1
|
Shahina Z, Dahms TES. A Comparative Review of Eugenol and Citral Anticandidal Mechanisms: Partners in Crimes Against Fungi. Molecules 2024; 29:5536. [PMID: 39683696 DOI: 10.3390/molecules29235536] [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: 10/23/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Candida albicans is an emerging multidrug-resistant opportunistic pathogen that causes candidiasis, superficial infections on the mucosa, nails or skin, and life-threatening candidemia in deep tissue when disseminated through the bloodstream. Recently, there has been a sharp rise in resistant strains, posing a considerable clinical challenge for the treatment of candidiasis. There has been a resurged interest in the pharmacological properties of essential oils and their active components, for example, monoterpenes with alcohol (-OH) and aldehyde (-CHO) groups. Eugenol and citral have shown promising in vitro and in vivo activity against Candida species. Although there is substantial research on the efficacy of these essential oil components against C. albicans, a detailed knowledge of their mycological mechanisms is lacking. To explore the broad-spectrum effects of EOs, it is more meaningful and rational to study the whole essential oil, along with some of its major components. This review provides a comprehensive overview of eugenol and citral anticandidal and antivirulence activity, alone and together, along with the associated mechanisms and limitations of our current knowledge.
Collapse
Affiliation(s)
- Zinnat Shahina
- Department of Chemistry and Biochemistry, University of Regina, 3737 Wascana Parkway, Regina, SK S4S 0A2, Canada
| | - Tanya E S Dahms
- Department of Chemistry and Biochemistry, University of Regina, 3737 Wascana Parkway, Regina, SK S4S 0A2, Canada
| |
Collapse
|
2
|
Fong JL, Ong Eng Yong V, Yeo C, Adamson C, Li L, Zhang D, Qiao Y. Biochemical Characterization of Recombinant Enterococcus faecalis EntV Peptide to Elucidate Its Antihyphal and Antifungal Mechanisms against Candida albicans. ACS Infect Dis 2024; 10:3408-3418. [PMID: 39137394 DOI: 10.1021/acsinfecdis.4c00515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Candida albicans is a common opportunistic fungus in humans, whose morphological switch between yeast and hyphae forms represents a key virulence trait. Developing strategies to inhibit C. albicans hyphal growth may provide insights into designs of novel antivirulent therapeutics. Importantly, the gut commensal bacterium, Enterococcus faecalis, secretes a bacteriocin EntV which has potent antivirulent and antifungal effects against C. albicans in infection models; however, hampered by the challenges to access large quantities of bioactive EntV, the detailed understanding of its mechanisms on C. albicans has remained elusive. In this work, we biochemically reconstituted the proteolytic cleavage reaction to obtain recombinant EntV88-His6 on a large preparative scale, providing facile access to the C-terminal EntV construct. Under in vitro C. albicans hyphal assay with specific inducers, we demonstrated that EntV88-His6 exhibits potent bioactivity against GlcNAc-triggered hyphal growth. Moreover, with fluorescent FITC-EntV88-His6, we revealed that EntV88-His6 enters C. albicans via endocytosis and perturbs the proper localization of the polarisome scaffolding Spa2 protein. Our findings provide important clues on EntV's mechanism of action. Surprisingly, we showed that EntV88-His6 does not affect C. albicans yeast cell growth but potently exerts cytotoxicity against C. albicans under hyphal-inducing conditions in vitro. The combination of EntV88-His6 and GlcNAc displays rapid killing of C. albicans, rendering it a promising antivirulent and antifungal agent.
Collapse
Affiliation(s)
- Jia Li Fong
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Victor Ong Eng Yong
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore
| | - Claresta Yeo
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Christopher Adamson
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Lanxin Li
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Dan Zhang
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore
| | - Yuan Qiao
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
3
|
Yiu B, Robbins N, Cowen LE. Interdisciplinary approaches for the discovery of novel antifungals. Trends Mol Med 2024; 30:723-735. [PMID: 38777733 DOI: 10.1016/j.molmed.2024.04.018] [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: 02/16/2024] [Revised: 04/10/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Pathogenic fungi are an increasing public health concern. The emergence of antifungal resistance coupled with the scarce antifungal arsenal highlights the need for novel therapeutics. Fortunately, the past few years have witnessed breakthroughs in antifungal development. Here, we discuss pivotal interdisciplinary approaches for the discovery of novel compounds with efficacy against diverse fungal pathogens. We highlight breakthroughs in improving current antifungal scaffolds, as well as the utility of compound combinations to extend the lifespan of antifungals. Finally, we describe efforts to refine candidate chemical scaffolds by leveraging structure-guided approaches, and the use of functional genomics to expand our knowledge of druggable antifungal targets. Overall, we emphasize the importance of interdisciplinary collaborations in the endeavor to develop innovative antifungal strategies.
Collapse
Affiliation(s)
- Bonnie Yiu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada.
| |
Collapse
|
4
|
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
|
5
|
Zhou X, Chen X, Pan Q, Wang S, Li J, Yang Y. Exploring the role of candidalysin in the pathogenicity of Candida albicans by gene set enrichment analysis and evolutionary dynamics. Am J Transl Res 2024; 16:3191-3210. [PMID: 39114682 PMCID: PMC11301511 DOI: 10.62347/izym9087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/20/2024] [Indexed: 08/10/2024]
Abstract
AIMS To explore the pathogenic mechanisms of Candida albicans (C. albicans), focusing on its impact on human health, particularly through invasive infections in the gastrointestinal and respiratory tracts. METHODS In this study, we evaluated the demographic and clinical profiles of 7 pneumonia patients. Meanwhile, we used Gene Set Enrichment Analysis (GSEA) and Evolutionary Dynamics method to analyze the role of candidalysin in C. albicans pathogenicity. RESULTS By analyzing genomic data and conducting biomedical text mining, we identified novel mutation sites in the candidalysin coding gene ECE1-III, shedding light into the genetic diversity within C. albicans strains and their potential implications for antifungal resistance. Our results revealed significant associations between C. albicans and respiratory as well as gastrointestinal diseases, emphasizing the fungus's role in the pathogenesis of these diseases. Additionally, we identified a new mutation site in the C. albicans strain YF2-5, isolated from patients with pneumonia. This mutation may be associated with its heightened pathogenicity. CONCLUSION Our research advances the understanding of C. albicans pathogenicity and opens new avenues for developing targeted antifungal therapies. By focusing on the molecular basis of fungal virulence, we aim to contribute to the development of more effective treatment strategies, addressing the challenge of multidrug resistance in invasive fungal infections.
Collapse
Affiliation(s)
- Xingchen Zhou
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious DiseasesBeijing 100850, China
| | - Xiaolin Chen
- Sir Run Run Hospital, Nanjing Medical UniversityNanjing 210009, Jiangsu, China
| | - Qianglong Pan
- Sir Run Run Hospital, Nanjing Medical UniversityNanjing 210009, Jiangsu, China
| | - Shengqi Wang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious DiseasesBeijing 100850, China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical UniversityNanjing 210009, Jiangsu, China
| | - Ying Yang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious DiseasesBeijing 100850, China
| |
Collapse
|
6
|
Wang X, Jin X, Zhao F, Xu Z, Tan W, Zhang J, Xu Y, Luan X, Fang M, Xie Z, Chang W, Lou H. Structure-Based Optimization of Novel Sterol 24-C-Methyltransferase Inhibitors for the Treatment of Candida albicans Infections. J Med Chem 2024; 67:9318-9341. [PMID: 38764175 DOI: 10.1021/acs.jmedchem.4c00470] [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: 05/21/2024]
Abstract
Interfering with sterol biosynthesis is an important strategy for developing safe and effective antifungal drugs. We previously identified compound H55 as an allosteric inhibitor of the fungal-specific C-24 sterol methyltransferase Erg6 for treating Candida albicans infections. Herein, 62 derivatives of H55 were designed and synthesized based on target-ligand interactions to identify more active candidates. Among them, d28 displayed the most potent antivirulence ability (MHIC50 = 0.25 μg/mL) by targeting Erg6, exhibiting an 8-fold increase in potency compared with H55. Moreover, d28 significantly outperformed H55 in inhibiting cell adhesion and biofilm formation, and exhibited minimal cytotoxicity and negligible potential to induce drug resistance. Of note, the coadministration of d28 and other sterol biosynthesis inhibitors, such as tridemorph or terbinafine, demonstrated a strong synergistic antifungal action in vitro and in vivo in a murine skin infection model. These results support the potential application of d28 in the treatment of C. albicans infections.
Collapse
Affiliation(s)
- Xue Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xueyang Jin
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zejun Xu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenzhuo Tan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiaozhen Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuliang Xu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China
| | - Xiaoyi Luan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Min Fang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhiyu Xie
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang 461002, China
| | - Wenqiang Chang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
7
|
Yang W, Liu R, Li Z, Tu J, Xu D, Liu N, Sheng C. Discovery of New Tricyclic Oxime Sampangine Derivatives as Potent Antifungal Agents for the Treatment of Cryptococcosis and Candidiasis. J Med Chem 2024. [PMID: 38489247 DOI: 10.1021/acs.jmedchem.3c02331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Cryptococcus neoformans (C. neoformans) and Candida albicans (C. albicans) are classified as the critical priority groups among the pathogenic fungi, highlighting the urgent need for developing more effective antifungal therapies. On the basis of antifungal natural product sampangine, herein, a series of tricyclic oxime and oxime ether derivatives were designed. Among them, compound WZ-2 showed excellent inhibitory activity against C. neoformans (MIC80 = 0.016 μg/mL) and synergized with fluconazole to treat resistant C. albicans (FICI = 0.078). Interestingly, compound WZ-2 effectively inhibited virulence factors (e.g., capsule, biofilm, and yeast-to-hypha morphological transition), suggesting the potential to overcome drug resistance. In a mouse model of cryptococcal meningitis, compound WZ-2 (5 mg/kg) effectively reduced the brain C. neoformans H99 burden. Furthermore, compound WZ-2 alone and its combination with fluconazole also significantly reduced the kidney burden of the drug-resistant strain (0304103) and sensitive strain (SC5314) of C. albicans.
Collapse
Affiliation(s)
- 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
| | - Ruxiong 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
| | - Zhuang Li
- 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
| | - Dongjian Xu
- 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
| | - 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
|
8
|
Song S, Zhao S, Sun X, Meng L, Wang Z, Tan H, Liu J, Zhang M, Deng Y. Anti-virulence strategy of diaryl chalcogenide compounds against Candida albicans infection. Virulence 2023; 14:2265012. [PMID: 37771181 PMCID: PMC10549196 DOI: 10.1080/21505594.2023.2265012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 09/25/2023] [Indexed: 09/30/2023] Open
Abstract
Candida albicans is an important opportunistic pathogenic fungus that frequently causes serious systemic infection in humans. Due to the vital roles of biofilm formation and the yeast-to-hypha transition in the infection process, we have selected a series of diaryl chalcogenides and tested their efficacy against C. albicans SC5314 pathogenicity by the inhibition of biofilm formation and the yeast-to-hypha transition. The compounds 5-sulfenylindole and 5-selenylindole were found to have excellent abilities to inhibit both biofilm formation and hyphal formation in C. albicans SC5314. Intriguingly, the two leading compounds also markedly attenuated C. albicans SC5314 virulence in human cell lines and mouse infection models at micromolar levels. Furthermore, our results showed that the presence of the compounds at 100 µM resulted in a marked decrease in the expression of genes involved in the cAMP-PKA and MAPK pathways in C. albicans SC5314. Intriguingly, the compounds 5-sulfenylindole and 5-selenylindole not only attenuated the cytotoxicity of Candida species strains but also showed excellent synergistic effects with antifungal agents against the clinical drug-resistant C. albicans strain HCH12. The compound 5-sulfenylindole showed an obvious advantage over fluconazole as it could also restore the composition and richness of the intestinal microbiota in mice infected by C. albicans. Together, these results suggest that diaryl chalcogenides can potentially be designed as novel clinical therapeutic agents against C. albicans infection. The diaryl chalcogenides of 5-sulfenylindole and 5-selenylindole discovered in this study can provide new direction for developing antifungal agents against C. albicans infection.
Collapse
Affiliation(s)
- Shihao Song
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shuo Zhao
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Xiuyun Sun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Lili Meng
- Integrative Microbiology Research Center, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Zijie Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Hunan Children’s Hospital, Changsha, China
| | - Huihui Tan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Jingyun Liu
- Integrative Microbiology Research Center, College of Plant Protection, South China Agricultural University, Guangzhou, China
- Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Min Zhang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, China
| | - Yinyue Deng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
9
|
Fang T, Xiong J, Wang L, Feng Z, Hang S, Yu J, Li W, Feng Y, Lu H, Jiang Y. Unexpected Inhibitory Effect of Octenidine Dihydrochloride on Candida albicans Filamentation by Impairing Ergosterol Biosynthesis and Disrupting Cell Membrane Integrity. Antibiotics (Basel) 2023; 12:1675. [PMID: 38136708 PMCID: PMC10741164 DOI: 10.3390/antibiotics12121675] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Candida albicans filamentation plays a significant role in developing both mucosal and invasive candidiasis, making it a crucial virulence factor. Consequently, exploring and identifying inhibitors that impede fungal hyphal formation presents an intriguing approach toward antifungal strategies. In line with this anti-filamentation strategy, we conducted a comprehensive screening of a library of FDA-approved drugs to identify compounds that possess inhibitory properties against hyphal growth. The compound octenidine dihydrochloride (OCT) exhibits potent inhibition of hyphal growth in C. albicans across different hyphae-inducing media at concentrations below or equal to 3.125 μM. This remarkable inhibitory effect extends to biofilm formation and the disruption of mature biofilm. The mechanism underlying OCT's inhibition of hyphal growth is likely attributed to its capacity to impede ergosterol biosynthesis and induce the generation of reactive oxygen species (ROS), compromising the integrity of the cell membrane. Furthermore, it has been observed that OCT demonstrates protective attributes against invasive candidiasis in Galleria mellonella larvae through its proficient eradication of C. albicans colonization in infected G. mellonella larvae by impeding hyphal formation. Although additional investigation is required to mitigate the toxicity of OCT in mammals, it possesses considerable promise as a potent filamentation inhibitor against invasive candidiasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
10
|
Kalimuthu S, Pudipeddi A, Braś G, Tanner JA, Rapala-Kozik M, Leung YY, Neelakantan P. A heptadeca amino acid peptide subunit of cathelicidin LL-37 has previously unreported antifungal activity. APMIS 2023; 131:584-600. [PMID: 37150907 DOI: 10.1111/apm.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023]
Abstract
Yeasts such as Candida albicans, albeit being ubiquitous members of the skin, oral and vaginal microbiome, can cause superficial to life-threatening infections. Human cathelicidin LL-37-based peptides have antibacterial activity and yet, their antifungal activity remains to be thoroughly characterized. The aim of this study was to comprehensively investigate the activity of LL-37-based peptides against C. albicans. LL-37 and its derivatives were tested for their ability to kill C. albicans planktonic cells in the presence of various biological matrices (serum, plasma, saliva and urine), that have been reported to inactivate peptides. The antibiofilm activity, resistance development and biocompatibility were investigated for the lead peptide. GK-17, a 17 amino acid peptide, showed remarkable stability to fungal aspartyl proteases and rapidly killed planktonic C. albicans despite the presence of biological matrices. GK-17 also inhibited adhesion to biotic and abiotic substrates, inhibited biofilm formation and eradicated preformed biofilms in the presence of biological matrices. Compared to nystatin, GK-17 had a lower propensity to allow for resistance development by C. albicans. The peptide showed concentration-dependent biocompatibility to red blood cells, with only 30% hemolysis even at 4× the fungicidal concentration. Taken together, GK-17 is a novel antifungal peptide with promising effects against C. albicans.
Collapse
Affiliation(s)
- Shanthini Kalimuthu
- Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong SAR
| | - Akhila Pudipeddi
- Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong SAR
| | - Grażyna Braś
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Julian A Tanner
- School of Biomedical Sciences, The University of Hong Kong, Sai Ying Pun, Hong Kong SAR
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Yiu Yan Leung
- Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong SAR
| | | |
Collapse
|
11
|
Sushmitha TJ, Rajeev M, Kathirkaman V, Shivam S, Rao TS, Pandian SK. 3-Hydroxy coumarin demonstrates anti-biofilm and anti-hyphal efficacy against Candida albicans via inhibition of cell-adhesion, morphogenesis, and virulent genes regulation. Sci Rep 2023; 13:11687. [PMID: 37468600 DOI: 10.1038/s41598-023-37851-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023] Open
Abstract
Candida albicans, a common fungus of human flora, can become an opportunistic pathogen and causes invasive candidiasis in immunocompromised individuals. Biofilm formation is the prime cause of antibiotic resistance during C. albicans infections and treating biofilm-forming cells is challenging due to their intractable and persistent nature. The study intends to explore the therapeutic potential of naturally produced compounds by competitive marine bacteria residing in marine biofilms against C. albicans biofilm. To this end, 3-hydroxy coumarin (3HC), a compound identified from the cell-free culture supernatant of the marine bacterium Brevundimonas abyssalis, was found to exhibit anti-biofilm and anti-hyphal activity against both reference and clinical isolates of C. albicans. The compound demonstrated significant inhibitory effects on biofilms and impaired the yeast-to-hyphal transition, wrinkle, and filament morphology at the minimal biofilm inhibitory concentration (MBIC) of 250 µg mL-1. Intriguingly, quantitative PCR analysis of 3HC-treated C. albicans biofilm revealed significant downregulation of virulence genes (hst7, ume6, efg1, cph1, ras1, als1) associated with adhesion and morphogenesis. Moreover, 3HC displayed non-fungicidal and non-toxic characteristics against human erythrocytes and buccal cells. In conclusion, this study showed that marine biofilms are a hidden source of diverse therapeutic drugs, and 3HC could be a potent drug to treat C. albicans infections.
Collapse
Affiliation(s)
- T J Sushmitha
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Meora Rajeev
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
- Department of Biological Sciences and Bioengineering, Inha University, Inharo 100, Incheon, 22212, Republic of Korea
| | - Vellaisamy Kathirkaman
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Singh Shivam
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Toleti Subba Rao
- School of Arts and Sciences, Sai University, OMR, Paiyanur, Tamil Nadu, 603105, India
| | - Shunmugiah Karutha Pandian
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India.
| |
Collapse
|
12
|
Zeng H, Stadler M, Abraham WR, Müsken M, Schrey H. Inhibitory Effects of the Fungal Pigment Rubiginosin C on Hyphal and Biofilm Formation in Candida albicans and Candida auris. J Fungi (Basel) 2023; 9:726. [PMID: 37504715 PMCID: PMC10381533 DOI: 10.3390/jof9070726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023] Open
Abstract
The two fungal human pathogens, Candida auris and Candida albicans, possess a variety of virulence mechanisms. Among them are the formation of biofilms to protect yeast against harsh conditions through the development of (pseudo)hyphae whilst also facilitating the invasion of host tissues. In recent years, increased rates of antifungal resistance have been associated with C. albicans and C. auris, posing a significant challenge for the effective treatment of fungal infections. In the course of our ongoing search for novel anti-infectives, six selected azaphilones were tested for their cytotoxicity and antimicrobial effects as well as for their inhibitory activity against biofilm and hyphal formation. This study revealed that rubiginosin C, derived from stromata of the ascomycete Hypoxylon rubiginosum, effectively inhibited the formation of biofilms, pseudohyphae, and hyphae in both C. auris and C. albicans without lethal effects. Crystal violet staining assays were utilized to assess the inhibition of biofilm formation, while complementary microscopic techniques, such as confocal laser scanning microscopy, scanning electron microscopy, and optical microscopy, were used to investigate the underlying mechanisms. Rubiginosin C is one of the few substances known to effectively target both biofilm formation and the yeast-to-hyphae transition of C. albicans and C. auris within a concentration range not affecting host cells, making it a promising candidate for therapeutic intervention in the future.
Collapse
Affiliation(s)
- Haoxuan Zeng
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Marc Stadler
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Wolf-Rainer Abraham
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Hedda Schrey
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| |
Collapse
|
13
|
MacAlpine J, Robbins N, Cowen LE. Bacterial-fungal interactions and their impact on microbial pathogenesis. Mol Ecol 2023; 32:2565-2581. [PMID: 35231147 PMCID: PMC11032213 DOI: 10.1111/mec.16411] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
Microbial communities of the human microbiota exhibit diverse effects on human health and disease. Microbial homeostasis is important for normal physiological functions and changes to the microbiota are associated with many human diseases including diabetes, cancer, and colitis. In addition, there are many microorganisms that are either commensal or acquired from environmental reservoirs that can cause diverse pathologies. Importantly, the balance between health and disease is intricately connected to how members of the microbiota interact and affect one another's growth and pathogenicity. However, the mechanisms that govern these interactions are only beginning to be understood. In this review, we outline bacterial-fungal interactions in the human body, including examining the mechanisms by which bacteria govern fungal growth and virulence, as well as how fungi regulate bacterial pathogenesis. We summarize advances in the understanding of chemical, physical, and protein-based interactions, and their role in exacerbating or impeding human disease. We focus on the three fungal species responsible for the majority of systemic fungal infections in humans: Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. We conclude by summarizing recent studies that have mined microbes for novel antimicrobials and antivirulence factors, highlighting the potential of the human microbiota as a rich resource for small molecule discovery.
Collapse
Affiliation(s)
- Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
14
|
Baghiat Esfahani M, Khodavandi A, Alizadeh F, Bahador N. Biofilm-associated genes as potential molecular targets of nano-Fe 3O 4 in Candida albicans. Pharmacol Rep 2023; 75:682-694. [PMID: 36930446 DOI: 10.1007/s43440-023-00467-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND There are few effective treatments for Candida biofilm-associated infections. The present study demonstrated changes in the expression of biofilm-associated genes in Candida albicans treated with magnetic iron oxide nanoparticles (denoted as nano-Fe3O4). METHODS Nano-Fe3O4 was biologically synthesized using Bacillus licheniformis, Bacillus cereus, and Fusarium oxysporum. Additionally, the biologically synthesized nano-Fe3O4 was characterized by visual observation; ultraviolet-visible spectroscopy, scanning electron microscopy, X-ray diffraction spectroscopy, and Fourier transform infrared spectroscopy. The biologically synthesized nano-Fe3O4 was tested for growth and biofilm formation in C. albicans. Furthermore, quantitative real-time reverse transcriptase-polymerase chain reaction (RT-PCR) was used to study the inhibition of biofilm-associated genes in C. albicans treated with nano-Fe3O4. RESULTS The production of biologically synthesized nano-Fe3O4 was confirmed using extensive characterization methods. The nano-Fe3O4 inhibited growth and biofilm formation. Nano-Fe3O4 exhibited growth inhibition with minimum inhibition concentrations (MICs) of 50 to 200 μg mL-1. The anti-biofilm effects of nano-Fe3O4 were shown by 2,3-bis (2-methoxy-4-nitro-5 sulfophenyl)-5-[(phenylamino) carbonyl]-2H-tetrazolium hydroxide (XTT) reduction assay, crystal violet staining, and light field microscopy. The gene expression results showed that the downregulation of BCR1, ALS1, ALS3, HWP1, and ECE1 genes inhibited the biofilm formation in C. albicans. ALS1 reduction was greater than others, with downregulation of 1375.83-, 1178.71-, and 768.47-fold at 2 × MIC, 1 × MIC, and ½ × MIC of nano-Fe3O4, respectively. CONCLUSION Biofilm-associated genes as potential molecular targets of nano-Fe3O4 in C. albicans may be an effective novel treatment strategy for biofilm-associated infections.
Collapse
Affiliation(s)
| | - Alireza Khodavandi
- Department of Biology, Gachsaran Branch, Islamic Azad University, Gachsaran, Iran.
| | - Fahimeh Alizadeh
- Department of Biology, Gachsaran Branch, Islamic Azad University, Gachsaran, Iran
| | - Nima Bahador
- Department of Microbiology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| |
Collapse
|
15
|
Candida albicans Reactive Oxygen Species (ROS)-Dependent Lethality and ROS-Independent Hyphal and Biofilm Inhibition by Eugenol and Citral. Microbiol Spectr 2022; 10:e0318322. [PMID: 36394350 PMCID: PMC9769929 DOI: 10.1128/spectrum.03183-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Candida albicans is part of the normal human flora but is most frequently isolated as the causative opportunistic pathogen of candidiasis. Plant-based essential oils and their components have been extensively studied as antimicrobials, but their antimicrobial impacts are poorly understood. Phenylpropenoids and monoterpenes, for example, eugenol from clove and citral from lemon grass, are potent antifungals against a wide range of pathogens. We report the cellular response of C. albicans to eugenol and citral, alone and combined, using biochemical and microscopic assays. The MICs of eugenol and citral were 1,000 and 256 μg/mL, respectively, with the two exhibiting additive effects based on a fractional inhibitory concentration index of 0.83 ± 0.14. High concentrations of eugenol caused membrane damage, oxidative stress, vacuole segregation, microtubule dysfunction and cell cycle arrest at the G1/S phase, and while citral had similar impacts, they were reactive oxygen species (ROS) independent. At sublethal concentrations (1/2 to 1/4 MIC), both oils disrupted microtubules and hyphal and biofilm formation in an ROS-independent manner. While both compounds disrupt the cell membrane, eugenol had a greater impact on membrane dysfunction. This study shows that eugenol and citral can induce vacuole and microtubule dysfunction, along with the inhibition of hyphal and biofilm formation. IMPORTANCE Candida albicans is a normal resident on and in the human body that can cause relatively benign infections. However, when our immune system is severely compromised (e.g., cancer chemotherapy patients) or underdeveloped (e.g., newborns), this fungus can become a deadly pathogen, infecting the bloodstream and organs. Since there are only a few effective antifungal agents that can be used to combat fungal infections, these fungi have been exposed to them over and over again, allowing the fungi to develop resistance. Instead of developing antifungal agents that kill the fungi, some of which have undesirable side effects on the human host, researchers have proposed to target the fungal traits that make the fungus more virulent. Here, we show how two components of plant-based essential oils, eugenol and citral, are effective inhibitors of C. albicans virulence traits.
Collapse
|
16
|
Fan F, Liu Y, Liu Y, Lv R, Sun W, Ding W, Cai Y, Li W, Liu X, Qu W. Candida albicans biofilms: antifungal resistance, immune evasion, and emerging therapeutic strategies. Int J Antimicrob Agents 2022; 60:106673. [PMID: 36103915 DOI: 10.1016/j.ijantimicag.2022.106673] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Candida albicans is a fungal pathogen that can form biofilms on medical devices and host tissue, resulting in serious, life-threatening infections. These fungal biofilms are inherently resistant to traditional antifungal therapies and the host immune system; therefore, biofilm-associated infections are a huge clinical challenge. This review summarizes the most important insights into C. albicans biofilm-associated antifungal drug resistance mechanisms and immune evasion strategies. In addtion, this review also discusses the strategies for antifungal drug use to combat these processes, providing further evidence for novel drugs research and clinical therapies.
Collapse
Affiliation(s)
- FangMei Fan
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Yi Liu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - YiQing Liu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - RuiXue Lv
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Wei Sun
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - WenJing Ding
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - YanXing Cai
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - WeiWei Li
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Xing Liu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Wei Qu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China.
| |
Collapse
|
17
|
Memariani M, Memariani H, Poursafavi Z, Baseri Z. Anti-fungal Effects and Mechanisms of Action of Wasp Venom-Derived Peptide Mastoparan-VT1 Against Candida albicans. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Synergistic Antibiofilm Effects of Pseudolaric Acid A Combined with Fluconazole against Candida albicans via Inhibition of Adhesion and Yeast-To-Hypha Transition. Microbiol Spectr 2022; 10:e0147821. [PMID: 35297651 PMCID: PMC9045105 DOI: 10.1128/spectrum.01478-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Candida albicans biofilms are resistant to several clinical antifungal agents. Thus, it is necessary to develop new antibiofilm intervention measures. Pseudolaric acid A (PAA), a diterpenoid mainly derived from the pine bark of Pseudolarix kaempferi, has been reported to have an inhibitory effect on C. albicans. The primary aim of the current study was to investigate the antibiofilm effect of PAA when combined with fluconazole (FLC) and explore the underlying mechanisms. Biofilm activity was assessed by tetrazolium {XTT [2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide salt]} reduction assays. PAA (4 μg/mL) combined with FLC (0.5 μg/mL) significantly inhibited early, developmental, and mature biofilm formation compared with the effect of PAA or FLC alone (P < 0.05). Furthermore, PAA (4 μg/mL) combined with FLC (0.5 μg/mL) produced a 56% reduction in C. albicans biofilm adhesion. The combination of PAA (4 μg/mL) and FLC (0.5 μg/mL) also performed well in inhibiting yeast-to-hypha transition. Transcriptome analysis using RNA sequencing and quantitative reverse transcription PCR indicated that the PAA-FLC combination treatment produced a strong synergistic inhibitory effect on the expression of genes involved in adhesion (ALS1, ALS4, and ALS2) and yeast-to-hypha transition (ECE1, PRA1, and TEC1). Notably, PAA, rather than FLC, may have a primary role in suppressing the expression of ALS1. In conclusion, these findings demonstrate, for the first time, that the combination of PAA and FLC has an improved antibiofilm effect against the formation of C. albicans biofilms by inhibiting adhesion and yeast-to-hypha transition; this may provide a novel therapeutic strategy for treating C. albicans biofilm-associated infection. IMPORTANCE Biofilms are the primary cause of antibiotic-resistant candida infections associated with medical implants and devices worldwide. Treating biofilm-associated infections is a challenge for clinicians because these infections are intractable and persistent. Candida albicans readily forms extensive biofilms on the surface of medical implants and mucosa. In this study, we demonstrated, for the first time, an inhibitory effect of pseudolaric acid A alone and in combination with fluconazole on C. albicans biofilms. Moreover, pseudolaric acid A in combination with fluconazole exerted an antibiofilm effect through multiple pathways, including inhibition of yeast-to-hypha transition and adhesion. This research not only provides new insights into the synergistic mechanisms of antifungal drug combinations but also brings new possibilities for addressing C. albicans drug resistance.
Collapse
|
19
|
Kalimuthu S, Alshanta OA, Krishnamoorthy AL, Pudipeddi A, Solomon AP, McLean W, Leung YY, Ramage G, Neelakantan P. Small molecule based anti-virulence approaches against Candida albicans infections. Crit Rev Microbiol 2022; 48:743-769. [PMID: 35232325 DOI: 10.1080/1040841x.2021.2025337] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Fungi are considered "silent killers" due to the difficulty of, and delays in diagnosis of infections and lack of effective antifungals. This challenge is compounded by the fact that being eukaryotes, fungi share several similarities with human cellular targets, creating obstacles to drug discovery. Candida albicans, a ubiquitous microbe in the human body is well-known for its role as an opportunistic pathogen in immunosuppressed people. Significantly, C. albicans is resistant to all the three classes of antifungals that are currently clinically available. Over the past few years, a paradigm shift has been recommended in the management of C. albicans infections, wherein anti-virulence strategies are considered an alternative to the discovery of new antimycotics. Small molecules, with a molecular weight <900 Daltons, can easily permeate the cell membrane and modulate the signal transduction pathways to elicit desired virulence inhibitory actions against pathogens. This review dissects in-depth, the discoveries that have been made with small-molecule anti-virulence approaches to tackle C. albicans infections.
Collapse
Affiliation(s)
| | - Om Alkhir Alshanta
- Glasgow Endodontology Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | - Akshaya Lakshmi Krishnamoorthy
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.,Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Akhila Pudipeddi
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - William McLean
- Glasgow Endodontology Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | - Yiu Yan Leung
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Gordon Ramage
- Glasgow Endodontology Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | | |
Collapse
|
20
|
Chen Z, Luo T, Huang F, Yang F, Luo W, Chen G, Cao M, Wang F, Zhang J. Kangbainian Lotion Ameliorates Vulvovaginal Candidiasis in Mice by Inhibiting the Growth of Fluconazole-Resistant Candida albicans and the Dectin-1 Signaling Pathway Activation. Front Pharmacol 2022; 12:816290. [PMID: 35140608 PMCID: PMC8819624 DOI: 10.3389/fphar.2021.816290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/27/2021] [Indexed: 11/18/2022] Open
Abstract
Vulvovaginal candidiasis (VVC) is an infectious disease caused by Candida species, which affects millions of women worldwide every year. The resistance to available antifungal drugs for clinical treatment is a growing problem. The treatment of refractory VVC caused by azole-resistant Candida is still facing challenges. However, research on new antifungal drugs is progressing slowly. Although a lot of reports on new antifungal drugs, only three new antifungal drugs (Isavuconazole, ibrexafungerp, and rezafungin) and two new formulations of posaconazole were marketed over the last decade. Chinese botanical medicine has advantages in the treatment of drug-resistant VVC, such as outstanding curative effects and low adverse reactions, which can improve patients’ comfort and adherence to therapy. Kangbainian lotion (KBN), a Chinese botanical formulation, has achieved very good clinical effects in the treatment of VVC. In this study, we investigated the antifungal and anti-inflammatory effects of KBN at different doses in fluconazole-resistant (FLC-resistant) VVC model mice. We further studied the antifungal mechanism of KBN against FLC-resistant Candida albicans (C. albicans) and the anti-inflammatory mechanism correlated with the Dectin-1 signaling pathway. In vivo and in vitro results showed that KBN had strong antifungal and anti-inflammatory effects in FLC-resistant VVC, such as inhibiting the growth of C. albicans and vaginal inflammation. Further studies showed that KBN inhibited the biofilm and hypha formation, reduced adhesion, inhibited ergosterol synthesis and the expression of ergosterol synthesis-related genes ERG11, and reduced the expression of drug-resistant efflux pump genes MDR1 and CDR2 of FLC-resistant C. albicans in vitro. In addition, in vivo results showed that KBN reduced the expression of inflammatory factor proteins TNF-α, IL-1β, and IL-6 in vaginal tissues, and inhibited the expression of proteins related to the Dectin-1 signaling pathway. In conclusion, our study revealed that KBN could ameliorate vaginal inflammation in VVC mice caused by FLC-resistance C. albicans. This effect may be related to inhibiting the growth of FLC-resistance C. albicans and Dectin-1 signaling pathway activation.
Collapse
Affiliation(s)
- Zewei Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tengshuo Luo
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Fengke Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuzhen Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenting Luo
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guanfeng Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengfei Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengyun Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Jun Zhang, ; Fengyun Wang,
| | - Jun Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Jun Zhang, ; Fengyun Wang,
| |
Collapse
|
21
|
Khona DK, Roy S, Ghatak S, Huang K, Jagdale G, Baker LA, Sen CK. Ketoconazole resistant Candida albicans is sensitive to a wireless electroceutical wound care dressing. Bioelectrochemistry 2021; 142:107921. [PMID: 34419917 PMCID: PMC8788813 DOI: 10.1016/j.bioelechem.2021.107921] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/27/2021] [Accepted: 07/31/2021] [Indexed: 01/22/2023]
Abstract
Wireless electroceutical dressing (WED) fabric kills bacteria and disrupts bacterial biofilm. This work tested, comparing with standard of care topical antibiotic ketoconazole, whether the weak electric field generated by WED is effective to manage infection caused by ketoconazole-resistant yeast Candida albicans. WED inhibited Candida albicans biofilm formation and planktonic growth. Unlike ketoconazole, WED inhibited yeast to hyphal transition and downregulated EAP1 curbing cell attachment. In response to WED-dependent down-regulation of biofilm-forming BRG1 and ROB1, BCR1 expression was markedly induced in what seems to be a futile compensatory response. WED induced NRG1 and TUP1, negative regulators of filamentation; it down-regulated EFG1, a positive regulator of hyphal pathway. Consistent with the anti-hyphal properties of WED, the expression of ALS3 and HWP1 were diminished. Ketoconazole failed to reproduce the effects of WED on NRG1, TUP1 and EFG1. WED blunted efflux pump activity; this effect was in direct contrast to that of ketoconazole. WED exposure compromised cellular metabolism. In the presence of ketoconazole, the effect was synergistic. Unlike ketoconazole, WED caused membrane depolarization, changes in cell wall composition and loss of membrane integrity. This work presents first evidence that weak electric field is useful in managing pathogens which are otherwise known to be antibiotic resistant.
Collapse
Affiliation(s)
- Dolly K Khona
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Subhadip Ghatak
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Kaixiang Huang
- Department of Chemistry, Indiana University, Bloomington, IN 47405, United States
| | - Gargi Jagdale
- Department of Chemistry, Indiana University, Bloomington, IN 47405, United States
| | - Lane A Baker
- Department of Chemistry, Indiana University, Bloomington, IN 47405, United States
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
22
|
MacAlpine J, Daniel-Ivad M, Liu Z, Yano J, Revie NM, Todd RT, Stogios PJ, Sanchez H, O'Meara TR, Tompkins TA, Savchenko A, Selmecki A, Veri AO, Andes DR, Fidel PL, Robbins N, Nodwell J, Whitesell L, Cowen LE. A small molecule produced by Lactobacillus species blocks Candida albicans filamentation by inhibiting a DYRK1-family kinase. Nat Commun 2021; 12:6151. [PMID: 34686660 PMCID: PMC8536679 DOI: 10.1038/s41467-021-26390-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/01/2021] [Indexed: 11/23/2022] Open
Abstract
The fungus Candida albicans is an opportunistic pathogen that can exploit imbalances in microbiome composition to invade its human host, causing pathologies ranging from vaginal candidiasis to fungal sepsis. Bacteria of the genus Lactobacillus are colonizers of human mucosa and can produce compounds with bioactivity against C. albicans. Here, we show that some Lactobacillus species produce a small molecule under laboratory conditions that blocks the C. albicans yeast-to-filament transition, an important virulence trait. It remains unexplored whether the compound is produced in the context of the human host. Bioassay-guided fractionation of Lactobacillus-conditioned medium linked this activity to 1-acetyl-β-carboline (1-ABC). We use genetic approaches to show that filamentation inhibition by 1-ABC requires Yak1, a DYRK1-family kinase. Additional biochemical characterization of structurally related 1-ethoxycarbonyl-β-carboline confirms that it inhibits Yak1 and blocks C. albicans biofilm formation. Thus, our findings reveal Lactobacillus-produced 1-ABC can prevent the yeast-to-filament transition in C. albicans through inhibition of Yak1.
Collapse
Affiliation(s)
- Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | | - Zhongle Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Junko Yano
- Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center School of Dentistry, New Orleans, LA, USA
| | - Nicole M Revie
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Robert T Todd
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter J Stogios
- BioZone, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Center for Structural Genomics of Infectious Diseases (CSGID), Chicago, IL, USA
| | - Hiram Sanchez
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Teresa R O'Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Thomas A Tompkins
- Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montreal, QC, Canada
| | - Alexei Savchenko
- BioZone, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Center for Structural Genomics of Infectious Diseases (CSGID), Chicago, IL, USA
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Amanda O Veri
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - David R Andes
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Paul L Fidel
- Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center School of Dentistry, New Orleans, LA, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Justin Nodwell
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
23
|
Khan F, Bamunuarachchi NI, Tabassum N, Jo DM, Khan MM, Kim YM. Suppression of hyphal formation and virulence of Candida albicans by natural and synthetic compounds. BIOFOULING 2021; 37:626-655. [PMID: 34284656 DOI: 10.1080/08927014.2021.1948538] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
Candida albicans undergoes a morphological yeast-to-hyphal transition during infection, which plays a significant role in its pathogenesis. The filamentous morphology of the hyphal form has been identified as a virulence factor as it facilitates surface adherence, intertwining with biofilm, invasion, and damage to host tissues and organs. Hence, inhibition of filamentation in addition to biofilm formation is considered a viable strategy against C. albicans infections. Furthermore, a good understanding of the signaling pathways involved in response to environmental cues driving hyphal growth is also critical to an understanding of C. albicans pathogenicity and to develop novel therapies. In this review, first the clinical significance and transcriptional control of C. albicans hyphal morphogenesis are addressed. Then, various strategies employed to suppress filamentation, prevent biofilm formation, and reduce virulence are discussed. These strategies include the inhibition of C. albicans filament formation using natural or synthetic compounds, and their combination with other agents or nanoformulations.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea
| | - Nilushi Indika Bamunuarachchi
- Department of Food Science and Technology, Pukyong National University, Busan, South Korea
- Department of Fisheries and Marine Sciences, Ocean University of Sri Lanka, Tangalle, Sri Lanka
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, South Korea
| | - Du-Min Jo
- Department of Food Science and Technology, Pukyong National University, Busan, South Korea
| | - Mohammad Mansoob Khan
- Chemical Sciences, Faculty of Science, University Brunei Darussalam, Gadong, Brunei Darussalam
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea
- Department of Food Science and Technology, Pukyong National University, Busan, South Korea
| |
Collapse
|
24
|
Computational Drug Repurposing Resources and Approaches for Discovering Novel Antifungal Drugs against Candida albicans N-Myristoyl Transferase. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is a yeast that is an opportunistic fungal pathogen and also identified as ubiquitous polymorphic species that is mainly linked with major fungal infections in humans, particularly in the immunocompromised patients including transplant recipients, chemotherapy patients, HIV-infected patients as well as in low-birth-weight infants. Systemic Candida infections have a high mortality rate of around 29 to 76%. For reducing its infection, limited drugs are existing such as caspofungin, fluconazole, terbinafine, and amphotericin B, etc. which contain unlikable side effects and also toxic. This review intends to utilize advanced bioinformatics technologies such as Molecular docking, Scaffold hopping, Virtual screening, Pharmacophore modeling, Molecular dynamics (MD) simulation for the development of potentially new drug candidates with a drug-repurpose approach against Candida albicans within a limited time frame and also cost reductive.
Collapse
|
25
|
Atriwal T, Azeem K, Husain FM, Hussain A, Khan MN, Alajmi MF, Abid M. Mechanistic Understanding of Candida albicans Biofilm Formation and Approaches for Its Inhibition. Front Microbiol 2021; 12:638609. [PMID: 33995297 PMCID: PMC8121174 DOI: 10.3389/fmicb.2021.638609] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
In recent years, the demand for novel antifungal therapies has increased several- folds due to its potential to treat severe biofilm-associated infections. Biofilms are made by the sessile microorganisms attached to the abiotic or biotic surfaces, enclosed in a matrix of exopolymeric substances. This results in new phenotypic characteristics and intrinsic resistance from both host immune response and antimicrobial drugs. Candida albicans biofilm is a complex association of hyphal cells that are associated with both abiotic and animal tissues. It is an invasive fungal infection and acts as an important virulent factor. The challenges linked with biofilm-associated diseases have urged scientists to uncover the factors responsible for the formation and maturation of biofilm. Several strategies have been developed that could be adopted to eradicate biofilm-associated infections. This article presents an overview of the role of C. albicans biofilm in its pathogenicity, challenges it poses and threats associated with its formation. Further, it discusses strategies that are currently available or under development targeting prostaglandins, quorum-sensing, changing surface properties of biomedical devices, natural scaffolds, and small molecule-based chemical approaches to combat the threat of C. albicans biofilm. This review also highlights the recent developments in finding ways to increase the penetration of drugs into the extracellular matrix of biofilm using different nanomaterials against C. albicans.
Collapse
Affiliation(s)
- Tanu Atriwal
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Kashish Azeem
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammed Nadeem Khan
- Department of Tashreehul Badan, Faculty of Unani Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
26
|
Lee Y, Puumala E, Robbins N, Cowen LE. Antifungal Drug Resistance: Molecular Mechanisms in Candida albicans and Beyond. Chem Rev 2021; 121:3390-3411. [PMID: 32441527 PMCID: PMC8519031 DOI: 10.1021/acs.chemrev.0c00199] [Citation(s) in RCA: 378] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fungal infections are a major contributor to infectious disease-related deaths across the globe. Candida species are among the most common causes of invasive mycotic disease, with Candida albicans reigning as the leading cause of invasive candidiasis. Given that fungi are eukaryotes like their human host, the number of unique molecular targets that can be exploited for antifungal development remains limited. Currently, there are only three major classes of drugs approved for the treatment of invasive mycoses, and the efficacy of these agents is compromised by the development of drug resistance in pathogen populations. Notably, the emergence of additional drug-resistant species, such as Candida auris and Candida glabrata, further threatens the limited armamentarium of antifungals available to treat these serious infections. Here, we describe our current arsenal of antifungals and elaborate on the resistance mechanisms Candida species possess that render them recalcitrant to therapeutic intervention. Finally, we highlight some of the most promising therapeutic strategies that may help combat antifungal resistance, including combination therapy, targeting fungal-virulence traits, and modulating host immunity. Overall, a thorough understanding of the mechanistic principles governing antifungal drug resistance is fundamental for the development of novel therapeutics to combat current and emerging fungal threats.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Emily Puumala
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| |
Collapse
|
27
|
Rosiana S, Zhang L, Kim GH, Revtovich AV, Uthayakumar D, Sukumaran A, Geddes-McAlister J, Kirienko NV, Shapiro RS. Comprehensive genetic analysis of adhesin proteins and their role in virulence of Candida albicans. Genetics 2021; 217:iyab003. [PMID: 33724419 PMCID: PMC8045720 DOI: 10.1093/genetics/iyab003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
Candida albicans is a microbial fungus that exists as a commensal member of the human microbiome and an opportunistic pathogen. Cell surface-associated adhesin proteins play a crucial role in C. albicans' ability to undergo cellular morphogenesis, develop robust biofilms, colonize, and cause infection in a host. However, a comprehensive analysis of the role and relationships between these adhesins has not been explored. We previously established a CRISPR-based platform for efficient generation of single- and double-gene deletions in C. albicans, which was used to construct a library of 144 mutants, comprising 12 unique adhesin genes deleted singly, and every possible combination of double deletions. Here, we exploit this adhesin mutant library to explore the role of adhesin proteins in C. albicans virulence. We perform a comprehensive, high-throughput screen of this library, using Caenorhabditis elegans as a simplified model host system, which identified mutants critical for virulence and significant genetic interactions. We perform follow-up analysis to assess the ability of high- and low-virulence strains to undergo cellular morphogenesis and form biofilms in vitro, as well as to colonize the C. elegans host. We further perform genetic interaction analysis to identify novel significant negative genetic interactions between adhesin mutants, whereby combinatorial perturbation of these genes significantly impairs virulence, more than expected based on virulence of the single mutant constituent strains. Together, this study yields important new insight into the role of adhesins, singly and in combinations, in mediating diverse facets of virulence of this critical fungal pathogen.
Collapse
Affiliation(s)
- Sierra Rosiana
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON NIG 2W1, Canada
| | - Liyang Zhang
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Grace H Kim
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON NIG 2W1, Canada
| | | | - Deeva Uthayakumar
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON NIG 2W1, Canada
| | - Arjun Sukumaran
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON NIG 2W1, Canada
| | | | | | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON NIG 2W1, Canada
| |
Collapse
|
28
|
Rajasekar V, Darne P, Prabhune A, Kao RYT, Solomon AP, Ramage G, Samaranayake L, Neelakantan P. A curcumin-sophorolipid nanocomplex inhibits Candida albicans filamentation and biofilm development. Colloids Surf B Biointerfaces 2021; 200:111617. [PMID: 33592455 DOI: 10.1016/j.colsurfb.2021.111617] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/20/2022]
Abstract
Candida albicans is an opportunistic fungal pathogen that is highly resistant to contemporary antifungals, due to their biofilm lifestyle. The ability of C. albicans to invade human tissues is due to its filamentation. Therefore, inhibition of biofilms and filamentation of the yeast are high value targets to develop the next-generation antifungals. Curcumin (CU) is a natural polyphenol with excellent pharmacological attributes, but limitations such as poor solubility, acid, and enzyme tolerance have impeded its practical utility. Sophorolipids (SL) are biologically-derived surfactants that serve as efficient carriers of hydrophobic molecules such as curcumin into biofilms. Here, we synthesised a curcumin-sophorolipid nanocomplex (CUSL), and comprehensively evaluated its effects on C. albicans biofilms and filamentation. Our results demonstrated that sub-inhibitory concentration of CUSL (9.37 μg/mL) significantly inhibited fungal adhesion to substrates, and subsequent biofilm development, maturation, and filamentation. This effect was associated with significant downregulation of a select group of biofilm, adhesins, and hyphal regulatory genes. In conclusion, the curcumin-sophorolipid nanocomplex is a potent inhibitor of the two major virulence attributes of C. albicans, biofilm formation and filamentation, thus highlighting its promise as a putative anti-fungal agent with biofilm penetrative potential.
Collapse
Affiliation(s)
- Vidhyashree Rajasekar
- Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region; Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to Be University, Thanjavur, India
| | - Priti Darne
- Green Pyramid Biotech Private Limited, Pune, India
| | | | - Richard Y T Kao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to Be University, Thanjavur, India
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Lakshman Samaranayake
- Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Prasanna Neelakantan
- Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
29
|
Inhibitory Effect of Nepeta deflersiana on Climax Bacterial Community Isolated from the Oral Plaque of Patients with Periodontal Disease. Molecules 2021; 26:molecules26010202. [PMID: 33401536 PMCID: PMC7795411 DOI: 10.3390/molecules26010202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The red-complex bacteria are one of the most significant complexes found simultaneously in subgingival plaque next to the periodontal pocket. The current antibacterial treatment is not adequate, and multidrug resistance to it is developing. Henceforth, the antibacterial effect of the ethanolic extract of Nepeta deflersiana was put to test against red-complex bacteria in patients with chronic periodontitis. METHODS Well diffusion and micro broth dilution procedure by Alamar blue were applied to assess the zone of inhibition (ZOI), the minimum inhibitory concentration (MIC), and the minimum bactericidal concentration (MBC). Anti-virulence efficacies of the plant extract that comprise of adherence and formation of biofilms were examined by the process of adherence and biofilm production assay. RESULTS The crude extract of Nepeta deflersiana exhibited significant inhibitory outcome against periodontopathic bacteria with noteworthy MIC (0.78-3.12 mg/mL), inhibitory zone (12-20 mm), as well as MBC (3.12-12.50 mg/mL). The N. deflersiana extract inhibited bacterial adhesion ranging from 41% to 52%, 53% to 66%, and 60% to 79% at the given MIC × 0.5, MIC × 1, and MIC × 2 in succession. Substantial suppression was also developed in the biofilm production of the investigated periodontopathic strains following exposure to numerous concentrations of N. deflersianan extract for a period of 24 and 48 h. CONCLUSION These outcomes divulge a new concept that N. deflersiana extract can be utilized to manufacture valuable antibacterial compounds to treat chronic and acute periodontitis. This identifies N. deflersiana as an essential natural source for future drug development.
Collapse
|
30
|
Inhibition of Distinct Proline- or N-Acetylglucosamine-Induced Hyphal Formation Pathways by Proline Analogs in Candida albicans. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7245782. [PMID: 33274221 PMCID: PMC7695494 DOI: 10.1155/2020/7245782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 11/22/2022]
Abstract
Candida albicans undergoes a yeast-to-hyphal transition that has been recognized as a virulence property as well as a turning point leading to biofilm formation associated with candidiasis. It is known that yeast-to-hyphal transition is induced under complex environmental conditions including temperature (above 35°C), pH (greater than 6.5), CO2, N-acetylglucosamine (GlcNAc), amino acids, RPMI-1640 synthetic culture medium, and blood serum. To identify the hyphal induction factor in the RPMI-1640 medium, we examined each component of RPMI-1640 and established a simple hyphal induction condition, that is, incubation in L-proline solution at 37°C. Incubation in GlcNAc solution alone, which is not contained in RPMI-1640, without any other materials was also identified as another simple hyphal induction condition. To inhibit hyphal formation, proline and GlcNAc analogs were examined. Among the proline analogs used, L-azetidine-2-carboxylic acid (AZC) inhibited hyphal induction under both induction conditions, but L-4-thiazolidinecarboxylic acid (T4C) specifically inhibited proline-induced hyphal formation only, while α-N-methyl-L-proline (mPro) selectively inhibited GlcNAc-induced hyphal formation. Hyphal formation in fetal bovine serum was also inhibited by AZC or T4C together with mPro without affecting the proliferation of yeast form. These results indicate that these proline analogs are ideal inhibitors of yeast-to-hyphal transition in C. albicans.
Collapse
|
31
|
Kunyeit L, K A AA, Rao RP. Application of Probiotic Yeasts on Candida Species Associated Infection. J Fungi (Basel) 2020; 6:jof6040189. [PMID: 32992993 PMCID: PMC7711718 DOI: 10.3390/jof6040189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 01/01/2023] Open
Abstract
Superficial and life-threatening invasive Candida infections are a major clinical challenge in hospitalized and immuno-compromised patients. Emerging drug-resistance among Candida species is exacerbated by the limited availability of antifungals and their associated side-effects. In the current review, we discuss the application of probiotic yeasts as a potential alternative/ combination therapy against Candida infections. Preclinical studies have identified several probiotic yeasts that effectively inhibit virulence of Candida species, including Candida albicans, Candida tropicalis, Candida glabrata, Candida parapsilosis, Candida krusei and Candida auris. However, Saccharomyces cerevisiae var. boulardii is the only probiotic yeast commercially available. In addition, clinical studies have further confirmed the in vitro and in vivo activity of the probiotic yeasts against Candida species. Probiotics use a variety of protective mechanisms, including posing a physical barrier, the ability to aggregate pathogens and render them avirulent. Secreted metabolites such as short-chain fatty acids effectively inhibit the adhesion and morphological transition of Candida species. Overall, the probiotic yeasts could be a promising effective alternative or combination therapy for Candida infections. Additional studies would bolster the application of probiotic yeasts.
Collapse
Affiliation(s)
- Lohith Kunyeit
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological Research Institute (CFTRI), Mysuru 570020, India; (L.K.); (A.K.A.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Anu-Appaiah K A
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological Research Institute (CFTRI), Mysuru 570020, India; (L.K.); (A.K.A.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Reeta P. Rao
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
- Correspondence: ; Tel.: +1-508-831-5000
| |
Collapse
|
32
|
Vera-González N, Shukla A. Advances in Biomaterials for the Prevention and Disruption of Candida Biofilms. Front Microbiol 2020; 11:538602. [PMID: 33042051 PMCID: PMC7527432 DOI: 10.3389/fmicb.2020.538602] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Candida species can readily colonize a multitude of indwelling devices, leading to biofilm formation. These three-dimensional, surface-associated Candida communities employ a multitude of sophisticated mechanisms to evade treatment, leading to persistent and recurrent infections with high mortality rates. Further complicating matters, the current arsenal of antifungal therapeutics that are effective against biofilms is extremely limited. Antifungal biomaterials are gaining interest as an effective strategy for combating Candida biofilm infections. In this review, we explore biomaterials developed to prevent Candida biofilm formation and those that treat existing biofilms. Surface functionalization of devices employing clinically utilized antifungals, other antifungal molecules, and antifungal polymers has been extremely effective at preventing fungi attachment, which is the first step of biofilm formation. Several mechanisms can lead to this attachment inhibition, including contact killing and release-based killing of surrounding planktonic cells. Eliminating mature biofilms is arguably much more difficult than prevention. Nanoparticles have shown the most promise in disrupting existing biofilms, with the potential to penetrate the dense fungal biofilm matrix and locally target fungal cells. We will describe recent advances in both surface functionalization and nanoparticle therapeutics for the treatment of Candida biofilms.
Collapse
Affiliation(s)
- Noel Vera-González
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Anita Shukla
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
- Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| |
Collapse
|
33
|
Gutierrez D, Weinstock A, Antharam VC, Gu H, Jasbi P, Shi X, Dirks B, Krajmalnik-Brown R, Maldonado J, Guinan J, Thangamani S. Antibiotic-induced gut metabolome and microbiome alterations increase the susceptibility to Candida albicans colonization in the gastrointestinal tract. FEMS Microbiol Ecol 2020; 96:5643884. [PMID: 31769789 PMCID: PMC6934136 DOI: 10.1093/femsec/fiz187] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023] Open
Abstract
Antibiotic-induced alterations in the gut ecosystem increases the susceptibility to Candida albicans, yet the mechanisms involved remains poorly understood. Here we show that mice treated with the broad-spectrum antibiotic cefoperazone promoted the growth, morphogenesis and gastrointestinal (GI) colonization of C. albicans. Using metabolomics, we revealed that the cecal metabolic environment of the mice treated with cefoperazone showed a significant alteration in intestinal metabolites. Levels of carbohydrates, sugar alcohols and primary bile acids increased, whereas carboxylic acids and secondary bile acids decreased in antibiotic treated mice susceptible to C. albicans. Furthermore, using in-vitro assays, we confirmed that carbohydrates, sugar alcohols and primary bile acids promote, whereas carboxylic acids and secondary bile acids inhibit the growth and morphogenesis of C. albicans. In addition, in this study we report changes in the levels of gut metabolites correlated with shifts in the gut microbiota. Taken together, our in-vivo and in-vitro results indicate that cefoperazone-induced metabolome and microbiome alterations favor the growth and morphogenesis of C. albicans, and potentially play an important role in the GI colonization of C. albicans.
Collapse
Affiliation(s)
- Daniel Gutierrez
- College of Veterinary Medicine, Midwestern University, 19555 N. 59th Ave. Glendale, AZ 85308, USA
| | - Anthony Weinstock
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Ave. Glendale, AZ 85308, USA
| | - Vijay C Antharam
- Department of Chemistry, School of Science and Human Development, Methodist University, 5400 Ramsey St, Fayetteville, NC 28311, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85259, USA
| | - Paniz Jasbi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85259, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85259, USA
| | - Blake Dirks
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ 85280, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ 85280, USA.,School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ 85287, USA.,Biodesign Center for Fundamental and Applied Microbiomics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Juan Maldonado
- Biodesign Center for Fundamental and Applied Microbiomics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Jack Guinan
- College of Veterinary Medicine, Midwestern University, 19555 N. 59th Ave. Glendale, AZ 85308, USA
| | - Shankar Thangamani
- Department of Pathology and Population Medicine, College of Veterinary Medicine, Midwestern University, 19555 N. 59th Ave. Glendale, AZ 85308, USA
| |
Collapse
|
34
|
Ahmad I, Wahab S, Nisar N, Dera AA, Alshahrani MY, Abullias SS, Irfan S, Alam MM, Srivastava S. Evaluation of antibacterial properties of Matricaria aurea on clinical isolates of periodontitis patients with special reference to red complex bacteria. Saudi Pharm J 2020; 28:1203-1209. [PMID: 33132714 PMCID: PMC7584800 DOI: 10.1016/j.jsps.2020.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/11/2020] [Indexed: 01/18/2023] Open
Abstract
Background Chronic periodontitis has an interplay between different species of bacteria found in dental biofilms act a crucial role in pathogenesis and disease progression. The existing antibacterial therapy is inadequate, associated with many side effects as well as evolving multidrug resistance. Hence, novel drugs development with minimum or no toxicity is an immediate priority. Methods Antibacterial efficacy of ethanolic extract of Matricaria aurea was tested against clinical isolates, ie. Treponema denticol, Tannerella forsythia, Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis from the patients with chronic periodontitis. Zone of inhibition, the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) were investigated by well diffusion method and micro broth dilution assay using alamar blue. Anti-virulence properties of the extract, which include adherence property and the biofilm formation, were investigated by adherence as well as biofilm formation assay. Results Matricaria aurea extract showed potent inhibitory effect against pathogenic periodontal bacteria with the significant inhibitory zone (13–23 mm), MIC (0.39–1.56 mg/ml) as well as MBC (1.56–6.25 mg/ml). The M. aurea extract was able to inhibit bacterial adhesion ranged from 30 to 45%, 35 to 63% and 55 to 80% of MIC at MIC × 0.5, MIC × 1 and MIC × 2 respectively. Significant inhibition was found in biofilm formation to all the tested periodontal bacterial strains after the treatment with various concentrations of M. aurea extract for 24 and 48hrs. Conclusion These results reveal for the first time that the Matricaria aurea extract might be the source of various compounds to be applied for chronic periodontitis therapy, which might draw these valuable compounds to the subsequent phase of development of the drug.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Nazima Nisar
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ayed A Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Shahabe Saquib Abullias
- Department of Periodontics and Community Dental Sciences, College of Dentistry, King Khalid University, Saudi Arabia
| | - Safia Irfan
- Department of Biosciences, Faculty of Sciences, Integral University, Lucknow, UP, India
| | - Mohammad Mahtab Alam
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Srivastava
- Department of Pharmaceutics, Era College of Pahrmacy, Era University, Lucknow, UP, India
| |
Collapse
|
35
|
Martin H, Somers T, Dwyer M, Robson R, Pfeffer FM, Bjornsson R, Krämer T, Kavanagh K, Velasco-Torrijos T. Scaffold diversity for enhanced activity of glycosylated inhibitors of fungal adhesion. RSC Med Chem 2020; 11:1386-1401. [PMID: 34095846 DOI: 10.1039/d0md00224k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
Candida albicans is one of the most prevalent fungal pathogens involved in hospital acquired infections. It binds to glycans at the surface of epithelial cells and initiates infection. This process can be blocked by synthetic carbohydrates that mimic the structure of cell surface glycans. Herein we report the evaluation of a series of divalent glycosides featuring aromatic (benzene, squaramide) and bicyclic aliphatic (norbornene) scaffolds, with the latter being the first examples of their kind as small molecule anti-adhesion glycoconjugates. Galactosides 1 and 6, built on an aromatic core, were most efficient inhibitors of adhesion of C. albicans to buccal epithelial cells, displacing up to 36% and 48%, respectively, of yeast already attached to epithelial cells at 138 μM. Remarkably, cis-endo-norbornene 21 performed comparably to benzene-core derivatives. Conformational analysis reveals a preference for compounds 1 and 21 to adopt folded conformations. These results highlight the potential of norbornenes as a new class of aliphatic scaffolds for the synthesis of anti-adhesion compounds.
Collapse
Affiliation(s)
- Harlei Martin
- Department of Chemistry, Maynooth University Maynooth Co. Kildare Ireland
| | - Tara Somers
- Department of Biology, Maynooth University Maynooth Co. Kildare Ireland
| | - Mathew Dwyer
- Department of Biology, Maynooth University Maynooth Co. Kildare Ireland
| | - Ryan Robson
- School of Life and Environmental Sciences, Deakin University Geelong Victoria 3217 Australia
| | - Frederick M Pfeffer
- School of Life and Environmental Sciences, Deakin University Geelong Victoria 3217 Australia
| | - Ragnar Bjornsson
- Department of Inorganic Spectroscopy, Max Planck Institute for Chemical Energy Conversion Stiftstrasse 34-36 45470 Mülheim an der Ruhr Germany
| | - Tobias Krämer
- Department of Chemistry, Maynooth University Maynooth Co. Kildare Ireland .,The Hamilton Institute, Maynooth University Maynooth Co. Kildare Ireland
| | - Kevin Kavanagh
- Department of Biology, Maynooth University Maynooth Co. Kildare Ireland.,The Kathleen Lonsdale Institute for Human Health Research, Maynooth University Maynooth Co. Kildare Ireland
| | - Trinidad Velasco-Torrijos
- Department of Chemistry, Maynooth University Maynooth Co. Kildare Ireland .,The Kathleen Lonsdale Institute for Human Health Research, Maynooth University Maynooth Co. Kildare Ireland
| |
Collapse
|
36
|
Abstract
Fungal infections with increasing resistance to conventional therapies are a growing concern. Candida albicans is a major opportunistic yeast responsible for mucosal and invasive infections. Targeting the initial step of the infection process (i.e., C. albicans adhesion to the host cell) is a promising strategy. A wide variety of molecules can interfere with adhesion processes via an assortment of mechanisms. Herein, we focus on how small molecules disrupt biosynthesis of fungal cell wall components and membrane structure, prevent the localization of GPI-anchor proteins, inhibit production of enzymes involved in adhesion, downregulate genes encoding adhesins and competitively inhibit receptor interactions. As a result, adhesion of C. albicans to host cells is reduced, paving the way to new classes of antifungal agents.
Collapse
|
37
|
Lone SA, Ahmad A. Inhibitory effect of novel Eugenol Tosylate Congeners on pathogenicity of Candida albicans. BMC Complement Med Ther 2020; 20:131. [PMID: 32349730 PMCID: PMC7191809 DOI: 10.1186/s12906-020-02929-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The global prevalence of fungal diseases is increasing rapidly, which affects more than a billion people every year with significant mortality rate. On the other hand, the development of new drugs to treat these fungal infections is slow, while the current antifungal therapy is insufficient and associated with adverse side effects and emerging multidrug resistance. Therefore, development of novel antifungal drugs with least or no toxicity and multi-target mechanisms of action is an immediate priority. Natural products have long been known to possess antimicrobial activities and are source of new drugs. Currently, modifying natural products to synthesize derivatives/analogues are of great scientific focus for discovering novel drugs with improved potency and safety. Modifications in eugenol to synthesize eugenol derivatives with enhanced antifungal activity have already been reported. METHODS In this study, three most active novel eugenol tosylate congeners (ETC-5, ETC-6 and ETC-7) were selected from our previous study to investigate their effect on major virulence factors of Candida albicans which include adherence, morphogenesis, hydrolytic enzymes secretion, biofilm formation and on expression of genes related to these virulence factors. Adherence and biofilm formation were studied by alamarBlue dye and XTT reduction assays respectively, hydrolytic enzyme secretion was evaluated by plate assays. Further, morphological transition was monitored microscopically and RT-qPCR was used to assess the gene expression levels. RESULTS ETCs significantly inhibited adherence in C. albicans with an inhibition range of 16-66%, and completely inhibited the morphogenesis at MIC values. Inhibition of proteinase and phospholipase activity was in the range of 2-48% and 8-34% respectively. Test compounds also significantly inhibit biofilm formation in C. albicans in the range of 7-77%. Furthermore, RT-qPCR results indicated a significant down regulation in expression levels of genes (ALS1, ALS2, ALS3, ALS9, CPH1, HWP1, SAP1, SAP2, SAP3 and PLB1) in C. albicans cells after treated with ETCs. CONCLUSION The results indicated that these novel ETCs target major virulence factors of C. albicans and avert this commensal microbe to turn into pathogenic. However, further in-depth studies may facilitate the mechanisms involved by ETCs in targeting these virulence factors.
Collapse
Affiliation(s)
- Shabir Ahmad Lone
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa. .,Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg, 2193, South Africa.
| |
Collapse
|
38
|
Abstract
Candida albicans has remained the main etiological agent of candidiasis, challenges clinicians with high mortality and morbidity. The emergence of resistance to antifungal drugs, toxicity and lower efficacy have all contributed to an urgent need to develop alternative drugs aiming at novel targets in C. albicans. Targeting the production of virulence factors, which are essential processes for infectious agents, represents an attractive substitute for the development of newer anti-infectives. The present review highlights the recent developments made in the understanding of the pathogenicity of C. albicans. Production of hydrolytic enzymes, morphogenesis and biofilm formation, along with their molecular and metabolic regulation in Candida are discussed with regard to the development of novel antipathogenic drugs against candidiasis. Over the last decade, candidiasis has remained a major problematic disease worldwide. In spite of the existence of many antifungal drugs, the treatment of such diseases has still remained unsuccessful due to drug inefficacy. Therefore, there is a need to discover antifungals with different modes of action, such as antipathogenic drugs against Candida albicans. Here, we describe how various types of virulence factors such as proteinase, phospholipase, hemolysin, adhesion, morphogenesis and biofilm formation, could be targeted to develop novel therapeutics. We can inhibit production of these virulence factors by controlling their molecular/metabolic regulation.
Collapse
|
39
|
Low ZJ, Xiong J, Xie Y, Ma GL, Saw H, Thi Tran H, Wong SL, Pang LM, Fong J, Lu P, Hu JF, Yang L, Miao Y, Liang ZX. Discovery, biosynthesis and antifungal mechanism of the polyene-polyol meijiemycin. Chem Commun (Camb) 2019; 56:822-825. [PMID: 31848534 DOI: 10.1039/c9cc08908j] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Produced by a newly isolated Streptomycetes strain, meijiemycin is a gigantic linear polyene-polyol that exhibits structural features not seen in other members of the polyene-polyol family. We propose a biosynthetic mechanism and demonstrate that meijiemycin inhibits hyphal growth by inducing the aggregation of ergosterol and restructuring of the fungal plasma membrane.
Collapse
Affiliation(s)
- Zhen Jie Low
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Juan Xiong
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore. and School of Pharmacy, Fudan University, Shanghai, China
| | - Ying Xie
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Guang-Lei Ma
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Howard Saw
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Hoa Thi Tran
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Soo Lin Wong
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Li Mei Pang
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - July Fong
- The Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, 637551, Singapore
| | - Peng Lu
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Jin-Feng Hu
- School of Pharmacy, Fudan University, Shanghai, China
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Zhao-Xun Liang
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore. and The Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, 637551, Singapore
| |
Collapse
|
40
|
Dwivedi M, Muralidhar S, Saluja D. Hibiscus sabdariffa Extract Inhibits Adhesion, Biofilm Initiation and Formation in Candida albicans. Indian J Microbiol 2019; 60:96-106. [PMID: 32089579 DOI: 10.1007/s12088-019-00835-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 10/23/2019] [Indexed: 10/25/2022] Open
Abstract
Microbial biofilms act as reservoirs for pathogenic sessile microbes which reside inside the three dimensional matrix of the biofilm, and are thus protected against anti-microbial drugs. Most of the anti-microbial drugs fail to completely abolish the biofilm associated infections. In the present study, we provide evidence of Hibiscus sabdariffa (Hs) extract having possible anti-microbial activity, with emphasis on Candida albicans biofilm. The Hs extract was shown to be effective against C. albicans pre-formed biofilm at 3.125 mg/ml and was able to inhibit the hyphae initiation and adherence of cells. Furthermore, Hs extract was able to reduce the C. albicans load in C. elegans by effectively killing the Candida cells thereby reducing the viable colony count and effectively increasing the lifespan of worms. The percentage of viable hatched progeny of worms exposed to Hs extract (both at conc. 1.5 mg/ml and 6.25 mg/ml), was also comparable to that of the control untreated eggs. The Hs extract was also found to be significantly effective against fluconazole resistant C. albicans isolated from patients. Thus, we, for the first time, propose Hs extract as a prospective drug candidate and substitute for eradicating pre-formed biofilm and inhibiting the growth of C. albicans.
Collapse
Affiliation(s)
- Meenakshi Dwivedi
- 1Medical Biotechnology Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, 110007 India
| | - Sumathi Muralidhar
- 2Apex-Regional STD Centre and State Reference Laboratory for HIV, Department of Microbiology, Vardhman Mahavir Medical College, Safdarjung Hospital, New Delhi, 110029 India
| | - Daman Saluja
- 1Medical Biotechnology Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, 110007 India
| |
Collapse
|
41
|
Kunyeit L, Kurrey NK, Anu-Appaiah KA, Rao RP. Probiotic Yeasts Inhibit Virulence of Non -albicans Candida Species. mBio 2019; 10:e02307-19. [PMID: 31615960 PMCID: PMC6794482 DOI: 10.1128/mbio.02307-19] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 12/24/2022] Open
Abstract
Systemic infections of Candida species pose a significant threat to public health. Toxicity associated with current therapies and emergence of resistant strains present major therapeutic challenges. Here, we report exploitation of the probiotic properties of two novel, food-derived yeasts, Saccharomyces cerevisiae (strain KTP) and Issatchenkia occidentalis (strain ApC), as an alternative approach to combat widespread opportunistic fungal infections. Both yeasts inhibit virulence traits such as adhesion, filamentation, and biofilm formation of several non-albicans Candida species, including Candida tropicalis, Candida krusei, Candida glabrata, and Candida parapsilosis as well as the recently identified multidrug-resistant species Candida auris They inhibit adhesion to abiotic surfaces as well as cultured colon epithelial cells. Furthermore, probiotic treatment blocks the formation of biofilms of individual non-albicans Candida strains as well as mixed-culture biofilms of each non-albicans Candida strain in combination with Candida albicans The probiotic yeasts attenuated non-albicans Candida infections in a live animal. In vivo studies using Caenorhabditis elegans suggest that exposure to probiotic yeasts protects nematodes from infection with non-albicans Candida strains compared to worms that were not exposed to the probiotic yeasts. Furthermore, application of probiotic yeasts postinfection with non-albicans Candida alleviated pathogenic colonization of the nematode gut. The probiotic properties of these novel yeasts are better than or comparable to those of the commercially available probiotic yeast Saccharomyces boulardii, which was used as a reference strain throughout this study. These results indicate that yeasts derived from food sources could serve as an effective alternative to antifungal therapy against emerging pathogenic Candida species.IMPORTANCE Non-albicans Candida-associated infections have emerged as a major risk factor in the hospitalized and immunecompromised patients. Besides, antifungal-associated complications occur more frequently with these non-albicans Candida species than with C. albicans Therefore, as an alternative approach to combat these widespread non-albicans Candida-associated infections, here we showed the probiotic effect of two yeasts, Saccharomyces cerevisiae (strain KTP) and Issatchenkia occidentalis (ApC), in preventing adhesion and biofilm formation of five non-albicans Candida strains, Candida tropicalis, Candida krusei, Candida glabrata, Candida parapsilosis, and Candida auris The result would influence the current trend of the conversion of conventional antimicrobial therapy into beneficial probiotic microbe-associated antimicrobial treatment.
Collapse
Affiliation(s)
- Lohith Kunyeit
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute (CFTRI), Mysore, India
- Academy of Scientific and Innovative Research (AcSIR), CFTRI, Mysore, India
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Nawneet K Kurrey
- Department of Biochemistry, CSIR-Central Food Technological Research Institute (CFTRI), Mysore, India
| | - K A Anu-Appaiah
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute (CFTRI), Mysore, India
- Academy of Scientific and Innovative Research (AcSIR), CFTRI, Mysore, India
| | - Reeta P Rao
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
42
|
Caenorhabditis elegans as a model animal for investigating fungal pathogenesis. Med Microbiol Immunol 2019; 209:1-13. [PMID: 31555911 DOI: 10.1007/s00430-019-00635-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/18/2019] [Indexed: 12/27/2022]
Abstract
The morbidity and mortality associated with systemic fungal infections in humans cannot be underestimated. The nematode Caenorhabditis elegans has become popular for the in vivo study of the pathogenesis of human fungal pathogens and as an antifungal drug-screening tool. C. elegans offers many advantages as a model organism for the study of human fungal diseases, including lack of ethics requirements, easy maintenance in the laboratory, fully sequenced genome, availability of genetic mutants, and the possibility of liquid assays for high-throughput antifungal screening. Its major drawbacks include the inability to grow at 37 °C and absence of an adaptive immune response. However, several virulence factors involved in the pathogenesis of medically important fungal pathogens have been identified using the C. elegans model, consequently providing new leads for drug discovery and potential drug targets. We review the use of C. elegans as a model animal to understand the pathogenesis of medically important human fungal pathogens and the discovery of novel antifungal compounds. The review makes a case for C. elegans as a suitable invertebrate model for a plethora of practical applications in the investigation of fungal pathogenesis as well as its amenability for liquid-based high-throughput screening of potential antifungal compounds.
Collapse
|
43
|
Efficacy of Compounds Isolated from Streptomyces olivaceus against the Morphogenesis and Virulence of Candida albicans. Mar Drugs 2019; 17:md17080442. [PMID: 31357504 PMCID: PMC6723460 DOI: 10.3390/md17080442] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023] Open
Abstract
Candida albicans is a type of commensal fungi which causes serious infections in immunocompromised patients and contributes to high mortality. In the present study, we identified that the extract from Streptomyces olivaceus SCSIO T05 inhibited hypha and biofilm formation of C. albicans. Seven compounds were isolated and evaluated for their effects on the biological functions and virulence of C. albicans. Two leading compounds, compound 1 (sorbicillin) and compound 2 (3-methyl-N-(2′-phenethyl)-butyrylamide) were identified as exhibiting strong activity against C. albicans morphological transition, adhesion activity, cytotoxicity, and adhesion to human cells, in a dose-dependent manner. Notably, compound 2 inhibited C. albicans infection in mouse oral mucosal models. Transcriptomic analysis and real-time PCR results revealed that compound 2 most likely inhibited the biological functions of C. albicans cells by regulating the expression levels of HWP1, TEC1, ALS1, IFD6, and CSH1, which are associated with filament formation and cell adhesion. Our results suggest that the candidate compounds present excellent efficacy against C. albicans pathogenicity and that they can be developed as potential options for the clinical treatment of candidiasis.
Collapse
|
44
|
Regulatory mechanisms controlling morphology and pathogenesis in Candida albicans. Curr Opin Microbiol 2019; 52:27-34. [PMID: 31129557 DOI: 10.1016/j.mib.2019.04.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/08/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022]
Abstract
Candida albicans, a major human fungal pathogen, can cause a wide variety of both mucosal and systemic infections, particularly in immunocompromised individuals. Multiple lines of evidence suggest a strong association between virulence and the ability of C. albicans to undergo a reversible morphological transition from yeast to filamentous cells in response to host environmental cues. Most previous studies on mechanisms important for controlling the C. albicans morphological transition have focused on signaling pathways and sequence-specific transcription factors. However, in recent years a variety of novel mechanisms have been reported, including those involving global transcriptional regulation and translational control. A large-scale functional genomics screen has also revealed new roles in filamentation for certain key biosynthesis pathways. This review article will highlight several of these exciting recent discoveries and discuss how they are relevant to the development of novel antifungal strategies. Ultimately, components of mechanisms that control C. albicans morphogenesis and pathogenicity could potentially serve as viable antifungal targets.
Collapse
|
45
|
Yan Y, Tan F, Miao H, Wang H, Cao Y. Effect of Shikonin Against Candida albicans Biofilms. Front Microbiol 2019; 10:1085. [PMID: 31156594 PMCID: PMC6527961 DOI: 10.3389/fmicb.2019.01085] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 04/30/2019] [Indexed: 11/13/2022] Open
Abstract
Candidiasis is often associated with the formation of biofilms. Candida albicans biofilms are inherently resistant to many clinical antifungal agents and have increasingly been found to be the sources of C. albicans infections. Novel antifungal agents against C. albicans biofilms are urgently needed. The aim of this study was to investigate the effect of shikonin (SK) against C. albicans biofilms and to clarify the underlying mechanisms. XTT reduction assay showed that SK could not only inhibit the formation of biofilms but also destroy the maintenance of mature biofilms. In a mouse vulvovaginal candidiasis (VVC) model, the fungal burden was remarkably reduced upon SK treatment. Further study showed that SK could inhibit hyphae formation and reduce cellular surface hydrophobicity (CSH). Real-time reverse transcription-PCR analysis revealed that several hypha- and adhesion-specific genes were differentially expressed in SK-treated biofilm, including the downregulation of ECE1, HWP1, EFG1, CPH1, RAS1, ALS1, ALS3, CSH1 and upregulation of TUP1, NRG1, BCR1. Moreover, SK induced the production of farnesol, a quorum sensing molecule, and exogenous addition of farnesol enhanced the antibiofilm activity of SK. Taken together, these results indicated that SK could be a favorable antifungal agent in the clinical management of C. albicans biofilms.
Collapse
Affiliation(s)
- Yu Yan
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei Tan
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Miao
- Institute of Basic Medicine, Chengde Medical University, Chengde, China
| | - Hui Wang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - YingYing Cao
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Wall G, Montelongo-Jauregui D, Vidal Bonifacio B, Lopez-Ribot JL, Uppuluri P. Candida albicans biofilm growth and dispersal: contributions to pathogenesis. Curr Opin Microbiol 2019; 52:1-6. [PMID: 31085405 DOI: 10.1016/j.mib.2019.04.001] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/02/2019] [Indexed: 12/20/2022]
Abstract
The fungal species Candida albicans is most frequently associated with biofilm formation in immune-compromised and medically compromised patients, and it is now firmly established that biofilm formation represents a major virulence factor during candidiasis. A growing body of evidence has demonstrated that C. albicans biofilm development is a highly regulated and coordinated process, where adhesive interactions, morphogenetic conversions, and consortial behavior play significant roles. Cells within the biofilms are protected from environmental stresses including host immune defenses and antifungal treatment, which carries important clinical consequences for the treatment of biofilm-associated infections. Dispersal of cells from biofilms represents one of the hallmarks of the biofilm life-style, and in the case of C. albicans dispersed cells are responsible for candidemia and dissemination leading to the establishment of invasive disease.
Collapse
Affiliation(s)
- Gina Wall
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Daniel Montelongo-Jauregui
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Bruna Vidal Bonifacio
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Jose L Lopez-Ribot
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Priya Uppuluri
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, 90509, USA.
| |
Collapse
|
47
|
Díaz-García J, Marcos-Zambrano LJ, Muñoz P, Guinea J, Escribano P. Does the composition of polystyrene trays affect Candida spp. biofilm formation? Med Mycol 2019; 57:504-509. [PMID: 30202852 DOI: 10.1093/mmy/myy064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/22/2018] [Accepted: 07/14/2018] [Indexed: 01/01/2023] Open
Abstract
The biofilm formation ability of Candida species seems to have a role in the prognosis of patients with candidemia. Biofilm formation is usually tested using 96 well flat bottom polystyrene microtiter plates, although the type of plastic used is not commonly reported. This study compares biofilm formation by Candida spp. on six types of plates from three brands (three non-tissue-treated and three tissue-treated). Thirty isolates of each of the following species were selected: C. albicans, C. parapsilosis, C. glabrata, C. tropicalis, as well as 15 isolates of C. krusei (n = 135 isolates) from patients with candidemia. Biofilm production was evaluated by measuring biomass production and metabolic activity. Our results show higher biomass production and metabolic activity of biofilms formed on non-tissue-treated plates in comparison to those formed on tissue-treated plates (P < .001). We only found significant differences in metabolic activity of biofilms formed on non-tissue-treated plates (P < .003). All comparisons including biofilm formation and metabolic activity among plates of the same brand yielded higher biofilm formation on non-treated plates compared to treated plates (P < .001). Significant difference in biomass production by C. parapsilosis was only seen when comparing between the various tissue-treated plastics (P < .03). In contrast, comparisons of different non-tissue-treated tray brands yielded significant metabolic activity differences for all species except for C. parapsilosis (P < .05). Biofilm formation and metabolic activity is significantly affected by the plastic composition of non-tissue-treated trays leading to increased biofilm formation.
Collapse
Affiliation(s)
- Judith Díaz-García
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Madrid, Spain
| | - Laura Judith Marcos-Zambrano
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Madrid, Spain
| | - Patricia Muñoz
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain.,Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Jesús Guinea
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain.,Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Pilar Escribano
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Madrid, Spain
| |
Collapse
|
48
|
Sharma J, Rosiana S, Razzaq I, Shapiro RS. Linking Cellular Morphogenesis with Antifungal Treatment and Susceptibility in Candida Pathogens. J Fungi (Basel) 2019; 5:E17. [PMID: 30795580 PMCID: PMC6463059 DOI: 10.3390/jof5010017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023] Open
Abstract
Fungal infections are a growing public health concern, and an increasingly important cause of human mortality, with Candida species being amongst the most frequently encountered of these opportunistic fungal pathogens. Several Candida species are polymorphic, and able to transition between distinct morphological states, including yeast, hyphal, and pseudohyphal forms. While not all Candida pathogens are polymorphic, the ability to undergo morphogenesis is linked with the virulence of many of these pathogens. There are also many connections between Candida morphogenesis and antifungal drug treatment and susceptibility. Here, we review how Candida morphogenesis-a key virulence trait-is linked with antifungal drugs and antifungal drug resistance. We highlight how antifungal therapeutics are able to modulate morphogenesis in both sensitive and drug-resistant Candida strains, the shared signaling pathways that mediate both morphogenesis and the cellular response to antifungal drugs and drug resistance, and the connection between Candida morphology, drug resistance, and biofilm growth. We further review the development of anti-virulence drugs, and targeting Candida morphogenesis as a novel therapeutic strategy to target fungal pathogens. Together, this review highlights important connections between fungal morphogenesis, virulence, and susceptibility to antifungals.
Collapse
Affiliation(s)
- Jehoshua Sharma
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Sierra Rosiana
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Iqra Razzaq
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
49
|
Polvi EJ, Veri AO, Liu Z, Hossain S, Hyde S, Kim SH, Tebbji F, Sellam A, Todd RT, Xie JL, Lin ZY, Wong CJ, Shapiro RS, Whiteway M, Robbins N, Gingras AC, Selmecki A, Cowen LE. Functional divergence of a global regulatory complex governing fungal filamentation. PLoS Genet 2019; 15:e1007901. [PMID: 30615616 PMCID: PMC6336345 DOI: 10.1371/journal.pgen.1007901] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 01/17/2019] [Accepted: 12/16/2018] [Indexed: 01/17/2023] Open
Abstract
Morphogenetic transitions are prevalent in the fungal kingdom. For a leading human fungal pathogen, Candida albicans, the capacity to transition between yeast and filaments is key for virulence. For the model yeast Saccharomyces cerevisiae, filamentation enables nutrient acquisition. A recent functional genomic screen in S. cerevisiae identified Mfg1 as a regulator of morphogenesis that acts in complex with Flo8 and Mss11 to mediate transcriptional responses crucial for filamentation. In C. albicans, Mfg1 also interacts physically with Flo8 and Mss11 and is critical for filamentation in response to diverse cues, but the mechanisms through which it regulates morphogenesis remained elusive. Here, we explored the consequences of perturbation of Mfg1, Flo8, and Mss11 on C. albicans morphogenesis, and identified functional divergence of complex members. We observed that C. albicans Mss11 was dispensable for filamentation, and that overexpression of FLO8 caused constitutive filamentation even in the absence of Mfg1. Harnessing transcriptional profiling and chromatin immunoprecipitation coupled to microarray analysis, we identified divergence between transcriptional targets of Flo8 and Mfg1 in C. albicans. We also established that Flo8 and Mfg1 cooperatively bind to promoters of key regulators of filamentation, including TEC1, for which overexpression was sufficient to restore filamentation in the absence of Flo8 or Mfg1. To further explore the circuitry through which Mfg1 regulates morphogenesis, we employed a novel strategy to select for mutations that restore filamentation in the absence of Mfg1. Whole genome sequencing of filamentation-competent mutants revealed chromosome 6 amplification as a conserved adaptive mechanism. A key determinant of the chromosome 6 amplification is FLO8, as deletion of one allele blocked morphogenesis, and chromosome 6 was not amplified in evolved lineages for which FLO8 was re-located to a different chromosome. Thus, this work highlights rewiring of key morphogenetic regulators over evolutionary time and aneuploidy as an adaptive mechanism driving fungal morphogenesis. Fungal infections pose a severe burden to human health worldwide. Candida albicans is a leading cause of systemic fungal infections, with mortality rates approaching 40%. One of the key virulence traits of this fungus is its ability to transition between yeast and filamentous forms in response to diverse host-relevant cues. The model yeast Saccharomyces cerevisiae is also capable of filamentous growth in certain conditions, and previous work has identified a key transcriptional complex required for filamentation in both species. However, here we discover that the circuitry governed by this complex in C. albicans is largely distinct from that in the non-pathogenic S. cerevisiae. We also employ a novel selection strategy to perform experimental evolution, identifying chromosome triplication as a mechanism to restore filamentation in a non-filamentous mutant. This work reveals unique circuitry governing a key virulence trait in a leading fungal pathogen, identifying potential therapeutic targets to combat these life-threatening infections.
Collapse
Affiliation(s)
- Elizabeth J. Polvi
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Amanda O. Veri
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Zhongle Liu
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Saif Hossain
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Sabrina Hyde
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Sang Hu Kim
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Faiza Tebbji
- Infectious Disease Research Centre, Université Laval, Quebec, Canada
| | - Adnane Sellam
- Infectious Disease Research Centre, Université Laval, Quebec, Canada
| | - Robert T. Todd
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Jinglin L. Xie
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Zhen-Yuan Lin
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Ontario, Canada
| | - Cassandra J. Wong
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Ontario, Canada
| | - Rebecca S. Shapiro
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | | | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Ontario, Canada
| | - Anna Selmecki
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
50
|
Dioscin Inhibits Virulence Factors of Candida albicans. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4651726. [PMID: 30598996 PMCID: PMC6287159 DOI: 10.1155/2018/4651726] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/23/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023]
Abstract
Candida albicans infections present a heavy burden upon public health, with only a few drugs available, while biofilms formed by C. albicans worsen this situation. Dioscin has antitumor, anti-inflammatory, and hepatoprotective effects, and this study was conducted to evaluate the effects of dioscin on the biofilm formation and development, as well as other virulence factors of C. albicans such as morphological transition, adhesion, and extracellular secreted phospholipase. Our results showed dioscin inhibits these virulence factors and has low cytotoxicity against mammalian cells. Considering protective effects of dioscin against damage on liver and kidney, dioscin may be used as a potential candidate for antifungal development.
Collapse
|