)
| | - André Moraes Nicola
- Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil; (H.C.P.); (A.M.N.)
| | - Ildinete Silva-Pereira
- Laboratory of Molecular Biology of Fungi, University of Brasilia, Brasilia 70910-900, Brazil; (J.d.N.D.); (F.A.H.E.)
| | - Patrícia Albuquerque
- Laboratory of Molecular Biology of Fungi, University of Brasilia, Brasilia 70910-900, Brazil; (J.d.N.D.); (F.A.H.E.)
| |
Collapse
|
5
|
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
|
6
|
Cao L, Tan J, Zhang Z, Lin B, Mu Y, Jiang M, Jiang Y, Huang X, Han L. Discovery of Antifungal Norsesquiterpenoids from a Soil-Derived Streptomyces microflavus: Targeting Biofilm Formation and Synergistic Combination with Amphotericin B against Yeast-like Fungi. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8521-8535. [PMID: 38565849 DOI: 10.1021/acs.jafc.3c08707] [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: 04/04/2024]
Abstract
Thirty-five norsesquiterpenoids were isolated from the fermentation broth of Streptomyces microflavus from the forest soil of Ailaoshan in China. The structures of new compounds (1-5, 10-26) were elucidated by comprehensive spectroscopic analysis including data from experimental and calculated ECD spectra, as well as Mosher's reagent derivatives method. Norsesquiterpenoids showed different levels of antifungal activity with MIC80 values ranging from 25 to 200 μg/mL against Candida albicans, Candida parapsilosis, and Cryptococcus neoformans. The combining isolated norsesquiterpenoids with amphotericin B resulted in a synergistic interaction against test yeast-like fungi with a fractional inhibitory concentration index < 0.5. Compound 33 significantly inhibited biofilm formation and destroyed the preformed biofilm of fungi. Moreover, 33 downregulated the expression of adhesion-related genes HWP1, ALS1, ALS3, ECE1, EAP1, and BCR1 to inhibit the adhesion of C. albicans. Findings from the current study highlight the potential usage of norsesquiterpenoids from soil-derived Streptomyces for antifungal leads discovery.
Collapse
Affiliation(s)
- Lu Cao
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Junfeng Tan
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Zengguang Zhang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Bin Lin
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Mu
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Mingguo Jiang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University, Nanning 530008, China
| | - Yi Jiang
- Yunnan Institute of Microbiology, Yunnan University, Kunming 650091, China
| | - Xueshi Huang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Li Han
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| |
Collapse
|
7
|
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
|
8
|
Chen Y, Gao Y, Li Y, Yin J. Anti-Biofilm Activity of Assamsaponin A, Theasaponin E1, and Theasaponin E2 against Candida albicans. Int J Mol Sci 2024; 25:3599. [PMID: 38612411 PMCID: PMC11011434 DOI: 10.3390/ijms25073599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Biofilm formation plays a crucial role in the pathogenesis of Candida albicans and is significantly associated with resistance to antifungal agents. Tea seed saponins, a class of non-ionic triterpenes, have been proven to have fungicidal effects on planktonic C. albicans. However, their anti-biofilm activity and mechanism of action against C. albicans remain unclear. In this study, the effects of three Camellia sinensis seed saponin monomers, namely, theasaponin E1 (TE1), theasaponin E2 (TE2), and assamsaponin A (ASA), on the metabolism, biofilm development, and expression of the virulence genes of C. albicans were evaluated. The results of the XTT reduction assay and crystal violet (CV) staining assay demonstrated that tea seed saponin monomers concentration-dependently suppressed the adhesion and biofilm formation of C. albicans and were able to eradicate mature biofilms. The compounds were in the following order in terms of their inhibitory effects: ASA > TE1 > TE2. The mechanisms were associated with reductions in multiple crucial virulence factors, including cell surface hydrophobicity (CSH), adhesion ability, hyphal morphology conversion, and phospholipase activity. It was further demonstrated through qRT-PCR analysis that the anti-biofilm activity of ASA and TE1 against C. albicans was attributed to the inhibition of RAS1 activation, which consequently suppressed the cAMP-PKA and MAPK signaling pathways. Conversely, TE2 appeared to regulate the morphological turnover and hyphal growth of C. albicans via a pathway that was independent of RAS1. These findings suggest that tea seed saponin monomers are promising innovative agents against C. albicans.
Collapse
Affiliation(s)
- Yuhong Chen
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
- Tea Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Ying Gao
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
| | - Yifan Li
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
| | - Junfeng Yin
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China; (Y.C.); (Y.L.)
| |
Collapse
|
9
|
Ma Y, Sui J, Wang Y, Sun W, Yi G, Wu J, Qiu S, Wang L, Zhang A, He X. RNA-Seq-Based Transcriptomics and GC-MS Quantitative Analysis Reveal Antifungal Mechanisms of Essential Oil of Clausena lansium (Lour.) Skeels Seeds against Candida albicans. Molecules 2023; 28:8052. [PMID: 38138542 PMCID: PMC10745804 DOI: 10.3390/molecules28248052] [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: 10/31/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Infections caused by Candida albicans (C. albicans) and increasing resistance to commonly used drugs lead to a variety of mucosal diseases and systemic infectious diseases. We previously confirmed that the essential oil of Clausena lansium (Lour.) Skeels seeds (CSEO) had antifungal activity against C. albicans, but the detailed mechanism between the chemical components and antifungal activity is unclear. In this study, a quantitative analysis of five volatile components of CSEO, including sabinene, α-phellandrene, β-phellandrene, 4-terpineol, and β-caryophyllene, was carried out using the gas chromatography-mass spectrometry (GC-MS) method. Both the broth dilution and kinetic growth methods proved that the antifungal activity of CSEO against fluconazole-resistant C. albicans was better than that of its main components (sabinene and 4-terpineol). To further investigate the inhibitory mechanism, the transcriptional responses of C. albicans to CSEO, sabinene, and 4-terpineol treatment were determined based on RNA-seq. The Venn diagram and clustering analysis pattern of differential expression genes showed the mechanism of CSEO and 4-terpineol's anti-C. albicans activity might be similar from the perspective of the genes. Functional enrichment analysis suggested that CSEO regulated adherence-, hyphae-, and biofilm-formation-related genes, which may be CSEO's active mechanism of inhibiting the growth of fluconazole-resistant C. albicans. Overall, we preliminarily revealed the molecular mechanism between the chemical components and the antifungal activity of CSEO against C. albicans. This study provides new insights to overcome the azole resistance of C. albicans and promote the development and application of C. lansium (Lour.) Skeels seeds.
Collapse
Affiliation(s)
- Yinzheng Ma
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
- School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Jinlei Sui
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Yan Wang
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Wanying Sun
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Guohui Yi
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Jinyan Wu
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Shi Qiu
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Lili Wang
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Aihua Zhang
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
| | - Xiaowen He
- Public Research Center, Hainan Medical University, Haikou 571199, China; (Y.M.); (J.S.); (Y.W.); (W.S.); (G.Y.); (J.W.); (S.Q.); (L.W.)
- School of Pharmacy, Hainan Medical University, Haikou 571199, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island, Emergency Medicine of Chinese Academy of Medical Sciences, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
10
|
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: 12] [Impact Index Per Article: 6.0] [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
|
11
|
Bao MY, Li M, Bu QR, Yang Y, Song H, Wang CZ, Wang TM, Li N. The effect of herbal medicine in innate immunity to Candida albicans. Front Immunol 2023; 14:1096383. [PMID: 37483621 PMCID: PMC10359817 DOI: 10.3389/fimmu.2023.1096383] [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: 11/12/2022] [Accepted: 03/06/2023] [Indexed: 07/25/2023] Open
Abstract
Candida albicans (C. albicans) is an opportunistic pathogenic fungus that often causes mucosal and systemic infections. Several pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) and C-type lectin receptors (CLRs), have been implicated in the host recognition of C. albicans. These PRRs recognize the pathogen-associated molecular patterns (PAMPs) of C. albicans to activate innate immune cells, thereby rapidly inducing various inflammatory responses by activating intracellular signaling cascades. Herbal medicine and its active components deserve priority development due to their low toxicity and high antibacterial, antiviral and antifungal activities. This review discussed the activities of herbal compounds against C. albicans and their related mechanisms, especially their regulatory role on innate immune cells such as neutrophils, macrophages, and dendritic cells (DCs) implicated in C. albicans infections. Our work aims to find new therapeutic drugs and targets to prevent and treat diseases caused by C. albicans infection with the mechanisms by which this fungus interacts with the innate immune response.
Collapse
Affiliation(s)
- Meng-Yuan Bao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ming Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qing-Ru Bu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Yang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chang-Zhong Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tian-Ming Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ning Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Chudzik-Rząd B, Zalewski D, Kasela M, Sawicki R, Szymańska J, Bogucka-Kocka A, Malm A. The Landscape of Gene Expression during Hyperfilamentous Biofilm Development in Oral Candida albicans Isolated from a Lung Cancer Patient. Int J Mol Sci 2022; 24:ijms24010368. [PMID: 36613809 PMCID: PMC9820384 DOI: 10.3390/ijms24010368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The filamentation ability of Candida albicans represents one of the main virulence factors allowing for host tissue penetration and biofilm formation. The aim of this paper was to study the genetic background of the hyperfilamentous biofilm development in vitro in C. albicans isolated from the oral cavity of a lung cancer patient. Analyzed C. albicans isolates (CA1, CA2, CA3) were chosen based on their different structures of mature biofilm. The CA3 isolate possessing hyperfilamentation properties and forming high biofilm was compared with CA1 and CA2 isolates exhibiting low or average biofilm-forming ability, respectively. The detailed biofilm organization was studied with the use of confocal scanning laser microscopy. The whole transcriptome analysis was conducted during three stages of biofilm development (24 h, 48 h, 72 h). In contrast to CA1 and/or CA2 isolate, the CA3 isolate was characterized by a significant upregulation of genes encoding for cell wall proteins (HWP1, PGA13, PGA44, ALS3) and candidalysin (ECE1), as well as being involved in iron metabolism (FRE1, ALS3), sulfur metabolism (HAL21), the degradation of aromatic compounds (HQD2), and membrane transport (DIP5, PHO89, TNA1). In contrast, some genes (SCW11, FGR41, RBE1) in the CA3 were downregulated. We also observed the overexpression of a few genes over time-mainly FRE1, ATX1, CSA2 involved in iron metabolism. This is the first insight into the potential function of multiple genes in the hyperfilamentous biofilm formation in C. albicans, primarily isolated from host tissue, which may have an important clinical impact on cancer patients. Moreover, the presented data can lay the foundation for further research on novel pathogen-specific targets for antifungal drugs.
Collapse
Affiliation(s)
- Beata Chudzik-Rząd
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
| | - Daniel Zalewski
- Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Martyna Kasela
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
- Correspondence: (M.K.); (A.M.); Tel.: +48-81448-7100 (M.K. & A.M.)
| | - Rafał Sawicki
- Department of Biochemistry and Biotechnology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
| | - Jolanta Szymańska
- Department of Comprehensive Paediatric and Adult Dentistry, Medical University of Lublin, 6 Chodźki St., 20-093 Lublin, Poland
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
- Correspondence: (M.K.); (A.M.); Tel.: +48-81448-7100 (M.K. & A.M.)
| |
Collapse
|
13
|
Vahedi F, Ghasemi Y, Atapour A, Zomorodian K, Ranjbar M, Monabati A, Nezafat N, Savardashtaki A. B-Cell Epitope Mapping from Eight Antigens of Candida albicans to Design a Novel Diagnostic Kit: An Immunoinformatics Approach. Int J Pept Res Ther 2022; 28:110. [PMID: 35669279 PMCID: PMC9136830 DOI: 10.1007/s10989-022-10413-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2022] [Indexed: 12/24/2022]
Abstract
Invasive candidiasis is an emerging fungal infection and a leading cause of morbidity in health care facilities. Despite advances in antifungal therapy, increased antifungal drug resistance in Candida albicans has enhanced patient fatality. The most common method for Candida albicans diagnosing is blood culture, which has low sensitivity. Therefore, there is an urgent need to establish a valid diagnostic method. Our study aimed to use the bioinformatics approach to design a diagnostic kit for detecting Candida albicans with high sensitivity and specificity. Eight antigenic proteins of Candida albicans (HYR1, HWP1, ECE1, ALS, EAP1, SAP1, BGL2, and MET6) were selected. Next, a construct containing different immunodominant B-cell epitopes was derived from the antigens and connected using a suitable linker. Different properties of the final construct, such as physicochemical properties, were evaluated. Moreover, the designed construct underwent 3D modeling, reverse translation, and codon optimization. The results confirmed that the designed construct could identify Candida albicans with high sensitivity and specificity in serum samples of patients with invasive candidiasis. However, experimental studies are needed for final confirmation.
Collapse
Affiliation(s)
- Farzaneh Vahedi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamiar Zomorodian
- Department of Parasitology & Mycology, School of Medicines, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Ranjbar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Monabati
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Haghighi F, Andriasian L, Tran NC, Lux R. Effect of Cigarette and E-Cigarette Smoke Condensates on Candida albicans Biofilm Formation and Gene Expression. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:4626. [PMID: 35457494 PMCID: PMC9029603 DOI: 10.3390/ijerph19084626] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023]
Abstract
Smoking triggers environmental changes in the oral cavity and increases the risk of mucosal infections caused by Candida albicans such as oral candidiasis. While cigarette smoke has a significant impact on C. albicans, how e-cigarettes affect this oral pathogen is less clear. Here, we investigated the effect of cigarette and e-cigarette smoke condensates (CSC and ECSC) on C. albicans growth, biofilm formation, and gene expression. Whereas pure nicotine (N) at the minimum inhibitory concentration (MIC, 4 mg/mL) prevented C. albicans growth, enhanced biofilm formation was observed at 0.1 mg/mL. In contrast, at this nicotine sub-MIC (0.1 mg/mL) concentration, CSC and ECSC had no significant effect on C. albicans biofilm formation. Additionally, N, CSC, and ECSC increased the expression of HWP1 and SAP2 genes. The ECSC group exhibited elevated expression levels of the EAP1 and ALS3 genes, compared to the nicotine-free ECSC (-) control. Moreover, our in vitro study illustrated that the antifungal drugs, fluconazole and amphotericin B, alleviated the effect of nicotine on C. albicans gene expression. Overall, the results of the study indicated nicotine from different sources may affect the pathogenic characteristics of C. albicans, including hyphal growth, biofilm formation, and particularly the expression of virulence-related genes.
Collapse
Affiliation(s)
- Farnoosh Haghighi
- Section of Biosystems and Function, Division of Oral and Systematic Health Sciences, School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095-1668, USA;
| | - Leah Andriasian
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095-1668, USA;
| | - Nini Chaichanasakul Tran
- Section of Pediatric Dentistry, University of California Los Angeles, Los Angeles, CA 90095-1668, USA;
| | - Renate Lux
- Section of Biosystems and Function, Division of Oral and Systematic Health Sciences, School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095-1668, USA;
| |
Collapse
|
15
|
Rashid S, Correia-Mesquita TO, Godoy P, Omran RP, Whiteway M. SAGA Complex Subunits in Candida albicans Differentially Regulate Filamentation, Invasiveness, and Biofilm Formation. Front Cell Infect Microbiol 2022; 12:764711. [PMID: 35350439 PMCID: PMC8957876 DOI: 10.3389/fcimb.2022.764711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/11/2022] [Indexed: 11/24/2022] Open
Abstract
SAGA (Spt-Ada-Gcn5-acetyltransferase) is a highly conserved, multiprotein co-activator complex that consists of five distinct modules. It has two enzymatic functions, a histone acetyltransferase (HAT) and a deubiquitinase (DUB) and plays a central role in processes such as transcription initiation, elongation, protein stability, and telomere maintenance. We analyzed conditional and null mutants of the SAGA complex module components in the fungal pathogen Candida albicans; Ngg1, (the HAT module); Ubp8, (the DUB module); Tra1, (the recruitment module), Spt7, (the architecture module) and Spt8, (the TBP interaction unit), and assessed their roles in a variety of cellular processes. We observed that spt7Δ/Δ and spt8Δ/Δ strains have a filamentous phenotype, and both are highly invasive in yeast growing conditions as compared to the wild type, while ngg1Δ/Δ and ubp8Δ/Δ are in yeast-locked state and non-invasive in both YPD media and filamentous induced conditions compared to wild type. RNA-sequencing-based transcriptional profiling of SAGA mutants reveals upregulation of hyphal specific genes in spt7Δ/Δ and spt8Δ/Δ strains and downregulation of ergosterol metabolism pathway. As well, spt7Δ/Δ and spt8Δ/Δ confer susceptibility to antifungal drugs, to acidic and alkaline pH, to high temperature, and to osmotic, oxidative, cell wall, and DNA damage stresses, indicating that these proteins are important for genotoxic and cellular stress responses. Despite having similar morphological phenotypes (constitutively filamentous and invasive) spt7 and spt8 mutants displayed variation in nuclear distribution where spt7Δ/Δ cells were frequently binucleate and spt8Δ/Δ cells were consistently mononucleate. We also observed that spt7Δ/Δ and spt8Δ/Δ mutants were quickly engulfed by macrophages compared to ngg1Δ/Δ and ubp8Δ/Δ strains. All these findings suggest that the SAGA complex modules can have contrasting functions where loss of Spt7 or Spt8 enhances filamentation and invasiveness while loss of Ngg1 or Ubp8 blocks these processes.
Collapse
Affiliation(s)
| | | | | | | | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
16
|
Yu Z, Wu X, He J. Study on the antifungal activity and mechanism of tea saponin from Camellia oleifera cake. Eur Food Res Technol 2022. [DOI: 10.1007/s00217-021-03929-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
17
|
Kasabe UI, Kale KB, Rode NR, Shelar AV, Patil RH, Mhaske PC, Chaskar MG. Synthesis and antifungal screening of tetramethyl hexahydro-1 H-xanthene-1,8(2 H)-dione derivatives as potential inhibitors of morphogenesis and biofilm formation in Candida albicans. NEW J CHEM 2022; 46:2128-2139. [DOI: 10.1039/d1nj04168a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
A series of hexahydro-1H-xanthene-1,8(2H)-dione derivatives were synthesized. All the derivatives were screened for their anti-virulence properties againstCandida albicans.In silicostudies were performed to corroborate the experimentally observed facts.
Collapse
Affiliation(s)
- Umesh I. Kasabe
- Department of Chemistry, Baburaoji Gholap College (Affiliated to Savitribai Phule Pune University, Pune), Sangvi, Pune-411027, India
| | - Kishor B. Kale
- Department of Chemistry, Nowrosjee Wadia College (Affiliated to Savitribai Phule Pune University, Pune), Pune-411001, India
| | - Nitin R. Rode
- Department of Chemistry, Nowrosjee Wadia College (Affiliated to Savitribai Phule Pune University, Pune), Pune-411001, India
| | - Amruta V. Shelar
- Department of Technology, Savitribai Phule Pune University, Pune-411007, India
| | - Rajendra H. Patil
- Department of Biotechnology, Savitribai Phule Pune University, Pune-411007, India
| | - Pravin C. Mhaske
- Department of Chemistry, Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University, Pune), Pune-411030, India
| | - Manohar G. Chaskar
- Department of Chemistry, Baburaoji Gholap College (Affiliated to Savitribai Phule Pune University, Pune), Sangvi, Pune-411027, India
- Dean, Science and Technology, Savitribai Phule Pune University, Pune-411007, India
| |
Collapse
|
18
|
Kamli MR, Malik MA, Lone SA, Sabir JSM, Mattar EH, Ahmad A. Beta vulgaris Assisted Fabrication of Novel Ag-Cu Bimetallic Nanoparticles for Growth Inhibition and Virulence in Candida albicans. Pharmaceutics 2021; 13:1957. [PMID: 34834372 PMCID: PMC8621205 DOI: 10.3390/pharmaceutics13111957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 01/03/2023] Open
Abstract
Beta vulgaris extract contains water-soluble red pigment betanin and is used as a food colorant. In this study, the biogenic Ag-Cu bimetallic nanoparticles were synthesized and characterized by different spectroscopic and microscopic techniques, including UV-Visible, FTIR, TEM. SEM-EDX, XRD, and TGA. Further, Ag-Cu bimetallic nanoparticles capped with Beta vulgaris biomolecules were evaluated for their antifungal activity against Candida albicans via targeting its major virulence factors, including adherence, yeast to hyphae transition, extracellular enzyme secretion, biofilm formation, and the expression of genes related to these pathogenic traits by using standard methods. C. albicans is an opportunistic human fungal pathogen that causes significant morbidity and mortality, mainly in immunocompromised patients. The current antifungal therapy is limited with various shortcomings such as host toxicity and developing multidrug resistance. Therefore, the development of novel antifungal agents is urgently required. Furthermore, NPs were screened for cell viability and cytotoxicity effect. Antifungal susceptibility testing showed potent antifungal activity of the Ag-Cu bimetallic NPs with a significant inhibitory effect on adherence, yeast to hyphae transition, extracellular enzymes secretion, and formation of biofilms in C. albicans at sub-inhibitory and inhibitory concentrations. The RT-qPCR results at an MIC value of the NPs exhibited a varying degree of downregulation in expression levels of virulence genes. Results also revealed the dose-dependent effect of NPs on cellular viability (up to 100%) using MUSE cell analyzer. Moreover, the low cytotoxicity effect of bimetallic NPs has been observed using haemolytic assay. The overall results indicated that the newly synthesized Ag-Cu bimetallic NPs capped with Beta vulgaris are proven to possess a potent anticandidal activity, by affecting the vital pathogenic factors of C. albicans.
Collapse
Affiliation(s)
- Majid Rasool Kamli
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (J.S.M.S.); (E.H.M.)
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Maqsood Ahmad Malik
- Chemistry Department, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Shabir Ahmad Lone
- Clinical Microbiology and Infectious Diseases, Faculty of Health Sciences, School of Pathology, University of the Witwatersrand, Johannesburg 2193, South Africa; (S.A.L.); (A.A.)
| | - Jamal S. M. Sabir
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (J.S.M.S.); (E.H.M.)
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Ehab H. Mattar
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (J.S.M.S.); (E.H.M.)
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, Faculty of Health Sciences, School of Pathology, University of the Witwatersrand, Johannesburg 2193, South Africa; (S.A.L.); (A.A.)
- Infection Control Unit, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg 2193, South Africa
| |
Collapse
|
19
|
Montoya C, Kurylec J, Baraniya D, Tripathi A, Puri S, Orrego S. Antifungal Effect of Piezoelectric Charges on PMMA Dentures. ACS Biomater Sci Eng 2021; 7:4838-4846. [PMID: 34596379 DOI: 10.1021/acsbiomaterials.1c00926] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Candida-associated denture stomatitis is a recurring disease affecting up to 67% of denture wearers. Poly(methyl methacrylate) (PMMA) remains the main material employed in the fabrication of dentures due to its desirable physical, mechanical, and aesthetic properties. However, the improvement of its antimicrobial properties remains a challenge. To address this need, we developed PMMA composite filled with piezoelectric nanoparticles of barium titanate (BaTiO3) for therapeutic effects. Candida albicans biofilms were cultivated on the surface of the composites under continuous cyclic mechanical loading to activate the piezoelectric charges and to resemble mastication patterns. The interactions between biofilms and biomaterials were evaluated by measuring the biofilm biomass, metabolic activity, and the number of viable cells. To explore the antifungal mechanisms, changes in the expression of genes encoding adhesins and superoxide dismutase were assessed using reverse transcription-polymerase chain reaction. With the addition of piezoelectric nanoparticles, we observed a significant reduction in the biofilm formation and interference in the yeast-to-hyphae transition compared to the standard PMMA. Moreover, we observed that the cyclic deformation of biomaterial surfaces without antifungal agents produced increased biomass, metabolic activity, and a number of viable cells compared to the static/no-deformed surfaces. Cyclic deformation appears to be a novel mechanobiological signal that enables pathogenicity and virulence of C. albicans cells with increased expression of the yeast-to-hyphae transition genes. The outcome of this study opens new opportunities for the design of antifungal dentures for improved clinical service and reduced need for cleaning methods.
Collapse
Affiliation(s)
- Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University. Philadelphia, Pennsylvania 19140, United States
| | - Julia Kurylec
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University. Philadelphia, Pennsylvania 19140, United States
| | - Divyashri Baraniya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University. Philadelphia, Pennsylvania 19140, United States
| | - Aparna Tripathi
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University. Philadelphia, Pennsylvania 19140, United States
| | - Sumant Puri
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University. Philadelphia, Pennsylvania 19140, United States
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University. Philadelphia, Pennsylvania 19140, United States.,Bioengineering Department, College of Engineering, Temple University. Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
20
|
Effects of lipid emulsions on the formation of Escherichia coli-Candida albicans mixed-species biofilms on PVC. Sci Rep 2021; 11:16929. [PMID: 34413406 PMCID: PMC8376934 DOI: 10.1038/s41598-021-96385-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Patients receiving lipid emulsions are at increased risk of contracting catheter-related bloodstream infections (CRBSIs) in the clinic. More than 15% of CRBSIs are polymicrobial. The objective of this study was to explore the effects of lipid emulsions on the formation of Escherichia coli (E. coli)–Candida albicans (C. albicans) mixed-species biofilms (BFs) on polyvinyl chloride (PVC) surfaces and the underlying mechanism. Mixed-species BFs were produced by coculturing E. coli and C. albicans with PVC in various concentrations of lipid emulsions. Crystal violet staining and XTT assays were performed to test the mixed-species BF biomass and the viability of microbes in the BFs. The microstructures of the BFs were observed by an approach that combined confocal laser scanning microscopy, fluorescence in situ hybridization, and scanning electron microscopy. The study found that lipid emulsions could promote the formation of E. coli–C. albicans mixed-species BFs, especially with 10% lipid emulsions. The mechanism by which lipid emulsions promote mixed-species BF formation may involve significant upregulation of the expression of the flhDC, iha, HTA1, and HWP1 genes, which are associated with bacterial motility, adhesion, and BF formation. The results derived from this study necessitate strict aseptic precautions when handling lipid emulsions and avoiding the use of high concentrations of lipid emulsions for as long as possible.
Collapse
|
21
|
Ottaviano E, Baron G, Fumagalli L, Leite J, Colombo EA, Artasensi A, Aldini G, Borghi E. Candida albicans Biofilm Inhibition by Two Vaccinium macrocarpon (Cranberry) Urinary Metabolites: 5-(3',4'-DihydroxyPhenyl)-γ-Valerolactone and 4-Hydroxybenzoic Acid. Microorganisms 2021; 9:microorganisms9071492. [PMID: 34361928 PMCID: PMC8307188 DOI: 10.3390/microorganisms9071492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/22/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022] Open
Abstract
Candida spp. are pathobionts, as they can switch from commensals to pathogens, responsible for a variety of pathological processes. Adhesion to surfaces, morphological switch and biofilm-forming ability are the recognized virulence factors promoting yeast virulence. Sessile lifestyle also favors fungal persistence and antifungal tolerance. In this study, we investigated, in vitro, the efficacy of two urinary cranberry metabolites, 5-(3′,4′-dihydroxy phenyl)-γ-valerolactone (VAL) and 4-hydroxybenzoic acid (4-HBA), in inhibiting C. albicans adhesion and biofilm formation. Both the reference strain SC5314 and clinical isolates were used. We evaluated biomass reduction, by confocal microscopy and crystal violet assay, and the possible mechanisms mediating their inhibitory effects. Both VAL and 4-HBA were able to interfere with the yeast adhesion, by modulating the expression of key genes, HWP1 and ALS3. A significant dose-dependent reduction in biofilm biomass and metabolic activity was also recorded. Our data showed that the two cranberry metabolites VAL and 4-HBA could pave the way for drug development, for targeting the very early phases of biofilm formation and for preventing genitourinary Candida infections.
Collapse
Affiliation(s)
- Emerenziana Ottaviano
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (E.O.); (E.A.C.)
| | - Giovanna Baron
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Laura Fumagalli
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Jessica Leite
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Elisa Adele Colombo
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (E.O.); (E.A.C.)
| | - Angelica Artasensi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (E.O.); (E.A.C.)
- Correspondence: ; Tel.: +39-02-50323287
| |
Collapse
|
22
|
Lee J, Kim Y, Khadke SK, Lee J. Antibiofilm and antifungal activities of medium-chain fatty acids against Candida albicans via mimicking of the quorum-sensing molecule farnesol. Microb Biotechnol 2021; 14:1353-1366. [PMID: 33252828 PMCID: PMC8313291 DOI: 10.1111/1751-7915.13710] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/20/2022] Open
Abstract
Candida biofilms are tolerant to conventional antifungal therapeutics and the host immune system. The transition of yeast cells to hyphae is considered a key step in C. albicans biofilm development, and this transition is inhibited by the quorum-sensing molecule farnesol. We hypothesized that fatty acids mimicking farnesol might influence hyphal and biofilm formation by C. albicans. Among 31 saturated and unsaturated fatty acids, six medium-chain saturated fatty acids, that is, heptanoic acid, octanoic acid, nonanoic acid, decanoic acid, undecanoic acid and lauric acid, effectively inhibited C. albicans biofilm formation by more than 75% at 2 µg ml-1 with MICs in the range 100-200 µg ml-1 . These six fatty acids at 2 µg ml-1 and farnesol at 100 µg ml-1 inhibited hyphal growth and cell aggregation. The addition of fatty acids to C. albicans cultures decreased the productions of farnesol and sterols. Furthermore, down-regulation of several hyphal and biofilm-related genes caused by heptanoic or nonanoic acid closely resembled the changes caused by farnesol. In addition, nonanoic acid, the most effective compound diminished C. albicans virulence in a Caenorhabditis elegans model. Our results suggest that medium-chain fatty acids inhibit more effectively hyphal growth and biofilm formation than farnesol.
Collapse
Affiliation(s)
- Jin‐Hyung Lee
- School of Chemical EngineeringYeungnam UniversityGyeongsanKorea
| | - Yong‐Guy Kim
- School of Chemical EngineeringYeungnam UniversityGyeongsanKorea
| | | | - Jintae Lee
- School of Chemical EngineeringYeungnam UniversityGyeongsanKorea
| |
Collapse
|
23
|
Samot J, Rouabhia M. Effect of Dermaseptin S4 on C. albicans Growth and EAP1 and HWP1 Gene Expression. Probiotics Antimicrob Proteins 2021; 13:287-298. [PMID: 32691243 DOI: 10.1007/s12602-020-09685-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Increasing resistance and changes in the spectrum of Candida infections have generated considerable interest in the development of new antifungal molecules. The use of antimicrobial peptides (AMPs) appears to be a promising approach. Frog skin AMPs (such as dermaseptins) have shown antimicrobial activity against several pathogens. In this study, we aimed to test the antimicrobial efficacy of dermaseptin S4 (DS4) against C. albicans. We determined the minimal inhibitory concentration (MIC) of DS4, and investigated the effects of the DS4 at low concentrations on human primary gingival fibroblasts. Additionally, we evaluated the effect of DS4 on C. albicans growth, form changes, and biofilm formation, as well as the expression of certain virulent genes. Our data show that DS4 completely inhibits C. albicans growth at a concentration of 32 μg/mL referring to the MIC of DS4. It should be noted that even with low concentrations (below 16 μg/mL), DS4 still have significant growth reduction of C. albicans, but were not toxic to human gingival fibroblasts. DS4 inhibited the transition from yeast to hyphae, and decreased the biofilm formation by reducing the biofilm mass weight. Surface morphological changes in the yeast cell membrane were observed following exposure to DS4. The gene expression analyses revealed that DS4 significantly decreased the expression of EAP1 and HWP1 genes. Overall results suggest the potential use of DS4 as an antifungal therapy to prevent C. albicans pathogenesis.
Collapse
Affiliation(s)
- Johan Samot
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC, Canada.,Université de Bordeaux UFR d'odontologie, Bordeaux, France.,Unive. Bordeaux, ISVV, Unité de recherche Œnologie, USC 1366 INRAE, 4577, Villenave d'Ornon, EA, France.,Centre hospitalier universitaire de Bordeaux pôle de médecine et chirurgie bucco-dentaire, Bordeaux, France
| | - Mahmoud Rouabhia
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC, Canada.
| |
Collapse
|
24
|
Yang W, Tu J, Ji C, Li Z, Han G, Liu N, Li J, Sheng C. Discovery of Piperidol Derivatives for Combinational Treatment of Azole-Resistant Candidiasis. ACS Infect Dis 2021; 7:650-660. [PMID: 33593060 DOI: 10.1021/acsinfecdis.0c00849] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Effective strategies are needed to deal with invasive fungal infections caused by drug-resistant fungi. Previously, we designed a series of antifungal benzocyclane derivatives based on the drug repurposing of haloperidol. Herein, further structural optimization and antifungal mechanism studies were performed, leading to the discovery of new piperidol derivative B2 with improved synergistic antifungal potency, selectivity, and water solubility. In particular, the combination of compound B2 and fluconazole showed potent in vitro and in vivo antifungal activity against azole-resistant Candida albicans. Compound B2 inhibited important virulence factors by regulating virulence-associated genes and improved the efficacy of fluconazole by down-regulating the CYP51-coding gene and efflux pump gene. Taken together, the piperidol derivative B2 represents a promising lead compound for the combinational treatment of azole-resistant candidiasis.
Collapse
Affiliation(s)
- Wanzhen Yang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian 350122, China
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Jie Tu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Changjin Ji
- School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zhuang Li
- School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guiyan Han
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Na Liu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Jian Li
- School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Chunquan Sheng
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian 350122, China
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
25
|
Rodríguez-Cerdeira C, Martínez-Herrera E, Carnero-Gregorio M, López-Barcenas A, Fabbrocini G, Fida M, El-Samahy M, González-Cespón JL. Pathogenesis and Clinical Relevance of Candida Biofilms in Vulvovaginal Candidiasis. Front Microbiol 2020; 11:544480. [PMID: 33262741 PMCID: PMC7686049 DOI: 10.3389/fmicb.2020.544480] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/23/2020] [Indexed: 12/30/2022] Open
Abstract
The ability of Candida spp. to form biofilms is crucial for its pathogenicity, and thus, it should be considered an important virulence factor in vulvovaginal candidiasis (VVC) and recurrent VVC (RVVC). Its ability to generate biofilms is multifactorial and is generally believed to depend on the site of infection, species and strain involved, and the microenvironment in which the infection develops. Therefore, both cell surface proteins, such as Hwp1, Als1, and Als2, and the cell wall-related protein, Sun41, play a critical role in the adhesion and virulence of the biofilm. Immunological and pharmacological approaches have identified the NLRP3 inflammasome as a crucial molecular factor contributing to host immunopathology. In this context, we have earlier shown that Candida albicans associated with hyphae-secreted aspartyl proteinases (specifically SAP4-6) contribute to the immunopathology of the disease. Transcriptome profiling has revealed that non-coding transcripts regulate protein synthesis post-transcriptionally, which is important for the growth of Candida spp. Other studies have employed RNA sequencing to identify differences in the 1,245 Candida genes involved in surface and invasive cellular metabolism regulation. In vitro systems allow the simultaneous processing of a large number of samples, making them an ideal screening technique for estimating various physicochemical parameters, testing the activity of antimicrobial agents, and analyzing genes involved in biofilm formation and regulation (in situ) in specific strains. Murine VVC models are used to study C. albicans infection, especially in trials of novel treatments and to understand the cause(s) for resistance to conventional therapeutics. This review on the clinical relevance of Candida biofilms in VVC focuses on important advances in its genomics, transcriptomics, and proteomics. Moreover, recent experiments on the influence of biofilm formation on VVC or RVVC pathogenesis in laboratory animals have been discussed. A clear elucidation of one of the pathogenesis mechanisms employed by Candida biofilms in vulvovaginal candidiasis and its applications in clinical practice represents the most significant contribution of this manuscript.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cerdeira
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Health Research Institute, SERGAS-UVIGO, Vigo, Spain.,Department of Dermatology, Hospital do Meixoeiro and University of Vigo, Vigo, Spain.,European Women's Dermatologic and Venereologic Society, Tui, Spain.,Psychodermatology Task Force of the Ibero-Latin American College of Dermatology (CILAD), Buenos Aires, Argentina
| | - Erick Martínez-Herrera
- Psychodermatology Task Force of the Ibero-Latin American College of Dermatology (CILAD), Buenos Aires, Argentina.,Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Ixtapaluca, Mexico
| | - Miguel Carnero-Gregorio
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Health Research Institute, SERGAS-UVIGO, Vigo, Spain.,Department of Molecular Diagnosis (Array & NGS Division), Institute of Cellular and Molecular Studies, Lugo, Spain
| | - Adriana López-Barcenas
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Psychodermatology Task Force of the Ibero-Latin American College of Dermatology (CILAD), Buenos Aires, Argentina.,Section of Mycology, Department of Dermatology, Manuel Gea González hospital, Mexico City, Mexico
| | - Gabriella Fabbrocini
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Department of Dermatology, University of Naples Federico II, Naples, Italy
| | - Monika Fida
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Department of Dermatology, University of Medicine, Tirana, Tirana, Albania
| | - May El-Samahy
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Department of Dermatology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - José Luís González-Cespón
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Health Research Institute, SERGAS-UVIGO, Vigo, Spain
| |
Collapse
|
26
|
Microglial Response to Aspergillus flavus and Candida albicans: Implications in Endophthalmitis. J Fungi (Basel) 2020; 6:jof6030162. [PMID: 32899547 PMCID: PMC7558867 DOI: 10.3390/jof6030162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022] Open
Abstract
Aspergillus flavus is the most common etiology of fungal endophthalmitis in India, while Candida albicans is the causative agent in the West. In this study, we determined the role of microglial cells in evoking an inflammatory response following an infection with A. flavus and C. albicans strains isolated from patients with endophthalmitis. Microglia (CHME-3) cells were infected with A. flavus and C. albicans and the expression of Toll-Like Receptors (TLRs), cytokines and Matrix metalloproteinases (MMPs) were assessed at various time intervals. A. flavus infected cells induced higher expressions of TLR-1, -2, -5, -6, -7 and -9 and cytokines such as IL-1α, IL-6, IL-8, IL-10 and IL-17. In contrast, C. albicans infected microglia induced only TLR-2 along with the downregulation of IL-10 and IL-17. The expression of MMP-9 (Matrix metalloproteinase-9) was however upregulated in both A. flavus and C. albicans infected microglia. These results indicate that microglial cells have the ability to incite an innate response towards endophthalmitis causing fungal pathogens via TLRs and inflammatory mediators. Moreover, our study highlights the differential responses of microglia towards yeast vs. filamentous fungi.
Collapse
|
27
|
Aaron L, Torsten M. Candida albicans in celiac disease: A wolf in sheep's clothing. Autoimmun Rev 2020; 19:102621. [PMID: 32693029 DOI: 10.1016/j.autrev.2020.102621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022]
Abstract
Candida albicans is a commensal fungus with a potential pathogenicity and celiac disease is an autoimmune condition. Both share multiple pathophysiological junctions, including serological markers against cell-wall proteins of Candida, anti-gliadin antibodies are positive in both entities, gluten and a candidal virulence factor share sequence similarity and the autoantigen of celiac disease, the tissue transglutaminase, is pivotal in Candida albicans commensalism and hostile behavior and its covalently cross linked products are stable and resistant to breakdown in the two entities. Those autoimmune/infectious cross roads are the basis for the hypothesis that Candida albicans is an additional environmental factor for celiac disease autoimmunogenesis.
Collapse
|
28
|
Prasath KG, Tharani H, Kumar MS, Pandian SK. Palmitic Acid Inhibits the Virulence Factors of Candida tropicalis: Biofilms, Cell Surface Hydrophobicity, Ergosterol Biosynthesis, and Enzymatic Activity. Front Microbiol 2020; 11:864. [PMID: 32457728 PMCID: PMC7226919 DOI: 10.3389/fmicb.2020.00864] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/14/2020] [Indexed: 01/13/2023] Open
Abstract
Biofilm is the fortitude of Candida species infections which eventually causes candidiasis in human. C. tropicalis is one of the predominant Candida species commonly found in systemic infections, next to C. albicans. In Candida species, biofilm maturity initiates irreversible surface attachment of cells and barricades the penetration of conventional antifungals. Hence, the current study investigated the antifungal and antivirulence potency of palmitic acid (PA) against C. tropicalis mature biofilm and its associated virulence factors. In vitro results revealed an effective inhibition of biofilm in PA-treated C. tropicalis, compared to C. albicans and C. glabrata. Also, PA reduced C. tropicalis mature biofilm at various time points. Further, PA treatment triggered apoptosis in C. tropicalis through ROS mediated mitochondrial dysfunction as demonstrated by confocal microscopic observation of PI, DAPI and DCFDA staining. PA regulated other virulence factors such as cell surface hydrophobicity, ergosterol biosynthesis, protease and lipase after 48 h of treatment. Downregulation of ERG11 (Lanosterol 14-alpha demethylase) was contributed to the reduction of ergosterol in PA-treated C. tropicalis. However, enhanced hyphal growth was observed in PA-treated C. tropicalis through upregulation HWP1 (Hyphal wall protein) and EFG1 (Enhanced filamentous growth). This study highlighted the antibiofilm and antivirulence potency of PA against C. tropicalis. Hence, PA could be applied synergistically with other antifungal agents to increase the efficacy for regulating NCAC infections.
Collapse
|
29
|
de Barros PP, Rossoni RD, de Souza CM, Scorzoni L, Fenley JDC, Junqueira JC. Candida Biofilms: An Update on Developmental Mechanisms and Therapeutic Challenges. Mycopathologia 2020; 185:415-424. [PMID: 32277380 DOI: 10.1007/s11046-020-00445-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/26/2020] [Indexed: 12/18/2022]
Abstract
Fungi of the genus Candida are important etiological agents of superficial and life-threatening infections in individuals with a compromised immune system. One of the main characteristics of Candida is its ability to form highly drug tolerance biofilms in the human host. Biofilms are a dynamic community of multiple cell types whose formation over time is orchestrated by a network of transcription regulators. In this brief review, we provide an update of the processes involved in biofilm formation by Candida spp. (formation, treatment, and control), as well as the transcriptional circuitry that regulates its development and interactions with other microorganisms. Candida albicans is known to build mixed species biofilms with other Candida species and with various other bacterial species in different host niches. Taken together, these properties play a key role in Candida pathogenesis. In addition, this review gathers recent studies with new insights and perspectives for the treatment and control of Candida biofilms.
Collapse
Affiliation(s)
- Patrícia Pimentel de Barros
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, Avenida Engenheiro Francisco José Longo 777, São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil.
| | - Rodnei Dennis Rossoni
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, Avenida Engenheiro Francisco José Longo 777, São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| | - Cheyenne Marçal de Souza
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, Avenida Engenheiro Francisco José Longo 777, São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| | - Liliana Scorzoni
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, Avenida Engenheiro Francisco José Longo 777, São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| | - Juliana De Camargo Fenley
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, Avenida Engenheiro Francisco José Longo 777, São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, Avenida Engenheiro Francisco José Longo 777, São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| |
Collapse
|
30
|
Araújo D, Azevedo NM, Barbosa A, Almeida C, Rodrigues ME, Henriques M, Silva S. Application of 2'-OMethylRNA' Antisense Oligomer to Control Candida albicans EFG1 Virulence Determinant. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:508-517. [PMID: 31671344 PMCID: PMC6838528 DOI: 10.1016/j.omtn.2019.09.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/26/2019] [Accepted: 09/12/2019] [Indexed: 11/18/2022]
Abstract
Antisense oligomers and their analogs have been successfully utilized to silence gene expression for the treatment of many human diseases; however, the control of yeast’s virulence determinants has never been exploited before. In this sense, this work is based on the key hypothesis that if a pathogen’s genetic sequence is a determinant of virulence, it will be possible to synthesize a nucleic acid mimic based on antisense therapy (AST) that will bind to the mRNA produced, blocking its translation into protein and, consequently, reducing the pathogen virulence phenotype. EFG1 is an important determinant of virulence that is involved in the regulation of the Candida albicans switch from yeast to filamentous form. Thus, our main goal was to design and synthesize an antisense oligonucleotide (ASO) targeting the EFG1 mRNA and to validate its in vitro applicability. The results show that the anti-EFG1 2′-OMethylRNA (2′OMe) oligomer was able to significantly reduce the levels of EFG1 gene expression and of Efg1p protein translation (both approximately 60%), as well as effectively prevent filamentation of C. albicans cells (by 80%). Moreover, it was verified that anti-EFG1 2′OMe keeps the efficacy in different simulated human body fluids. Undeniably, this work provides potentially valuable information for future research into the management of Candida infections, regarding the development of a credible and alternative method to control C. albicans infections, based on AST methodology.
Collapse
Affiliation(s)
- Daniela Araújo
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Nuno Miguel Azevedo
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Ana Barbosa
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Carina Almeida
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; INIAV, IP-National Institute for Agrarian and Veterinary Research, Rua dos Lagidos, Lugar da Madalena, Vairão, 4485-655 Vila do Conde, Portugal
| | - Maria Elisa Rodrigues
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Mariana Henriques
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Sónia Silva
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal.
| |
Collapse
|
31
|
Lee JH, Kim YG, Khadke SK, Yamano A, Watanabe A, Lee J. Inhibition of Biofilm Formation by Candida albicans and Polymicrobial Microorganisms by Nepodin via Hyphal-Growth Suppression. ACS Infect Dis 2019; 5:1177-1187. [PMID: 31055910 DOI: 10.1021/acsinfecdis.9b00033] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Candida albicans is an opportunistic pathogenic yeast and is responsible for candidiasis. It readily colonizes host tissues and implant devices, and forms biofilms, which play an important role in pathogenesis and drug resistance. In this study, the antibiofilm, antihyphal, and antivirulence activities of nepodin, isolated from Rumex japonicus roots, were investigated against a fluconazole-resistant C. albicans strain and against polymicrobial-microorganism-biofilm formation. Nepodin effectively inhibited C. albicans biofilm formation without affecting its planktonic cell growth. Also, Rumex-root extract and nepodin both inhibited hyphal growth and cell aggregation of C. albicans. Interestingly, nepodin also showed antibiofilm activities against Candida glabrata, Candida parapsilosis, Staphylococcus aureus, and Acinetobacter baumannii strains and against dual biofilms of C. albicans and S. aureus or A. baumannii but not against Pseudomonas aeruginosa. Transcriptomic analysis performed by RNA-seq and qRT-PCR showed nepodin repressed the expression of several hypha- and biofilm-related genes (ECE1, HGT10, HWP1, and UME6) and increased the expression of several transport genes (CDR4, CDR11, and TPO2), which supported phenotypic changes. Moreover, nepodin reduced C. albicans virulence in a nematode-infection model and exhibited minimal cytotoxicity against the nematode and an animal cell line. These results demonstrate that nepodin and Rumex-root extract might be useful for controlling C. albicans infections and multispecies biofilms.
Collapse
Affiliation(s)
- Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Sagar Kiran Khadke
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Aki Yamano
- Okinawa Research Center Company, Ltd., 12-75 Ulumasi, Okinawa 904-2234, Japan
| | - Akio Watanabe
- Research Institute for Biological Functions, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
32
|
Raimondi S, Amaretti A, Gozzoli C, Simone M, Righini L, Candeliere F, Brun P, Ardizzoni A, Colombari B, Paulone S, Castagliuolo I, Cavalieri D, Blasi E, Rossi M, Peppoloni S. Longitudinal Survey of Fungi in the Human Gut: ITS Profiling, Phenotyping, and Colonization. Front Microbiol 2019; 10:1575. [PMID: 31354669 PMCID: PMC6636193 DOI: 10.3389/fmicb.2019.01575] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/25/2019] [Indexed: 12/20/2022] Open
Abstract
The fungal component of the intestinal microbiota of eight healthy subjects was studied over 12 months using metagenome survey and culture-based approaches. Aspergillus, Candida, Debaryomyces, Malassezia, Penicillium, Pichia, and Saccharomyces were the most recurrent and/or dominant fungal genera, according to metagenomic analysis. The biodiversity of fungal communities was lower and characterized by greater unevenness, when compared to bacterial microbiome. The dissimilarities both among subjects and over the time within the same subject suggested that most of the fungi passed through the gastro-intestinal tract (GIT) without becoming stable colonizers. Certain genera, such as Aspergillus and Penicillium, were isolated in a minority of cases, although they recurred abundantly and frequently in the metagenomics survey, likely being environmental or food-borne fungi that do not inhabit the GIT. Candida genus was recurrently detected. Candida albicans isolates dominated among the cultivable mycobiota and longitudinally persisted, likely as commensals inhabiting the intestine or regularly reaching it from Candida-colonized districts, such as the oral cavity. Other putative colonizers belonged to Candida zeylanoides, Geotrichum candidum, and Rhodotorula mucilaginosa, with persisting biotypes being identified. Phenotyping of fungal isolates indicated that C. albicans adhered to human epithelial cells more efficiently and produced greater amounts of biofilm in vitro than non-albicans Candida (NAC) and non-Candida fungi (NCF). The C. albicans isolates also induced the highest release of HBD-2 by human epithelial cells, further differing from NAC and NCF. Nine representative isolates were administered to mice to evaluate the ability to colonize the intestine. Only two out of three C. albicans strains persisted in stools of animals 2 weeks after the end of the oral administration, whereas NAC and NCF did not. These results confirm the allochthonous nature of most the intestinal fungi, while C. albicans appears to be commonly involved in stable colonization. A combination of specific genetic features in the microbe and in the host likely allow colonization from fungi normally present solely as passengers. It remains to be established if other species identified as potential colonizers, in addition to Candida, are true inhabitants of the GIT or rather reach the intestine spreading from other body districts.
Collapse
Affiliation(s)
- Stefano Raimondi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto Amaretti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Caterina Gozzoli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marta Simone
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Righini
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Candeliere
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Brun
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Andrea Ardizzoni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Bruna Colombari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Paulone
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Elisabetta Blasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Maddalena Rossi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Samuele Peppoloni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
33
|
Pakshir K, Sheykhi S, Zomorodian K, Nouraei H, Zare Shahrabadi Z. Evaluation of biofilm formation in the homozygous and heterozygous strains of vaginal Candida albicans isolates. Curr Med Mycol 2019; 5:37-40. [PMID: 31321337 PMCID: PMC6626708 DOI: 10.18502/cmm.5.2.1160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background and Purpose Candida albicans is one of the most opportunistic yeasts around the world. This species has two heterozygous and homozygous strains at hyphal wall protein 1 (hwp1) gene locus. A simple method for the discrimination of these two strains is the amplification of HWP1 gene. Regarding this, the aim of this study was to discriminate C. albicans heterozygous and homozygous strains via the amplification of hwp1 gene and evaluation of biofilm formation between the strains. Materials and Methods A total of 60 homozygous (n=30) and heterozygous (n=30) strains were discriminated among 126 C. albicans vaginal isolates by the amplification of HWP1 gene, using specific primers. The evaluation of biofilm formation was accomplished using the visual method. Results According to the results, the homozygous and heterozygous strains produced one and two DNA fragments, respectively. The frequency of homozygous strains among the C. albicans vaginal isolates was 76.2%. Biofilm formation activity in the heterozygous strains was more than that in the homozygous strains. However, statistical analysis showed no significant difference between the strains in terms of biofilm formation. Conclusion As the findings indicated, the frequency of the heterozygous strains in C. albicans was lower than that of the homozygous strains. Both of the strains could form biofilm in the different ranges of severity. High activity of biofilm formation in heterozygous strains may set the ground for its pathogenicity.
Collapse
Affiliation(s)
- Keyvan Pakshir
- Department of Parasitology and Mycology, Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Sheykhi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamiar Zomorodian
- Department of Parasitology and Mycology, Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hasti Nouraei
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Zare Shahrabadi
- Department of Parasitology and Mycology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Vaginal lactobacilli inhibit growth and hyphae formation of Candida albicans. Sci Rep 2019; 9:8121. [PMID: 31148560 PMCID: PMC6544633 DOI: 10.1038/s41598-019-44579-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Lactobacillus species are the predominant vaginal microbiota found in healthy women of reproductive age and help to prevent pathogen infection by producing lactic acid, H2O2 and anti-microbial compounds. Identification of novel vaginal Lactobacillus isolates that exhibit efficient colonisation and secrete anti-Candida factors is a promising strategy to prevent vulvovaginal candidiasis. The azole antifungal agents used to treat vulvovaginal candidiasis elicit adverse effects such as allergic responses and exhibit drug interactions. Candida strains with resistance to antifungal treatments are often reported. In this study, we isolated Lactobacillus species from healthy Korean women and investigated their antifungal effects against C. albicans in vitro and in vivo. Lactobacillus conditioned supernatant (LCS) of L. crispatus and L. fermentum inhibited C. albicans growth in vitro. A Lactobacillus-derived compound, which was not affected by proteolytic enzyme digestion and heat inactivation, inhibited growth and hyphal induction of C. albicans after adjustment to neutral pH. Combination treatment with neutral LCSs of L. crispatus and L. fermentum effectively inhibited propagation of C. albicans in a murine in vivo model of vulvovaginal candidiasis.
Collapse
|
35
|
Candida spp. and phagocytosis: multiple evasion mechanisms. Antonie van Leeuwenhoek 2019; 112:1409-1423. [PMID: 31079344 DOI: 10.1007/s10482-019-01271-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/02/2019] [Indexed: 01/01/2023]
Abstract
Invasive fungal infections are a global health problem, mainly in hospitals, where year by year hundreds of patients die because of these infections. Commensal yeasts may become pathogenic to human beings, affecting mainly immunocompromised patients. During infectious processes, the immune system uses phagocytes to eliminate invader microorganisms. In order to prevent or neutralize phagocyte attacks, pathogenic yeasts can use virulence factors to survive, as well as to colonize and infect the host. In this review, we describe how Candida spp., mainly Candida albicans, interact with phagocytes and use several factors that contribute to immune evasion. Polymorphism, biofilm formation, gene expression and enzyme production mediate distinct functions such as adhesion, invasion, oxidative stress response, proteolysis and escape from phagocytes. Fungal and human cells have similar structures and mechanisms that decrease the number of potential targets for antifungal drugs. Therefore, research on host-pathogen interaction may aid in the discovery of new targets and in the development of new drugs or treatments for these diseases and thus to save lives.
Collapse
|
36
|
Rodríguez-Cerdeira C, Gregorio MC, Molares-Vila A, López-Barcenas A, Fabbrocini G, Bardhi B, Sinani A, Sánchez-Blanco E, Arenas-Guzmán R, Hernandez-Castro R. Biofilms and vulvovaginal candidiasis. Colloids Surf B Biointerfaces 2018; 174:110-125. [PMID: 30447520 DOI: 10.1016/j.colsurfb.2018.11.011] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/04/2018] [Accepted: 11/05/2018] [Indexed: 01/18/2023]
Abstract
Candida species, including C. albicans, are part of the mucosal flora of most healthy women, and inhabit the gastrointestinal and genitourinary tracts. Under favourable conditions, they can colonize the vulvovaginal mucosa, giving rise to symptomatic vulvovaginal candidiasis (VVC). The mechanism by which Candida spp. produces inflammation is unknown. Both, the blastoconidia and the pseudohyphae are capable of destroying the vaginal epithelium by direct invasion. Although the symptoms are not always related to the fungal burden, in general, VVC is associated with a greater number of yeasts and pseudohyphae. Some years ago, C. albicans was the species most frequently involved in the different forms of VVC. However, infections by different species have emerged during the last two decades producing an increase in causative species of VVC such as C. glabrata, C. parapsilosis, C. krusei and C. tropicalis. Candida species are pathogenic organisms that have two forms of development: planktonic and biofilm. A biofilm is defined as a community of microorganisms attached to a surface and encompassed by an extracellular matrix. This form of presentation gives microorganisms greater resistance to antifungal agents. This review, about Candia spp. with a special emphasis on Candida albicans discusses specific areas such as biofilm structure and development, cell morphology and biofilm formation, biofilm-associated gene expression, the cell surface and adherence, the extracellular matrix, biofilm metabolism, and biofilm drug resistance in vulvovaginitis biofilms as an important virulence factor in fungi.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cerdeira
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Dermatology Department, Hospital do Meixoeiro and University of Vigo, Vigo, Spain; European Women's Dermatologic and Venereologic Society (EWDVS), Vigo, Spain.
| | - Miguel Carnero Gregorio
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain
| | - Alberto Molares-Vila
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Department of Department of Analytical & Food Chemistry, Universidade de Vigo (UVIGO), Spain
| | - Adriana López-Barcenas
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Mycology Service, Hospital Manuel Gea González, Mexico City, Mexico
| | | | | | - Ardiana Sinani
- Dermatology Service, Military Medical Unit, University Trauma Hospital, Tirana, Albania
| | | | | | | |
Collapse
|
37
|
Willaert RG. Adhesins of Yeasts: Protein Structure and Interactions. J Fungi (Basel) 2018; 4:jof4040119. [PMID: 30373267 PMCID: PMC6308950 DOI: 10.3390/jof4040119] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
The ability of yeast cells to adhere to other cells or substrates is crucial for many yeasts. The budding yeast Saccharomyces cerevisiae can switch from a unicellular lifestyle to a multicellular one. A crucial step in multicellular lifestyle adaptation is self-recognition, self-interaction, and adhesion to abiotic surfaces. Infectious yeast diseases such as candidiasis are initiated by the adhesion of the yeast cells to host cells. Adhesion is accomplished by adhesin proteins that are attached to the cell wall and stick out to interact with other cells or substrates. Protein structures give detailed insights into the molecular mechanism of adhesin-ligand interaction. Currently, only the structures of a very limited number of N-terminal adhesion domains of adhesins have been solved. Therefore, this review focuses on these adhesin protein families. The protein architectures, protein structures, and ligand interactions of the flocculation protein family of S. cerevisiae; the epithelial adhesion family of C. glabrata; and the agglutinin-like sequence protein family of C. albicans are reviewed and discussed.
Collapse
Affiliation(s)
- Ronnie G Willaert
- Alliance Research Group VUB-UGent NanoMicrobiology (NAMI), IJRG VUB-EPFL NanoBiotechnology & NanoMedicine (NANO), Research Group Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium.
- Department Bioscience Engineering, University Antwerp, 2020 Antwerp, Belgium.
| |
Collapse
|
38
|
Shibasaki S, Karasaki M, Aoki W, Ueda M. Molecular and Physiological Study of Candida albicans by Quantitative Proteome Analysis. Proteomes 2018; 6:proteomes6030034. [PMID: 30231513 PMCID: PMC6160938 DOI: 10.3390/proteomes6030034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/28/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022] Open
Abstract
Candida albicans is one of the major pathogens that cause the serious infectious condition known as candidiasis. C. albicans was investigated by proteome analysis to systematically examine its virulence factors and to promote the development of novel pharmaceuticals against candidiasis. Here, we review quantitative time-course proteomics data related to C. albicans adaptation to fetal bovine serum, which were obtained using a nano-liquid chromatography/tandem mass spectrometry system equipped with a long monolithic silica capillary column. It was revealed that C. albicans induced proteins involved in iron acquisition, detoxification of oxidative species, energy production, and pleiotropic stress tolerance. Native interactions of C. albicans with macrophages were also investigated with the same proteome-analysis system. Simultaneous analysis of C. albicans and macrophages without isolating individual living cells revealed an attractive strategy for studying the survival of C. albicans. Although those data were obtained by performing proteome analyses, the molecular physiology of C. albicans is discussed and trials related to pharmaceutical applications are also examined.
Collapse
Affiliation(s)
- Seiji Shibasaki
- General Education Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan.
| | - Miki Karasaki
- General Education Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan.
| | - Wataru Aoki
- Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Mitsuyoshi Ueda
- Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
39
|
Lee JH, Kim YG, Choi P, Ham J, Park JG, Lee J. Antibiofilm and Antivirulence Activities of 6-Gingerol and 6-Shogaol Against Candida albicans Due to Hyphal Inhibition. Front Cell Infect Microbiol 2018; 8:299. [PMID: 30211127 PMCID: PMC6121036 DOI: 10.3389/fcimb.2018.00299] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/07/2018] [Indexed: 11/17/2022] Open
Abstract
Candida albicans is an opportunistic pathogen and responsible for candidiasis. C. albicans readily forms biofilms on various biotic and abiotic surfaces, and these biofilms can cause local and systemic infections. C. albicans biofilms are more resistant than its free yeast to antifungal agents and less affected by host immune responses. Transition of yeast cells to hyphal cells is required for biofilm formation and is believed to be a crucial virulence factor. In this study, six components of ginger were investigated for antibiofilm and antivirulence activities against a fluconazole-resistant C. albicans strain. It was found 6-gingerol, 8-gingerol, and 6-shogaol effectively inhibited biofilm formation. In particular, 6-shogaol at 10 μg/ml significantly reduced C. albicans biofilm formation but had no effect on planktonic cell growth. Also, 6-gingerol and 6-shogaol inhibited hyphal growth in embedded colonies and free-living planktonic cells, and prevented cell aggregation. Furthermore, 6-gingerol and 6-shogaol reduced C. albicans virulence in a nematode infection model without causing toxicity at the tested concentrations. Transcriptomic analysis using RNA-seq and qRT-PCR showed 6-gingerol and 6-shogaol induced several transporters (CDR1, CDR2, and RTA3), but repressed the expressions of several hypha/biofilm related genes (ECE1 and HWP1), which supported observed phenotypic changes. These results highlight the antibiofilm and antivirulence activities of the ginger components, 6-gingerol and 6-shogaol, against a drug resistant C. albicans strain.
Collapse
Affiliation(s)
- Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Pilju Choi
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, South Korea
| | - Jungyeob Ham
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, South Korea
| | - Jae Gyu Park
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang, South Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
40
|
Farisa Banu S, Rubini D, Shanmugavelan P, Murugan R, Gowrishankar S, Karutha Pandian S, Nithyanand P. Effects of patchouli and cinnamon essential oils on biofilm and hyphae formation by Candida species. J Mycol Med 2018; 28:332-339. [PMID: 29571979 DOI: 10.1016/j.mycmed.2018.02.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/24/2018] [Accepted: 02/24/2018] [Indexed: 12/01/2022]
Abstract
The prevalence and fatality rates with biofilm-associated candidal infections have remained a challenge to the medical fraternity despite major advances in the field of antifungal therapy. Traditionally, essential oils (EOs) from the aromatic plants have been found to be excellent therapeutic agents to treat fungal ailments. The present study explores the antivirulent and antibiofilm effects of under explored leaf EOs of Indian patchouli EO extracted from Pogostemon heyneanus (PH), Indian cassia from Cinnamomum tamala (CT) and camphor EO from C. camphora (CC) against Candida species. The EOs were investigated for its efficacy to disrupt the young and preformed Candida spp. biofilms and to inhibit the yeast to hyphal transition, a hallmark virulent trait of C. albicans. The ability of these EOs to inhibit metabolically active cells was assessed through XTT assay. Of these three EOs, CT EO showed enhanced biofilm inhibition than others and hence it was further selected to study its biomass inhibition potential and exopolysaccharide layer disruption ability. The CT EO reduced the biomass of the preformed biofilms of all three Candida strains, which was supported by confocal microscopy. It also disrupted the exopolysaccharide layer of the Candida strains as shown by scanning electron microscopy. The present findings validate the effectiveness of EOs against the virulence of Candida spp. and emphasize the pharmaceutical potential of several native but yet unexplored wild aromatic plants in the prospect of therapeutic application.
Collapse
Affiliation(s)
- S Farisa Banu
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, Anusandhan Kendra II, SASTRA University, 613401 Thanjavur, Tamil Nadu, India
| | - D Rubini
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, Anusandhan Kendra II, SASTRA University, 613401 Thanjavur, Tamil Nadu, India
| | - P Shanmugavelan
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, Anusandhan Kendra II, SASTRA University, 613401 Thanjavur, Tamil Nadu, India
| | - R Murugan
- School of Chemical and Biotechnology, SASTRA University, 613401 Thanjavur, Tamil Nadu, India
| | - S Gowrishankar
- Department of Biotechnology, Science Campus, Alagappa University, 630003 Karaikudi, Tamil Nadu, India
| | - S Karutha Pandian
- Department of Biotechnology, Science Campus, Alagappa University, 630003 Karaikudi, Tamil Nadu, India
| | - P Nithyanand
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, Anusandhan Kendra II, SASTRA University, 613401 Thanjavur, Tamil Nadu, India; Centre for Research on Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA University, 613401 Thanjavur, Tamil Nadu, India.
| |
Collapse
|
41
|
Cavalieri D, Di Paola M, Rizzetto L, Tocci N, De Filippo C, Lionetti P, Ardizzoni A, Colombari B, Paulone S, Gut IG, Berná L, Gut M, Blanc J, Kapushesky M, Pericolini E, Blasi E, Peppoloni S. Genomic and Phenotypic Variation in Morphogenetic Networks of Two Candida albicans Isolates Subtends Their Different Pathogenic Potential. Front Immunol 2018; 8:1997. [PMID: 29403478 PMCID: PMC5780349 DOI: 10.3389/fimmu.2017.01997] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/22/2017] [Indexed: 01/29/2023] Open
Abstract
The transition from commensalism to pathogenicity of Candida albicans reflects both the host inability to mount specific immune responses and the microorganism’s dimorphic switch efficiency. In this study, we used whole genome sequencing and microarray analysis to investigate the genomic determinants of the phenotypic changes observed in two C. albicans clinical isolates (YL1 and YQ2). In vitro experiments employing epithelial, microglial, and peripheral blood mononuclear cells were thus used to evaluate C. albicans isolates interaction with first line host defenses, measuring adhesion, susceptibility to phagocytosis, and induction of secretory responses. Moreover, a murine model of peritoneal infection was used to compare the in vivo pathogenic potential of the two isolates. Genome sequence and gene expression analysis of C. albicans YL1 and YQ2 showed significant changes in cellular pathways involved in environmental stress response, adhesion, filamentous growth, invasiveness, and dimorphic transition. This was in accordance with the observed marked phenotypic differences in biofilm production, dimorphic switch efficiency, cell adhesion, invasion, and survival to phagocyte-mediated host defenses. The mutations in key regulators of the hyphal growth pathway in the more virulent strain corresponded to an overall greater number of budding yeast cells released. Compared to YQ2, YL1 consistently showed enhanced pathogenic potential, since in vitro, it was less susceptible to ingestion by phagocytic cells and more efficient in invading epithelial cells, while in vivo YL1 was more effective than YQ2 in recruiting inflammatory cells, eliciting IL-1β response and eluding phagocytic cells. Overall, these results indicate an unexpected isolate-specific variation in pathways important for host invasion and colonization, showing how the genetic background of C. albicans may greatly affect its behavior both in vitro and in vivo. Based on this approach, we propose that the co-occurrence of changes in sequence and expression in genes and pathways driving dimorphic transition and pathogenicity reflects a selective balance between traits favoring dissemination of the pathogen and traits involved in host defense evasion. This study highlights the importance of investigating strain-level, rather than species level, differences, when determining fungal–host interactions and defining commensal or pathogen behavior.
Collapse
Affiliation(s)
- Duccio Cavalieri
- Dipartimento di Biologia, Università di Firenze, Florence, Italy
| | - Monica Di Paola
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Università di Firenze, Florence, Italy
| | - Lisa Rizzetto
- Centro Ricerca e Innovazione, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Noemi Tocci
- Centro Ricerca e Innovazione, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Carlotta De Filippo
- Institute of Agricultural Biology and Biotechnology, National Research Council (CNR), Pisa, Italy
| | - Paolo Lionetti
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Università di Firenze, Florence, Italy
| | - Andrea Ardizzoni
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia, Modena, Italy
| | - Bruna Colombari
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia, Modena, Italy
| | - Simona Paulone
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia, Modena, Italy
| | - Ivo G Gut
- Centro Nacional de Anàlisi Genòmica, Barcelona, Spain
| | - Luisa Berná
- Unidad de Biologia Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Marta Gut
- Centro Nacional de Anàlisi Genòmica, Barcelona, Spain
| | - Julie Blanc
- Centro Nacional de Anàlisi Genòmica, Barcelona, Spain
| | - Misha Kapushesky
- European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | - Eva Pericolini
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia, Modena, Italy
| | - Elisabetta Blasi
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia, Modena, Italy
| | - Samuele Peppoloni
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia, Modena, Italy
| |
Collapse
|
42
|
Mail MH, Himratul-Aznita WH, Musa MY. Anti-hyphal properties of potential bioactive compounds for oral rinse in suppression of Candidagrowth. BIOTECHNOL BIOTEC EQ 2017. [DOI: 10.1080/13102818.2017.1348255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Mohd Hafiz Mail
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Wan Harun Himratul-Aznita
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Md Yusoff Musa
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Manoharan RK, Lee JH, Lee J. Antibiofilm and Antihyphal Activities of Cedar Leaf Essential Oil, Camphor, and Fenchone Derivatives against Candida albicans. Front Microbiol 2017; 8:1476. [PMID: 28824600 PMCID: PMC5541024 DOI: 10.3389/fmicb.2017.01476] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/21/2017] [Indexed: 01/09/2023] Open
Abstract
Candida albicans can form biofilms composed of yeast, hyphal, and pseudohyphal elements, and C. albicans cells in the hyphal stage could be a virulence factor. The present study describes the chemical composition, antibiofilm, and antihyphal activities of cedar leaf essential oil (CLEO), which was found to possess remarkable antibiofilm activity against C. albicans but not to affect its planktonic cell growth. Nineteen components were identified in CLEO by gas chromatography/mass spectrometry, and phenolics were the main constituents. Of these, camphor, fenchone, fenchyl alcohol, α-thujone, and borneol significantly reduced C. albicans biofilm formation. Notably, treatments with CLEO, camphor, or fenchyl alcohol at 0.01% clearly inhibited hyphal formation, and this inhibition appeared to be largely responsible for their antibiofilm effects. Transcriptomic analyses indicated that camphor and fenchyl alcohol downregulated some hypha-specific and biofilm related genes (ECE1, ECE2, RBT1, and EED1). Furthermore, camphor and fenchyl alcohol reduced C. albicans virulence in a Caenorhabditis elegans nematode model. These results demonstrate CLEO, camphor, and fenchyl alcohol might be useful for controlling C. albicans infections.
Collapse
Affiliation(s)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam UniversityGyeongsan, South Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam UniversityGyeongsan, South Korea
| |
Collapse
|
44
|
Wang S, Wang Q, Yang E, Yan L, Li T, Zhuang H. Antimicrobial Compounds Produced by Vaginal Lactobacillus crispatus Are Able to Strongly Inhibit Candida albicans Growth, Hyphal Formation and Regulate Virulence-related Gene Expressions. Front Microbiol 2017; 8:564. [PMID: 28421058 PMCID: PMC5378977 DOI: 10.3389/fmicb.2017.00564] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/20/2017] [Indexed: 01/09/2023] Open
Abstract
The female vaginal environment contains diverse microorganisms, and their interactions play significant roles in health and disease. Lactobacillus species are the predominant vaginal microorganisms in healthy women and relevant as a barrier to defense against pathogens, including Candida albicans. The yeast-to-hyphae transition is believed to be a determinant of C. albicans pathogenesis. In this study, we investigated the effects of vaginal isolates of L. crispatus (seven strains), L. gasseri (six strains), and L. jensenii (five strains) on growth, hyphal formation and virulence-related genes expression of C. albicans ATCC 10231. We found that the L. crispatus showed the most significant antimicrobial activities in microplate-based liquid medium assay (P < 0.05). All seven cell-free supernatants (CFS) from L. crispatus strains reduced the growth of C. albicans by >60%. The effects might be due to their productions of some secretory antimicrobial compounds in addition to H2O2 and organic acids. Furthermore, each of the CFS of Lactobacillus strains was found to significantly suppress the yeast-to-hyphae transition of C. albicans under hyphae-inducing conditions (RPMI 1640 medium supplemented with 10% fetal bovine serum). The hyphae inhibition rates of C. albicans treated by CFS from L. crispatus, L. gasseri, and L. jensenii were 88.3 ± 3.02%, 84.9 ± 6.0%, and 81.9 ± 6.2%, respectively. Moreover, the expression of hyphae-specific genes (ALS3, HWP1, ECE1, EAP1, and SAP5) and transcriptional regulatory genes (EFG1, TEC1, and NRG1) were analyzed using quantitative real-time PCR. The results demonstrated that L. crispatus CFS significantly down-regulated the expression of hyphae-specific genes ALS3 (0.140-fold)), HWP1 (0.075-fold), and ECE1 (0.045-fold), while up-regulated the expression of the negative transcriptional regulator gene NRG1 with 1.911-fold. The antimicrobial compounds from L. crispatus B145 against Candida growth were heat stable and protease resistance, but those against hyphal formation were partially sensitive to the same treatments. Our novel findings suggest that L. crispatus, a dominant Lactobacillus species associated with a healthy vagina, could strongly inhibit C. albicans growth and hyphal formation. L. crispatus might repress the expression of hyphae-specific genes (ALS3, HWP1, and ECE1) in a NRG1-dependent manner. Besides, L. crispatus B145 is highly worthwhile for probiotic investigation.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science CenterBeijing, China
| | - Qiangyi Wang
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science CenterBeijing, China
| | - Ence Yang
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science CenterBeijing, China
| | - Ling Yan
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science CenterBeijing, China
| | - Tong Li
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science CenterBeijing, China
| | - Hui Zhuang
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science CenterBeijing, China
| |
Collapse
|
45
|
Tong Y, Tang J. Candida albicans infection and intestinal immunity. Microbiol Res 2017; 198:27-35. [PMID: 28285659 DOI: 10.1016/j.micres.2017.02.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/27/2016] [Accepted: 02/09/2017] [Indexed: 12/22/2022]
Abstract
Fungal infections cause high rates of morbidity and mortality in intensive care and immunocompromised patients, and can represent a life-threatening disease. As a microorganism commonly found in the intestine, Candida albicans (C. albicans) can invade the gut epithelium barrier via microfold cells and enter the bloodstream. The defensive potential of the intestinal barrier against invasive C. albicans is dependent on innate and adaptive immune responses which enable the host to eliminate pathogenic fungi. The lamina propria layer of the intestine contains numerous immune cells capable of inducing an innate cellular immune response against invasive fungi. This review focuses on the immune response triggered by a C. albicans infection in the intestine.
Collapse
Affiliation(s)
- Yiqing Tong
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University; Shanghai 200240, PR China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University; Shanghai 200240, PR China.
| |
Collapse
|
46
|
Candida krusei and Candida glabrata reduce the filamentation of Candida albicans by downregulating expression of HWP1 gene. Folia Microbiol (Praha) 2017; 62:317-323. [DOI: 10.1007/s12223-017-0500-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/17/2017] [Indexed: 10/20/2022]
|
47
|
Li X, Yu C, Huang X, Sun S. Synergistic Effects and Mechanisms of Budesonide in Combination with Fluconazole against Resistant Candida albicans. PLoS One 2016; 11:e0168936. [PMID: 28006028 PMCID: PMC5179115 DOI: 10.1371/journal.pone.0168936] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/08/2016] [Indexed: 11/21/2022] Open
Abstract
Candida albicans is an important opportunistic pathogen, causing both superficial mucosal infections and life-threatening systemic diseases in the clinic. The emergence of drug resistance in Candida albicans has become a noteworthy phenomenon due to the extensive use of antifungal agents and the development of biofilms. This study showed that budesonide potentiates the antifungal effect of fluconazole against fluconazole-resistant Candida albicans strains both in vitro and in vivo. In addition, our results demonstrated, for the first time, that the combination of fluconazole and budesonide can reverse the resistance of Candida albicans by inhibiting the function of drug transporters, reducing the formation of biofilms, promoting apoptosis and inhibiting the activity of extracellular phospholipases. This is the first study implicating the effects and mechanisms of budesonide against Candida albicans alone or in combination with fluconazole, which may ultimately lead to the identification of new potential antifungal targets.
Collapse
Affiliation(s)
- Xiuyun Li
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province, People’s Republic of China
| | - Cuixiang Yu
- Respiration Medicine, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Province, People’s Republic of China
| | - Xin Huang
- Pharmaceutical Department, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Province, People’s Republic of China
| | - Shujuan Sun
- Pharmaceutical Department, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Province, People’s Republic of China
- * E-mail:
| |
Collapse
|
48
|
Koutsouras GW, Ramos RL, Martinez LR. Role of microglia in fungal infections of the central nervous system. Virulence 2016; 8:705-718. [PMID: 27858519 DOI: 10.1080/21505594.2016.1261789] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Most fungi are capable of disseminating into the central nervous system (CNS) commonly being observed in immunocompromised hosts. Microglia play a critical role in responding to these infections regulating inflammatory processes proficient at controlling CNS colonization by these eukaryotic microorganisms. Nonetheless, it is this inflammatory state that paradoxically yields cerebral mycotic meningoencephalitis and abscess formation. As peripheral macrophages and fungi have been investigated aiding our understanding of peripheral disease, ascertaining the key interactions between fungi and microglia may uncover greater abilities to treat invasive fungal infections of the brain. Here, we present the current knowledge of microglial physiology. Due to the existing literature, we have described to greater extent the opportunistic mycotic interactions with these surveillance cells of the CNS, highlighting the need for greater efforts to study other cerebral fungal infections such as those caused by geographically restricted dimorphic and rare fungi.
Collapse
Affiliation(s)
- George W Koutsouras
- a Department of Biomedical Sciences , NYIT College of Osteopathic Medicine, New York Institute of Technology , Old Westbury , NY , USA
| | - Raddy L Ramos
- a Department of Biomedical Sciences , NYIT College of Osteopathic Medicine, New York Institute of Technology , Old Westbury , NY , USA
| | - Luis R Martinez
- a Department of Biomedical Sciences , NYIT College of Osteopathic Medicine, New York Institute of Technology , Old Westbury , NY , USA
| |
Collapse
|
49
|
Araújo D, Henriques M, Silva S. Portrait of Candida Species Biofilm Regulatory Network Genes. Trends Microbiol 2016; 25:62-75. [PMID: 27717660 DOI: 10.1016/j.tim.2016.09.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/01/2016] [Accepted: 09/15/2016] [Indexed: 11/15/2022]
Abstract
Most cases of candidiasis have been attributed to Candida albicans, but Candida glabrata, Candida parapsilosis and Candida tropicalis, designated as non-C. albicans Candida (NCAC), have been identified as frequent human pathogens. Moreover, Candida biofilms are an escalating clinical problem associated with significant rates of mortality. Biofilms have distinct developmental phases, including adhesion/colonisation, maturation and dispersal, controlled by complex regulatory networks. This review discusses recent advances regarding Candida species biofilm regulatory network genes, which are key components for candidiasis.
Collapse
Affiliation(s)
- Daniela Araújo
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Mariana Henriques
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Sónia Silva
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal.
| |
Collapse
|
50
|
Salivary pellicles equalise surfaces’ charges and modulate the virulence of Candida albicans biofilm. Arch Oral Biol 2016; 66:129-40. [DOI: 10.1016/j.archoralbio.2016.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/01/2015] [Accepted: 02/25/2016] [Indexed: 02/06/2023]
|