1
|
Kim YG, Jeon H, Boya BR, Lee JH, Lee J. Targeting biofilm formation in Candida albicans with halogenated pyrrolopyrimidine derivatives. Eur J Med Chem 2025; 290:117528. [PMID: 40121868 DOI: 10.1016/j.ejmech.2025.117528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Growing concern over environmental contaminants, including pharmaceuticals and antifungal agents, highlights their role in promoting resistance and biofilm formation by microorganisms. Antifungal resistance, especially in drug-resistant Candida spp., poses a global threat, worsened by the widespread use of antifungal agents in both clinical applications and environmental contamination. This study investigates the antibiofilm properties of various halogenated pyrrolo pyrimidine derivatives, specifically 4-chloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (10) and 2,4-dichloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (16), against fluconazole-resistant C. albicans. Both compounds demonstrated strong biofilm inhibition, with 16 showing greater efficacy even at lower concentrations. qRT-PCR analysis revealed downregulation of key biofilm- and hyphae/germ tube-relating genes, including ALS3, HWP1, and ECE1, alongside upregulation of stress response and biofilm regulator genes such as CDR11, GST3, IFD6, UCF1, YWP1, and ZAP1, indicating complex regulatory responses to the treatments. Molecular docking analysis revealed that these compounds bind effectively to the binding cavity of the ALS3 protein, with halogen atoms playing a key role in stabilizing interaction. Compound 16 exhibited minimal cytotoxicity in Brassica rapa and Caenorhabditis elegans models, suggesting a favorable ADMET safety profile. Confocal microscopy analysis confirmed the compounds effectiveness in preventing biofilm formation when applied as biodegradable PLGA coatings on biomaterial surfaces. These findings suggest that 16 holds promise as a potent antifungal agent with reduced environmental impact, offering both efficacy and sustainability.
Collapse
Affiliation(s)
- Yong-Guy Kim
- The Institute of Clean Technology, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Hyejin Jeon
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Bharath Reddy Boya
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Jin-Hyung Lee
- The Institute of Clean Technology, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea.
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea.
| |
Collapse
|
2
|
Fayed B. Insights into Candida auris dispersal cells and their impact on antifungal resistance. BMC Microbiol 2025; 25:342. [PMID: 40442589 PMCID: PMC12121289 DOI: 10.1186/s12866-025-04055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/19/2025] [Indexed: 06/02/2025] Open
Abstract
The emerging Candidozyma auris (formerly known as Candida auris, C. auris) has caused several outbreaks globally. While several studies explored the resistant biofilm formed by C. auris, little is known regarding the cells dispersed following biofilm formation. Herein, I investigated and characterized the cells dispersed from C. auris biofilms. Cells dispersed from biofilm developed in 96 well plate were isolated and counted. The antifungal susceptibility testing showed that the dispersed cells display similar antifungal susceptibility as the parent planktonic cells, except amphotericin B. Gene expression analysis performed by quantitative real-time PCR indicated that dispersed cells can express genes coded for antifungal resistance (ERG2, ERG6, ERG11, FKS1, CHS1, CHS2, CDR1, MDR1) more than the parent planktonic cells. It was observed that dispersed cells can acquire resistance to caspofungin faster than the parent planktonic cells once exposed to caspofungin at sub MIC level. Furthermore, biofilms formed by dispersed cells displayed significantly higher metabolic activity, as indicated by the XTT analysis. To provide more insight, I explored the expression of genes coding for biofilm initiation and maturation and the data obtained indicated that dispersed cells overexpressed ALS5 and KRE6 genes. Further, GC-MS analysis indicated that dispersed cells exhibit altered metabolic profile that enhance cells survivability under stress and nutrient limit condition. The presented study is the first to explore C. auris dispersed cells and indicated that they are not able to revert to the planktonic mode once released from the biofilm.
Collapse
Affiliation(s)
- Bahgat Fayed
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El Bohouth Street, P.O. 12622, Dokki, Giza, Egypt.
| |
Collapse
|
3
|
Wei Y, Zhao X, Li C, Fu J, Gao W, Mao X. Vacuole and mitochondria patch protein Mcp1 of Saccharomyces boulardii impairs the oxidative stress response of Candida albicans by regulating 2-phenylethanol. Microb Cell Fact 2025; 24:92. [PMID: 40259315 PMCID: PMC12013155 DOI: 10.1186/s12934-025-02721-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Vacuole and mitochondria patch (vCLAMP) protein Mcp1 is crucial in eukaryotic cells response to environmental stress, but the mechanism of Mcp1 in Saccharomyces boulardii (S. boulardii) against pathogenic fungi is unclear. RESULTS This work first explored the role of Mcp1 in S. boulardii against Candida albicans (C. albicans). The results showed that Mcp1 located on the vacuolar and mitochondrial membrane of S. boulardii. Overexpression of Mcp1 inhibited the adhesion and hyphal formation of C. albicans in vitro. The mice model of intestinal infection revealed that WT-pGK1-MCP1 mutant enhanced the ability of S. boulardii antagonize C. albicans infecting gut. High performance liquid chromatography-mass spectrometry analysis demonstrated that overexpressing Mcp1 promoted the production of 2-phenylethanol. The latter is a secondary metabolite of S. boulardii, and can inhibit the adhesion and biofilm formation of C. albicans. The reverse transcription polymerase chain reaction and western blotting results confirmed Mcp1 promoted the production of 2-phenylethanol by regulating the expression level of Aro10. Notably, RNA-sequencing and Gene Ontology enrichment analyses showed that 2-phenylethanol impaired the oxidative stress response of C. albicans. CONCLUSION This work reveals the critical role of Mcp1 in S. boulardii against C. albicans by regulating 2-phenylethanol metabolism, which provide a theoretical basis for S. boulardii as antifungal biologic therapy to prevent and treat of Candida infection.
Collapse
Affiliation(s)
- Yunyun Wei
- Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, 250000, Shandong, China
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Xiaohui Zhao
- The Second Affiliated Hospital of Shandong First Medical University& Shandong Academy of Medical Sciences, Tai'an, China
| | - Chuanqi Li
- Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, 250000, Shandong, China
| | - Jianhao Fu
- Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, 250000, Shandong, China
| | - Wanli Gao
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaolong Mao
- Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, 250000, Shandong, China.
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
4
|
Lim SJ, Muhd Noor ND, Sabri S, Mohamad Ali MS, Salleh AB, Oslan SN. Features of the rare pathogen Meyerozyma guilliermondii strain SO and comprehensive in silico analyses of its adherence-contributing virulence factor agglutinin-like sequences. J Biomol Struct Dyn 2025; 43:3728-3748. [PMID: 38189364 DOI: 10.1080/07391102.2023.2300757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/17/2023] [Indexed: 01/09/2024]
Abstract
Meyerozyma guilliermondii is a rare yeast pathogen contributing to the deadly invasive candidiasis. M. guilliermondii strain SO, as a promising protein expression host, showed 99% proteome similarity with the clinically isolated ATCC 6260 (type strain) in a recent comparative genomic analysis. However, their in vitro virulence features and in vivo pathogenicity were uncharacterized. This study aimed to characterize the in vitro and in vivo pathogenicity of M. guilliermondii strain SO and analyze its Als proteins (MgAls) via comprehensive bioinformatics approaches. M. guilliermondii strain SO showed lower and higher sensitivity towards β-mercaptoethanol and lithium, respectively than the avirulent S. cerevisiae but exhibited the same tolerance towards cell wall-perturbing Congo Red with C. albicans. With 7.5× higher biofilm mass, M. guilliermondii strain SO also demonstrated 75% higher mortality rate in the zebrafish embryos with a thicker biofilm layer on the chorion compared to the avirulent S. cerevisiae. Being one of the most important Candida adhesins, sequence and structural analyses of four statistically identified MgAls showed that MgAls1056 was predicted to exhibit the most conserved amyloid-forming regions, tandem repeat domain and peptide binding cavity (PBC) compared to C. albicans Als3. Favoured from the predicted largest ligand binding site and druggable pockets, it showed the highest affinity towards hepta-threonine. Non-PBC druggable pockets in the most potent virulence contributing MgAls1056 provide new insights into developing antifungal drugs targeting non-albicans Candida spp. Virtual screening of available synthetic or natural bioactive compounds and MgAls1056 deletion from the fungal genome should be further performed and validated experimentally.
Collapse
Affiliation(s)
- Si Jie Lim
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Noor Dina Muhd Noor
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Suriana Sabri
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Mohd Shukuri Mohamad Ali
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Abu Bakar Salleh
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Siti Nurbaya Oslan
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| |
Collapse
|
5
|
Liu S, Wang Y, Xu C. Suppressive effects of lemon myrtle extract against the colonization and virulence factors of Candida spp. J Oral Biosci 2025:100657. [PMID: 40127778 DOI: 10.1016/j.job.2025.100657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVES Candida species (Candida spp.) are among the most common opportunistic pathogens inhabiting the oral cavity and frequently cause infection in immunocompromised individuals. Conventional antibiotic treatments for Candida infections face significant challenges, including the emergence of antimicrobial resistance. This highlights the urgent need for alternative therapeutic strategies, particularly those leveraging natural products. METHODS In this study, we evaluated the inhibitory effects of an aqueous lemon myrtle extract on the colonization and virulence of six Candida spp., including microbial adhesion, biofilm formation, extracellular polysaccharide production, hyphal production, and several invasion-associated virulence factors. RESULTS The extract significantly reduced Candida adhesion to hard surfaces and inhibited biofilm formation. Additionally, it suppressed the production of insoluble extracellular polysaccharides and various invasion-associated virulence factors, including phospholipase, ergosterol, protease, and hyphal formation. CONCLUSIONS These findings provide a better understanding of the potential role of lemon myrtle extract as a natural therapeutic agent for controlling Candida colonization and mitigating its invasive capabilities. This study provides a foundation for further exploration of lemon myrtle as a promising alternative for the management of Candida infections.
Collapse
Affiliation(s)
- Siyuan Liu
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Yi Wang
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Chun Xu
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| |
Collapse
|
6
|
Bekkal Brikci-Benhabib O. Navigating dual-species fungal biofilms: The competitive and cooperative dynamics of Candidaalbicans. Res Microbiol 2025; 176:104262. [PMID: 39615641 DOI: 10.1016/j.resmic.2024.104262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 03/24/2025]
Abstract
Research on microbial biofilms has primarily concentrated on bacterial-bacterial and bacterial-fungal interactions, leaving fungal-fungal dynamics underexplored. The present study examines interactions within dual-species fungal biofilms, with a particular emphasis on Candida albicans. The behavior and pathogenicity of this yeast are significantly influenced by its interactions with other fungal species in biofilms, where its ability to shift between yeast and hyphal forms contributes significantly to biofilm formation. These fungal species biofilms exhibit a complex interplay of synergistic cooperation and antagonistic competition, depending on the environmental context and resource availability. Understanding these interactions is essential for advancing our knowledge of fungal biofilm.
Collapse
|
7
|
Zhang H, Zhang Q, Zuo T, Wang Z, Liao J, Lu Y. 2-Chloromethyl anthraquinone inhibits Candida albicans biofilm formation by inhibiting the Ras1-cAMP-Efg1 pathway. Res Microbiol 2025:104280. [PMID: 40024356 DOI: 10.1016/j.resmic.2025.104280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Candida albicans is an opportunistic pathogen, and the formation of its biofilm makes it resistant to traditional antifungal therapy. Anthraquinones have universal antibacterial activity. We evaluated the inhibitory effects of 2-chloromethyl anthraquinone on C. albicans adhesion, mycelial morphology transformation, and biofilm formation. The results showed that 2-chloromethyl anthraquinone could inhibit C. albicans adhesion, mycelium formation, and biofilm formation in a dose-dependent manner at 2 μg/mL. In addition, 2-chloromethyl anthraquinone significantly inhibited the expression of biofilm formation-related genes in C. albicans, including ALS1, CPH1, ECE1, HWP1, TEC1, BCR1, and UME6. In addition, Ras1-cAMP-Efg1 pathway-related genes (RAC1, CYR1, and TPK2) were also significantly down-regulated, indicating that the inhibitory effect of 2-chloromethyl anthraquinone on C. albicans biofilms may be related to the Ras1-cAMP-Efg1 signaling pathway. In summary, the results of this study confirmed the inhibitory mechanism of 2-chloromethyl anthraquinone on the virulence factors of C. albicans, which laid a theoretical foundation for its use as an anti-biofilm agent against C. albicans.
Collapse
Affiliation(s)
- Haoying Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ting Zuo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziqi Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Liao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
8
|
Harine A, Ranjani S, Hemalatha S. Antifungal efficacy of Citrusfusion mediated silver nanoparticles in Candida species. BMC Biotechnol 2025; 25:18. [PMID: 39979871 PMCID: PMC11841014 DOI: 10.1186/s12896-025-00952-y] [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: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Candida species are commensal fungi that can become opportunistic pathogens under specific host and environmental conditions. The emergence of multidrug-resistant Candida strains poses a significant challenge. Nanotechnology represents a cutting-edge field offering precise and targeted delivery systems for combating fungal infections, leveraging the unique properties of plant-derived bioactive compounds. This investigation employed a biogenic approach utilizing polyherbal leaf extracts from Citrus limon and Citrus medica, known for their abundant Citral content. RESULTS Citrus sp. extracts were used to synthesize Citrusfusion silver nanoparticles (CitAgNPs) through a green synthesis method. Characterization of CitAgNPs was carried out using advanced analytical methods ensuring the quality, uniformity, size, and charge. The synthesized CitAgNPs exhibited non toxic effect when tested on Vigna radiata and Danio rerio, highlighting their potential for sustainability and safe therapeutic use. Antifungal assays demonstrated the potent efficacy of CitAgNPs in various Candida strains, with low MIC and MFC. CitAgNPs exhibited remarkable biofilm inhibition capabilities and elucidated specific mechanisms of action in Candida species, surpassing the performance of fluconazole. CONCLUSION This study underscores the immense potential of nanotechnology-driven approaches harnessing Citrus leaf extract for synthesizing highly effective antifungal nanoparticles. The fusion of biogenic nanoparticles with Citrus bioactive compounds presents a sustainable strategy for addressing the escalating challenge of azole-resistant Candida infections. The research outcomes suggest that CitAgNPs have promising applications in inhibiting Candida biofilms, offering potential solutions for infections caused by diaper rashes and onychomycosis, providing safe and effective alternatives to antifungal therapies.
Collapse
Affiliation(s)
- A Harine
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, Tamil Nadu, 600048, India
| | - S Ranjani
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, Tamil Nadu, 600048, India
| | - S Hemalatha
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, Tamil Nadu, 600048, India.
| |
Collapse
|
9
|
Niu H, Zhu D, Leng J, Wang Z, Liu D, Chen Y, Yang P, Ying H. Biofilm-based immobilized fermentation of engineered Komagataella phaffii for xylanase production. BIORESOURCE TECHNOLOGY 2025; 418:131918. [PMID: 39622419 DOI: 10.1016/j.biortech.2024.131918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
This study presented an immobilized fermentation process of engineered Komagataella phaffii with improved biofilm-forming abilities for continuous xylanase production and provided the first insights into the molecular basis of biofilm-based immobilized fermentation of K. phaffii. Overexpression of PAS_chr2-2_0178 and PAS_FragB_0067 in K. phaffii facilitated biofilm formation with 31.6% and 113.8% increasement, respectively. Subsequently, a biofilm-based immobilized fermentation process was developed for the PAS_FragB_0067-overexpressing strain. Xylanase production over five batches by GS115-0067* was better than that of GS115-xyn, with an overall average of 35.4 % higher enzyme activity. PAS_FragB_0067 overexpression resulted in better adhesion of K. phaffii cells on the carrier, and enhanced biofilms could provide more active cells in the immobilized fermentation process. Transcriptome analysis revealed that overexpression of the biofilm-related gene promoted central carbon metabolism. These findings offer a valuable reference strategy to improve production efficiency of K. phaffii cells in continuous fermentation processes.
Collapse
Affiliation(s)
- Huanqing Niu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China.
| | - Daoguang Zhu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Jing Leng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zhenyu Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Dong Liu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Yong Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Pengpeng Yang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Hanjie Ying
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
10
|
Cosio T, Romeo A, Pistoia ES, Pica F, Freni C, Iacovelli F, Orlandi A, Falconi M, Campione E, Gaziano R. Retinoids as Alternative Antifungal Agents Against Candida albicans: In Vitro and In Silico Evidence. Microorganisms 2025; 13:237. [PMID: 40005604 PMCID: PMC11857849 DOI: 10.3390/microorganisms13020237] [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: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Candida albicans (C. albicans) is the most common pathogen responsible for a wide spectrum of human infections ranging from superficial mucocutaneous mycoses to systemic life-threatening diseases. Its main virulence factors are the morphological transition between yeast and hyphal forms and the ability to produce biofilm. Novel antifungal strategies are required given the severity of systemic candidiasis, especially in immunocompromised patients, and the lack of effective anti-biofilm treatments. We previously demonstrated that all-trans retinoic acid (ATRA), an active metabolite of vitamin A, exerted an inhibitory effect on Candida growth, yeast-hyphal transition and biofilm formation. Here, we further investigated the possible anti-Candida potential of trifarotene and tazarotene, which are the other two molecules belonging to the retinoid family, compared to ATRA. The results indicate that both drugs were able to suppress Candida growth, germination and biofilm production, although trifarotene was proven to be more effective than tazarotene, showing effectiveness comparable to ATRA. In silico studies suggest that all three retinoids may exert antifungal activity through their molecular interactions with the heat shock protein (Hsp) 90 and 14α-demethylase of C. albicans. Moreover, interactions between retinoids and ergosterol have been observed, suggesting that those compounds have great potential against C. albicans infections.
Collapse
Affiliation(s)
- Terenzio Cosio
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Alice Romeo
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Enrico Salvatore Pistoia
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
| | - Francesca Pica
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
| | - Claudia Freni
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Augusto Orlandi
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Mattia Falconi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (A.R.); (C.F.); (F.I.); (M.F.)
| | - Elena Campione
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (T.C.); (E.S.P.); (F.P.)
| |
Collapse
|
11
|
Jørgensen MR. Pathophysiological microenvironments in oral candidiasis. APMIS 2024; 132:956-973. [PMID: 38571459 DOI: 10.1111/apm.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024]
Abstract
Oral candidiasis (OC), a prevalent opportunistic infection of the oral mucosa, presents a considerable health challenge, particularly in individuals with compromised immune responses, advanced age, and local predisposing conditions. A considerable part of the population carries Candida in the oral cavity, but only few develop OC. Therefore, the pathogenesis of OC may depend on factors other than the attributes of the fungus, such as host factors and other predisposing factors. Mucosal trauma and inflammation compromise epithelial integrity, fostering a conducive environment for fungal invasion. Molecular insights into the immunocompromised state reveal dysregulation in innate and adaptive immunity, creating a permissive environment for Candida proliferation. Detailed examination of Candida species (spp.) and their virulence factors uncovers a nuanced understanding beyond traditional C. albicans focus, which embrace diverse Candida spp. and their strategies, influencing adhesion, invasion, immune evasion, and biofilm formation. Understanding the pathophysiological microenvironments in OC is crucial for the development of targeted therapeutic interventions. This review aims to unravel the diverse pathophysiological microenvironments influencing OC development focusing on microbial, host, and predisposing factors, and considers Candida resistance to antifungal therapy. The comprehensive approach offers a refined perspective on OC, seeking briefly to identify potential therapeutic targets for future effective management.
Collapse
Affiliation(s)
- Mette Rose Jørgensen
- Section of Oral Pathology and Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Aydin M, Unusan N, Sumlu E, Korucu EN. Rosmarinic Acid Exhibits Antifungal and Antibiofilm Activities Against Candida albicans: Insights into Gene Expression and Morphological Changes. J Fungi (Basel) 2024; 10:751. [PMID: 39590670 PMCID: PMC11595412 DOI: 10.3390/jof10110751] [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: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Candida species, opportunistic pathogens that cause various infections, pose a significant threat due to their ability to form biofilms that resist antifungal treatments and immune responses. The increasing resistance of Candida spp. and the limited availability of effective treatments have prompted the research of natural compounds as alternative therapies. This study assessed the antifungal properties of RA against Candida species, focusing on its impact on C. albicans biofilms and the underlying mechanisms. The antifungal efficacy of RA was evaluated using the CLSI M27-A3 microdilution method on both fluconazole-susceptible and -resistant strains. Biofilm formation by C. albicans was assessed through a crystal violet assay, while its antibiofilm activity was analyzed using an MTT assay and field emission scanning electron microscopy (FESEM). Gene expression related to biofilm formation was studied using quantitative real-time PCR (qRT-PCR), and statistical analysis was performed with an ANOVA. Among the 28 Candida strains tested, RA exhibited minimum inhibitory concentration (MIC) values ranging from 160 to 1280 μg/mL. At a 640 μg/mL concentration, it significantly reduced the expression of genes associated with adhesion (ALS3, HWP1, and ECE1), hyphal development (UME6 and HGC1), and hyphal cAMP-dependent protein kinase regulators (CYR1, RAS1, and EFG1) in RAS1-cAMP-EFG1 pathway (p < 0.05). FESEM analysis revealed a reduction in hyphal networks and disruptions on the cell surface. Our study is the first to demonstrate the effects of RA on C. albicans adhesion, hyphae development, and biofilm formation through gene expression analysis with findings supported by FESEM. This approach distinguishes our study from previous studies on the effect of RA on Candida. However, the high MIC values of RA limit its antifungal potential. Therefore, more extensive research using innovative methods is required to increase the antifungal effect of RA.
Collapse
Affiliation(s)
- Merve Aydin
- Department of Medical Microbiology, Faculty of Medicine, KTO Karatay University, Konya 42020, Turkey
| | - Nurhan Unusan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, KTO Karatay University, Konya 42020, Turkey;
| | - Esra Sumlu
- Department of Medical Pharmacology, Faculty of Medicine, KTO Karatay University, Konya 42020, Turkey;
| | - Emine Nedime Korucu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya 42090, Turkey;
| |
Collapse
|
13
|
Aldulaijan S, Alruwili R, Almulaify R, Alhassan FA, Al-Dulaijan YA, Alshahrani FA, Mokeem L, Gad MM, Melo MAS, Balhaddad AA. Benzyldimethyldodecyl Ammonium Chloride-Doped Denture-Based Resin: Impact on Strength, Surface Properties, Antifungal Activities, and In Silico Molecular Docking Analysis. J Funct Biomater 2024; 15:310. [PMID: 39452608 PMCID: PMC11508443 DOI: 10.3390/jfb15100310] [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: 09/06/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Candida albicans (C. albicans) adhering to denture-based resins (DBRs) is a known cause of denture stomatitis. A new approach to prevent denture stomatitis is to include antimicrobial substances within DBRs. Here, we examined the mechanical performance and antifungal properties of DBRs containing benzyldimethyldodecyl ammonium chloride (C12BDMA-Cl) as an antimicrobial compound. C12BDMA-Cl is a quaternary ammonium compound, and its antifungal properties have never been investigated when combined with dental acrylic resin. Therefore, we modified a commercially available heat-polymerized acrylic DBR to contain 3 and 5 wt.% of C12BDMA-Cl. Unmodified DBR was used as a control group. Specimens were prepared using the conventional heat processing method. The specimen's flexural strength, elastic modulus, microhardness, and surface roughness were evaluated. C. albicans biofilm was grown on the specimens and assessed via colony-forming units (CFUs) and scanning electron microscopy (SEM). In silico molecular docking was applied to predict the potential C12BDMA-Cl inhibition activity as an antifungal drug. The 3% C12BDMA-Cl DBR demonstrated antifungal activities without a deterioration effect on the mechanical performance. SEM images indicated fewer colonies in DBR containing C12BDMA-Cl, which can be a potential approach to managing denture stomatitis. In conclusion, C12BDMA-Cl is a promising antifungal agent for preventing and treating denture stomatitis.
Collapse
Affiliation(s)
- Sarah Aldulaijan
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Raghad Alruwili
- College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (F.A.A.)
| | - Rawan Almulaify
- College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (F.A.A.)
| | - Fatimah A. Alhassan
- College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (F.A.A.)
| | - Yousif A. Al-Dulaijan
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (M.M.G.)
| | - Faris A. Alshahrani
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (M.M.G.)
| | - Lamia Mokeem
- Department of Restorative Dentistry, College of Medicine and Dentistry, Riyadh Elm University, Riyadh 13244, Saudi Arabia
| | - Mohammed M. Gad
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia (M.M.G.)
| | - Mary Anne S. Melo
- Department of Comprehensive Dentistry, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Abdulrahman A. Balhaddad
- Department of Restorative Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
14
|
Nayak A, Khedri A, Chavarria A, Sanders KN, Ghalei H, Khoshnevis S. Sinefungin, a natural nucleoside analog of S-adenosyl methionine, impairs the pathogenicity of Candida albicans. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:23. [PMID: 39268078 PMCID: PMC11391927 DOI: 10.1038/s44259-024-00040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/19/2024] [Indexed: 09/15/2024]
Abstract
Candida albicans, an opportunistic fungal pathogen, causes life-threatening infections in immunocompromised patients. Current antifungals are limited by toxicity, drug-drug interactions, and emerging resistance, underscoring the importance of identifying novel treatment approaches. Here, we elucidate the impact of sinefungin, an analog of S-adenosyl methionine, on the virulence of C. albicans strain SC5314 and clinical isolates. Our data indicate that sinefungin impairs pathogenic traits of C. albicans including hyphal morphogenesis, biofilm formation, adhesion to epithelial cells, and virulence towards Galleria mellonella, highlighting sinefungin as an avenue for therapeutic intervention. We determine that sinefungin particularly disturbs N6-methyladenosine (m6A) formation. Transcriptome analysis of C. albicans hyphae upon sinefungin treatment reveals an increase in transcripts related to the yeast form and decrease in those associated with hyphae formation and virulence. Collectively, our data propose sinefungin as a potent molecule against C. albicans and emphasize further exploration of post-transcriptional control mechanisms of pathogenicity for antifungal design.
Collapse
Affiliation(s)
- Anushka Nayak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Azam Khedri
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Alejandro Chavarria
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Kyla N. Sanders
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Sohail Khoshnevis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
15
|
Fayed B, Shakartalla SB, Sabbah H, Dalle H, Tannira M, Senok A, Soliman SSM. Transcriptome Analysis of Human Dermal Cells Infected with Candida auris Identified Unique Pathogenesis/Defensive Mechanisms Particularly Ferroptosis. Mycopathologia 2024; 189:65. [PMID: 38990436 DOI: 10.1007/s11046-024-00868-9] [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: 12/23/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Candida auris is an emerging multi-drug resistant yeast that can cause life-threatening infections. A recent report clarified the ability of C. auris to form a biofilm with enhanced drug resistance properties in the host skin's deep layers. The formed biofilm may initiate further bloodstream spread and immune escape. Therefore, we propose that secreted chemicals from the biofilm may facilitate fungal pathogenesis. In response to this interaction, the host skin may develop potential defensive mechanisms. Comparative transcriptomics was performed on the host dermal cells in response to indirect interaction with C. auris biofilm through Transwell inserts compared to planktonic cells. Furthermore, the effect of antifungals including caspofungin and fluconazole was studied. The obtained data showed that the dermal cells exhibited different transcriptional responses. Kyoto Encyclopedia of Genes and Genomes and Reactome analyses identified potential defensive responses employed by the dermal cells and potential toxicity induced by C. auris. Additionally, our data indicated that the dominating toxic effect was mediated by ferroptosis; which was validated by qRT-PCR, cytotoxicity assay, and flow cytometry. On the other hand, the viability of C. auris biofilm was enhanced and accompanied by upregulation of MDR1, and KRE6 upon interaction with dermal cells; both genes play significant roles in drug resistance and biofilm maturation, respectively. This study for the first-time shed light on the dominating defensive responses of human dermal cells, microbe colonization site, to C. auris biofilm and its toxic effects. Further, it demonstrates how C. auris biofilm responds to the defensive mechanisms developed by the human dermal cells.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Chemistry of Natural and Microbial Products, National Research Centre, Cairo, Egypt
| | - Sarra B Shakartalla
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Faculty of Pharmacy, University of Gezira, P.O.Box. 21111, Wad Medani, Sudan
| | - Hassan Sabbah
- AbbVie BioPharmaceuticals, P.O. Box 118052, Dubai, UAE
| | - Hala Dalle
- AbbVie BioPharmaceuticals, Kuwait City, Kuwait
| | | | - Abiola Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14 Dubai Healthcare City, P.O.Box 505055, Dubai, UAE
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
| |
Collapse
|
16
|
Kumar D, Kumar A. Molecular Determinants Involved in Candida albicans Biofilm Formation and Regulation. Mol Biotechnol 2024; 66:1640-1659. [PMID: 37410258 DOI: 10.1007/s12033-023-00796-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
Candida albicans is known for its pathogenicity, although it lives within the human body as a commensal member. The commensal nature of C. albicans is well controlled and regulated by the host's immune system as they live in the harmonized microenvironment. However, the development of certain unusual microhabitat conditions (change in pH, co-inhabiting microorganisms' population ratio, debilitated host-immune system) pokes this commensal fungus to transform into a pathogen in such a way that it starts to propagate very rapidly and tries to breach the epithelial barrier to enter the host's systemic circulations. In addition, Candida is infamous as a major nosocomial (hospital-acquired infection) agent because it enters the human body through venous catheters or medical prostheses. The hysterical mode of C. albicans growth builds its microcolony or biofilm, which is pathogenic for the host. Biofilms propose additional resistance mechanisms from host immunity or extracellular chemicals to aid their survival. Differential gene expressions and regulations within the biofilms cause altered morphology and metabolism. The genes associated with adhesiveness, hyphal/pseudo-hyphal growth, persister cell transformation, and biofilm formation by C. albicans are controlled by myriads of cell-signaling regulators. These genes' transcription is controlled by different molecular determinants like transcription factors and regulators. Therefore, this review has focused discussion on host-immune-sensing molecular determinants of Candida during biofilm formation, regulatory descriptors (secondary messengers, regulatory RNAs, transcription factors) of Candida involved in biofilm formation that could enable small-molecule drug discovery against these molecular determinants, and lead to disrupt the well-structured Candida biofilms effectively.
Collapse
Affiliation(s)
- Dushyant Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, 492010, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, 492010, India.
| |
Collapse
|
17
|
Wang SH, Zheng T, Fawzi NL. Structure and interactions of prion-like domains in transcription factor Efg1 phase separation. Biophys J 2024; 123:1481-1493. [PMID: 38297837 PMCID: PMC11163291 DOI: 10.1016/j.bpj.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 02/02/2024] Open
Abstract
Candida albicans, a prominent member of the human microbiome, can make an opportunistic switch from commensal coexistence to pathogenicity accompanied by an epigenetic shift between the white and opaque cell states. This transcriptional switch is under precise regulation by a set of transcription factors (TFs), with Enhanced Filamentous Growth Protein 1 (Efg1) playing a central role. Previous research has emphasized the importance of Efg1's prion-like domain (PrLD) and the protein's ability to undergo phase separation for the white-to-opaque transition of C. albicans. However, the underlying molecular mechanisms of Efg1 phase separation have remained underexplored. In this study, we delved into the biophysical basis of Efg1 phase separation, revealing the significant contribution of both N-terminal (N) and C-terminal (C) PrLDs. Through NMR structural analysis, we found that Efg1 N-PrLD and C-PrLD are mostly disordered but have prominent partial α-helical secondary structures in both domains. NMR titration experiments suggest that the partially helical structures in N-PrLD act as hubs for self-interaction as well as Efg1 interaction with RNA. Using condensed-phase NMR spectroscopy, we uncovered diverse amino acid interactions underlying Efg1 phase separation. Particularly, we highlight the indispensable role of tyrosine residues within the transient α-helical structures of PrLDs particularly in the N-PrLD compared to the C-PrLD in stabilizing phase separation. Our study provides evidence that the transient α-helical structure is present in the phase-separated state and highlights the particular importance of aromatic residues within these structures for phase separation. Together, these results enhance the understanding of C. albicans transcription factor interactions that lead to virulence and provide a crucial foundation for potential antifungal therapies targeting the transcriptional switch.
Collapse
Affiliation(s)
- Szu-Huan Wang
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island
| | - Tongyin Zheng
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island.
| | - Nicolas L Fawzi
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island.
| |
Collapse
|
18
|
Xu Z, Li Y, Xu A, Xue L, Soteyome T, Yuan L, Ma Q, Seneviratne G, Hong W, Mao Y, Kjellerup BV, Liu J. Differential alteration in Lactiplantibacillus plantarum subsp. plantarum quorum-sensing systems and reduced Candida albicans yeast survival and virulence gene expression in dual-species interaction. Microbiol Spectr 2024; 12:e0035324. [PMID: 38717160 PMCID: PMC11237386 DOI: 10.1128/spectrum.00353-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/15/2024] [Indexed: 06/06/2024] Open
Abstract
Candida albicans (C. albicans) and Lactiplantibacillus plantarum subsp. plantarum (L. plantarum) are frequently identified in various niches, but their dual-species interaction, especially with C. albicans in yeast form, remains unclear. This study aimed to investigate the dual-species interaction of L. plantarum and C. albicans, including proliferation, morphology, and transcriptomes examined by selective agar plate counting, microscopy, and polymicrobial RNA-seq, respectively. Maintaining a stable and unchanged growth rate, L. plantarum inhibited C. albicans yeast cell proliferation but not hyphal growth. Combining optical microscopy and atomic force microscopy, cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed during dual-species interaction. Reduced C. albicans yeast cell proliferation in mixed culture was partially due to L. plantarum cell-free culture supernatant but not the acidic environment. Upon polymicrobial transcriptomics analysis, interesting changes were identified in both L. plantarum and C. albicans gene expression. First, two L. plantarum quorum-sensing systems showed contrary changes, with the activation of lamBDCA and repression of luxS. Second, the upregulation of stress response-related genes and downregulation of cell cycle, cell survival, and cell integrity-related pathways were identified in C. albicans, possibly connected to the stress posed by L. plantarum and the reduced yeast cell proliferation. Third, a large scale of pathogenesis and virulence factors were downregulated in C. albicans, indicating the potential interruption of pathogenic activities by L. plantarum. Fourth, partial metabolism and transport pathways were changed in L. plantarum and C. albicans. The information in this study might aid in understanding the behavior of L. plantarum and C. albicans in dual-species interaction.IMPORTANCEThe anti-Candida albicans activity of Lactiplantibacillus plantarum has been explored in the past decades. However, the importance of C. albicans yeast form and the effect of C. albicans on L. plantarum had also been omitted. In this study, the dual-species interaction of L. plantarum and C. albicans was investigated with a focus on the transcriptomes. Cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed. Upon polymicrobial transcriptomics analysis, interesting changes were identified, including contrary changes in two L. plantarum quorum-sensing systems and reduced cell survival-related pathways and pathogenesis determinants in C. albicans.
Collapse
Affiliation(s)
- Zhenbo Xu
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yaqin Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou, China
| | - Liang Xue
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, China, Guangzhou, Guangdong
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qin Ma
- Key Laboratory of Functional Foods, Ministry of Agriculture, Guangdong Key Laboratory of Agricultural Products Processing, Sericultural and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | | | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuzhu Mao
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Birthe V. Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Junyan Liu
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, College of Light Industry and Food Science, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
19
|
Nouraei H, Zare S, Nemati M, Amirzadeh N, Motamedi M, Shabanzadeh S, Zomorodian K, Pakshir K. Comparative analysis of enzymatic profiles and biofilm formation in clinical and environmental Candida kefyr isolates. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e13282. [PMID: 38923398 PMCID: PMC11194042 DOI: 10.1111/1758-2229.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/30/2024] [Indexed: 06/28/2024]
Abstract
The global landscape of Candida infections has seen a significant shift. Previously, Candida albicans was the predominant species. However, there has been an emergence of non-albicans Candida species, which are often less susceptible to antifungal treatment. Candida kefyr, in particular, has been increasingly associated with infections. This study aimed to investigate the profiles of enzymatic activity and biofilm formation in both clinical and non-clinical isolates of C. kefyr. A total of 66 C. kefyr isolates were analysed. The activities of proteinase and phospholipase were assessed using bovine serum albumin and egg yolk agar, respectively. Haemolysin, caseinolytic and esterase activities were evaluated using specific methods. Biofilm formation was investigated using crystal violet staining. The findings indicated that biofilm and proteinase activity were detected in 81.8% and 93.9% of all the isolates, respectively. Haemolysin activity was observed with the highest occurrence (95.5%) among normal microbiota isolates. Esterase activity was predominantly identified in dairy samples and was absent in hospital samples. Caseinase production was found with the highest occurrence (18.2%) in normal microbiota and hospital samples. Phospholipase activity was limited, found in only 3% of all the isolates. These findings reveal variations in enzyme activity between clinical and non-clinical C. kefyr isolates. This sheds light on their pathogenic potential and has implications for therapeutic strategies.
Collapse
Affiliation(s)
- Hasti Nouraei
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Samira Zare
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Maryam Nemati
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Neda Amirzadeh
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Marjan Motamedi
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Shafigheh Shabanzadeh
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Kamiar Zomorodian
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
- Basic Sciences in Infectious Diseases Research Center, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Keyvan Pakshir
- Department of Parasitology and Mycology, School of MedicineShiraz University of Medical SciencesShirazIran
- Basic Sciences in Infectious Diseases Research Center, School of MedicineShiraz University of Medical SciencesShirazIran
| |
Collapse
|
20
|
Patil SB, Basrani ST, Chougule SA, Gavandi TC, Karuppayil SM, Jadhav AK. Butyl isothiocyanate exhibits antifungal and anti-biofilm activity against Candida albicans by targeting cell membrane integrity, cell cycle progression and oxidative stress. Arch Microbiol 2024; 206:251. [PMID: 38727840 DOI: 10.1007/s00203-024-03983-9] [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: 02/15/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/18/2024]
Abstract
The prevalence of Candida albicans infection has increased during the past few years, which contributes to the need for new, effective treatments due to the increasing concerns regarding antifungal drug toxicity and multidrug resistance. Butyl isothiocyanate (butylITC) is a glucosinolate derivative, and has shown a significant antifungal effect contrary to Candida albicans. Additionally, how butylITC affects the virulence traits of C. albicans and molecular mode of actions are not well known. Present study shows that at 17.36 mM concentration butylITC inhibit planktonic growth. butylITC initially slowed the hyphal transition at 0.542 mM concentration. butylITC hampered biofilm development, and inhibits biofilm formation at 17.36 mM concentration which was analysed using metabolic assay (XTT assay) and Scanning Electron Microscopy (SEM). In addition, it was noted that butylITC inhibits ergosterol biosynthesis. The permeability of cell membranes was enhanced by butylITC treatment. Moreover, butylITC arrests cells at S-phase and induces intracellular Reactive Oxygen Species (ROS) accumulation in C. albicans. The results suggest that butylITC may have a dual mode of action, inhibit virulence factors and modulate cellular processes like inhibit ergosterol biosynthesis, cell cycle arrest, induces ROS production which leads to cell death in C. albicans.
Collapse
Affiliation(s)
- Shivani Balasaheb Patil
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416003, Maharashtra, India
| | - Sargun Tushar Basrani
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416003, Maharashtra, India
| | - Sayali Ashok Chougule
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416003, Maharashtra, India
| | - Tanjila Chandsaheb Gavandi
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416003, Maharashtra, India
| | - Sankunny Mohan Karuppayil
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416003, Maharashtra, India.
| | - Ashwini Khanderao Jadhav
- Department of Stem Cell and Regenerative Medicine, Medical Biotechnology Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416003, Maharashtra, India.
| |
Collapse
|
21
|
Zhu X, Wang A, Zheng Y, Li D, Wei Y, Gan M, Li Y, Si S. Anti-Biofilm Activity of Cocultimycin A against Candida albicans. Int J Mol Sci 2023; 24:17026. [PMID: 38069349 PMCID: PMC10707031 DOI: 10.3390/ijms242317026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Candida albicans (C. albicans), the most common fungal pathogen, has the ability to form a biofilm, leading to enhanced virulence and antibiotic resistance. Cocultimycin A, a novel antifungal antibiotic isolated from the co-culture of two marine fungi, exhibited a potent inhibitory effect on planktonic C. albicans cells. This study aimed to evaluate the anti-biofilm activity of cocultimycin A against C. albicans and explore its underlying mechanism. Crystal violet staining showed that cocultimycin A remarkably inhibited biofilm formation in a dose-dependent manner and disrupted mature biofilms at higher concentrations. However, the metabolic activity of mature biofilms treated with lower concentrations of cocultimycin A significantly decreased when using the XTT reduction method. Cocultimycin A could inhibit yeast-to-hypha transition and mycelium formation of C. albicans colonies, which was observed through the use of a light microscope. Scanning electron microscopy revealed that biofilms treated with cocultimycin A were disrupted, yeast cells increased, and hypha cells decreased and significantly shortened. The adhesive ability of C. albicans cells treated with cocultimycin A to the medium and HOEC cells significantly decreased. Through the use of a qRT-PCR assay, the expression of multiple genes related to adhesion, hyphal formation and cell membrane changes in relation to biofilm cells treated with cocultimycin A. All these results suggested that cocultimycin A may be considered a potential novel molecule for treating and preventing biofilm-related C. albicans infections.
Collapse
Affiliation(s)
| | | | | | | | | | - Maoluo Gan
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (A.W.); (Y.Z.); (D.L.); (Y.W.); (S.S.)
| | - Yan Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (A.W.); (Y.Z.); (D.L.); (Y.W.); (S.S.)
| | | |
Collapse
|
22
|
Khodavandi P, Hosseini A, Khodavandi A, Alizadeh F, Azizi A, Gerami M. Hyphae-specific genes: Possible molecular targets for magnetic iron oxide nanoparticles alone and combined with visible light in Candida albicans. Photodiagnosis Photodyn Ther 2023; 44:103822. [PMID: 37778716 DOI: 10.1016/j.pdpdt.2023.103822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/03/2023]
Abstract
Candida albicans readily develops resistance to fluconazole. Magnetic iron oxide nanoparticles (denoted as MION) and antimicrobial photodynamic therapy are attracting attention as therapeutic agents. This study aims to investigate the inhibitory efficacy of MION alone and combined with visible light against C. albicans and expression analysis of hyphal wall protein 1 (HWP1) and agglutinin-like sequence 1 (ALS1) genes in C. albicans. Antifungal susceptibility testing, photodynamic activity assay, reactive oxygen species (ROS) production assay and gene expression analysis were determined in C. albicans treated with MION alone and combined with visible light. MION at 1 × minimum inhibitory concentration (MIC) level (500 μg/mL) exhibited antifungal activity against C. albicans isolates. Further, 1 × MIC levels of MION alone and combined with visible light displayed remarkable fungicidal effects at 24 and 48 h after treatment. The MION combined with visible light caused the highest levels of ROS production by all C. albicans isolates. The relative RT-PCR data showed significant downregulation of HWP1 and ALS1 genes which are the key virulence genes in C. albicans. Differences in gene expression of HWP1 and ALS1 were more significant in MION combined with visible light treatments than MION alone. Our study sheds a novel light on facile development of effective treatment of C. albicans especially fluconazole-resistant C. albicans infections. The hyphae-specific genes HWP1 and ALS1 could be probable molecular targets for MION alone and combined with visible light in C. albicans.
Collapse
Affiliation(s)
| | - Asma Hosseini
- Department of Microbiology, Yasuj Branch, Islamic Azad University, Yasuj, Iran
| | - Alireza Khodavandi
- Department of Biology, Gachsaran Branch, Islamic Azad University, Gachsaran, Iran.
| | - Fahimeh Alizadeh
- Department of Biology, Gachsaran Branch, Islamic Azad University, Gachsaran, Iran.
| | - Arsalan Azizi
- Department of Pathology, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Majid Gerami
- Education Research Center, Yasuj University, Yasuj, Iran
| |
Collapse
|
23
|
Ganser C, Staples MI, Dowell M, Frazer C, Dainis J, Sircaik S, Bennett RJ. Filamentation and biofilm formation are regulated by the phase-separation capacity of network transcription factors in Candida albicans. PLoS Pathog 2023; 19:e1011833. [PMID: 38091321 PMCID: PMC10718430 DOI: 10.1371/journal.ppat.1011833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
The ability of the fungus Candida albicans to filament and form biofilms contributes to its burden as a leading cause of hospital-acquired infections. Biofilm development involves an interconnected transcriptional regulatory network (TRN) consisting of nine transcription factors (TFs) that bind both to their own regulatory regions and to those of the other network TFs. Here, we show that seven of the nine TFs in the C. albicans biofilm network contain prion-like domains (PrLDs) that have been linked to the ability to form phase-separated condensates. Construction of PrLD mutants in four biofilm TFs reveals that these domains are essential for filamentation and biofilm formation in C. albicans. Moreover, biofilm PrLDs promote the formation of phase-separated condensates in the nuclei of live cells, and PrLD mutations that abolish phase separation (such as the removal of aromatic residues) also prevent biofilm formation. Biofilm TF condensates can selectively recruit other TFs through PrLD-PrLD interactions and can co-recruit RNA polymerase II, implicating condensate formation in the assembly of active transcriptional complexes. Finally, we show that PrLD mutations that block the phase separation of biofilm TFs also prevent filamentation in an in vivo model of gastrointestinal colonization. Together, these studies associate transcriptional condensates with the regulation of filamentation and biofilm formation in C. albicans, and highlight how targeting of PrLD-PrLD interactions could prevent pathogenesis by this species.
Collapse
Affiliation(s)
- Collin Ganser
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| | - Mae I. Staples
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| | - Maureen Dowell
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| | - Corey Frazer
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| | - Joseph Dainis
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| | - Shabnam Sircaik
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| | - Richard J. Bennett
- Molecular Microbiology and Immunology Department, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
24
|
Bonincontro G, Scuderi SA, Marino A, Simonetti G. Synergistic Effect of Plant Compounds in Combination with Conventional Antimicrobials against Biofilm of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida spp. Pharmaceuticals (Basel) 2023; 16:1531. [PMID: 38004397 PMCID: PMC10675371 DOI: 10.3390/ph16111531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Bacterial and fungal biofilm has increased antibiotic resistance and plays an essential role in many persistent diseases. Biofilm-associated chronic infections are difficult to treat and reduce the efficacy of medical devices. This global problem has prompted extensive research to find alternative strategies to fight microbial chronic infections. Plant bioactive metabolites with antibiofilm activity are known to be potential resources to alleviate this problem. The phytochemical screening of some medicinal plants showed different active groups, such as stilbenes, tannins, alkaloids, terpenes, polyphenolics, flavonoids, lignans, quinones, and coumarins. Synergistic effects can be observed in the interaction between plant compounds and conventional drugs. This review analyses and summarises the current knowledge on the synergistic effects of plant metabolites in combination with conventional antimicrobials against biofilms of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans. The synergism of conventional antimicrobials with plant compounds can modify and inhibit the mechanisms of acquired resistance, reduce undesirable effects, and obtain an appropriate therapeutic effect at lower doses. A deeper knowledge of these combinations and of their possible antibiofilm targets is needed to develop next-generation novel antimicrobials and/or improve current antimicrobials to fight drug-resistant infections attributed to biofilm.
Collapse
Affiliation(s)
- Graziana Bonincontro
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Andreana Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Giovanna Simonetti
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| |
Collapse
|
25
|
Nayak A, Chavarria A, Sanders KN, Ghalei H, Khoshnevis S. Sinefungin, a natural nucleoside analog of S-adenosyl methionine, impairs the pathogenicity of Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.562127. [PMID: 37873365 PMCID: PMC10592816 DOI: 10.1101/2023.10.12.562127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Candida albicans, an opportunistic fungal human pathogen, is a major threat to the healthcare system due to both infections in immunocompromised individuals and the emergence of antifungal resistance. Fungal infection caused by C. albicans, candidiasis, is a life-threatening condition in immunocompromised patients and the current treatments are mostly restricted to polyenes, azoles, and echinocandins. Use of these antifungals is limited by toxicity, drug-drug interactions, and the emergence of resistance, underscoring the importance of identifying novel therapeutic targets and the need for new treatment approaches. C. albicans can undergo a morphological transition from yeast to hyphae and this transition is central to C. albicans virulence. Here, we determine the impact of sinefungin, a natural nucleoside analog of S-adenosyl methionine, on the virulence of C. albicans strain SC5314 by evaluating treatment effects on the morphological transition, human epithelial cell adhesion, and biofilm formation. Our data indicate that sinefungin impairs pathogenic traits of C. albicans including hyphal lengthening, biofilm formation and the adhesion to the human epithelial cell lines, without adversely affecting human cells, therefore highlighting sinefungin as a potential avenue for therapeutic intervention. We determine that the formation of N6-methyladenosine (m6A) is particularly disturbed by sinefungin. More broadly, this study underscores the importance of considering the post-transcriptional control mechanisms of pathogenicity when designing therapeutic solutions to fungal infection.
Collapse
Affiliation(s)
- Anushka Nayak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Alejandro Chavarria
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Kyla N. Sanders
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Sohail Khoshnevis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
26
|
Domi OKB, Darmani H. Syzygium aromaticum extracts debilitate Candida albicans by radically inhibiting its morphological plasticity and biofilm formation. JOURNAL OF HERBS, SPICES & MEDICINAL PLANTS 2023; 29:392-404. [DOI: 10.1080/10496475.2023.2196463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 01/03/2025]
Affiliation(s)
- Omar K.H. Bani Domi
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Homa Darmani
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
27
|
Rapala-Kozik M, Surowiec M, Juszczak M, Wronowska E, Kulig K, Bednarek A, Gonzalez-Gonzalez M, Karkowska-Kuleta J, Zawrotniak M, Satała D, Kozik A. Living together: The role of Candida albicans in the formation of polymicrobial biofilms in the oral cavity. Yeast 2023; 40:303-317. [PMID: 37190878 DOI: 10.1002/yea.3855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
The oral cavity of humans is colonized by diversity of microbial community, although dominated by bacteria, it is also constituted by a low number of fungi, often represented by Candida albicans. Although in the vast minority, this usually commensal fungus under certain conditions of the host (e.g., immunosuppression or antibiotic therapy), can transform into an invasive pathogen that adheres to mucous membranes and also to medical or dental devices, causing mucosal infections. This transformation is correlated with changes in cell morphology from yeast-like cells to hyphae and is supported by numerous virulence factors exposed by C. albicans cells at the site of infection, such as multifunctional adhesins, degradative enzymes, or toxin. All of them affect the surrounding host cells or proteins, leading to their destruction. However, at the site of infection, C. albicans can interact with different bacterial species and in its filamentous form may produce biofilms-the elaborated consortia of microorganisms, that present increased ability to host colonization and resistance to antimicrobial agents. In this review, we highlight the modification of the infectious potential of C. albicans in contact with different bacterial species, and also consider the mutual bacterial-fungal relationships, involving cooperation, competition, or antagonism, that lead to an increase in the propagation of oral infection. The mycofilm of C. albicans is an excellent hiding place for bacteria, especially those that prefer low oxygen availability, where microbial cells during mutual co-existence can avoid host recognition or elimination by antimicrobial action. However, these microbial relationships, identified mainly in in vitro studies, are modified depending on the complexity of host conditions and microbial dominance in vivo.
Collapse
Affiliation(s)
- Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Magdalena Surowiec
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Aneta Bednarek
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Miriam Gonzalez-Gonzalez
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satała
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
28
|
Watchaputi K, Jayasekara LACB, Ratanakhanokchai K, Soontorngun N. Inhibition of cell cycle-dependent hyphal and biofilm formation by a novel cytochalasin 19,20‑epoxycytochalasin Q in Candida albicans. Sci Rep 2023; 13:9724. [PMID: 37322086 PMCID: PMC10272203 DOI: 10.1038/s41598-023-36191-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023] Open
Abstract
Biofilm-mediated drug resistance is a key virulence factor of pathogenic microbes that cause a serious global health threat especially in immunocompromised individuals. Here, we investigated the antihyphal and antibiofilm activity of 19,20‑epoxycytochalasin Q (ECQ), a cytochalasin actin inhibitor isolated from medicinal mushroom Xylaria sp. BCC1067 against Candida albicans. Remarkably, 256 µg/ml of ECQ inhibited over 95% of C. albicans hyphal formation after 24 h-treatment. Combined ECQ and lipid-based biosurfactant effectively enhanced the antihyphal activity, lowering required ECQ concentrations. Hyphal fragmentation and reduction of biofilm biomass, shown by SEM and AFM visualization of ECQ-treated biofilms, were well corelated to the reduced metabolic activities of young and 24 h-preformed C. albicans biofilms. Induced intracellular accumulation of reactive oxygen species (ROS) also occurred in accompany with the leakage of shrunken cell membrane and defective cell wall at increasing ECQ concentrations. Transcriptomic analyses via RNA-sequencing revealed a massive change (> 1300 genes) in various biological pathways, following ECQ-treatment. Coordinated expression of genes, associated with cellular response to drugs, filamentous growth, cell adhesion, biofilm formation, cytoskeleton organization, cell division cycle, lipid and cell wall metabolisms was confirmed via qRT-PCR. Protein-protein association tool identified coupled expression between key regulators of cell division cyclin-dependent kinases (Cdc19/28) and a gamma-tubulin (Tub4). They coordinated ECQ-dependent hyphal specific gene targets of Ume6 and Tec1 during different phases of cell division. Thus, we first highlight the antihyphal and antibiofilm property of the novel antifungal agent ECQ against one of the most important life-threatening fungal pathogens by providing its key mechanistic detail in biofilm-related fungal infection.
Collapse
Affiliation(s)
- Kwanrutai Watchaputi
- Excellent Research Laboratory for Yeast Innovation, Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi (KMUTT), Bangkok, 10150, Thailand
| | - L A Channa Bhathiya Jayasekara
- Excellent Research Laboratory for Yeast Innovation, Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi (KMUTT), Bangkok, 10150, Thailand
| | - Khanok Ratanakhanokchai
- Excellent Center of Enzyme Technology and Microbial Utilization, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (KMUTT), Bangkok, 10150, Thailand
| | - Nitnipa Soontorngun
- Excellent Research Laboratory for Yeast Innovation, Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi (KMUTT), Bangkok, 10150, Thailand.
| |
Collapse
|
29
|
Wijaya M, Halleyantoro R, Kalumpiu JF. Biofilm: The invisible culprit in catheter-induced candidemia. AIMS Microbiol 2023; 9:467-485. [PMID: 37649801 PMCID: PMC10462453 DOI: 10.3934/microbiol.2023025] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 09/01/2023] Open
Abstract
Candidemia is the most common form of invasive fungal infection associated with several risk factors, and one of them is the use of medical devices, to which microbial biofilms can attach. Candidemia related to the use of peripheral intravascular and central venous catheters (CVC) is referred to as Candida catheter-related bloodstream infection, with more than 90% being related to CVC usage. The infection is associated with a higher morbidity and mortality rate than nosocomial bacterial infections. Candida spp. can protect themselves from the host immune system and antifungal drugs because of the biofilm structure, which is potentiated by the extracellular matrix (ECM). Candida albicans and Candida parapsilosis are the most pathogenic species often found to form biofilms associated with catheter usage. Biofilm formation of C. albicans includes four mechanisms: attachment, morphogenesis, maturation and dispersion. The biofilms formed between C. albicans and non-albicans spp. differ in ECM structure and composition and are associated with the persistence of colonization to infection for various catheter materials and antifungal resistance. Efforts to combat Candida spp. biofilm formation on catheters are still challenging because not all patients, especially those who are critically ill, can be recommended for catheter removal; also to be considered are the characteristics of the biofilm itself, which readily colonizes the permanent medical devices used. The limited choice and increasing systemic antifungal resistance also make treating it more difficult. Hence, alternative strategies have been developed to manage Candida biofilm. Current options for prevention or therapy in combination with systemic antifungal medications include lock therapy, catheter coating, natural peptide products and photodynamic inactivation.
Collapse
Affiliation(s)
- Meiliyana Wijaya
- Department of Parasitology, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Ryan Halleyantoro
- Department of Parasitology, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Jane Florida Kalumpiu
- Department of Parasitology, Faculty of Medicine, Pelita Harapan University, Banten, Indonesia
| |
Collapse
|
30
|
Kulshrestha A, Gupta P. Secreted aspartyl proteases family: a perspective review on the regulation of fungal pathogenesis. Future Microbiol 2023; 18:295-309. [PMID: 37097060 DOI: 10.2217/fmb-2022-0143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Secreted aspartyl proteases (SAPs) are important enzymes for fungal pathogenicity, playing a significant role in infection and survival. This article provides insight into how SAPs facilitate the transformation of yeast cells into hyphae and engage in biofilm formation, invasion and degradation of host cells and proteins. SAPs and their isoenzymes are prevalent during fungal infections, making them a potential target for antifungal and antibiofilm therapies. By targeting SAPs, critical stages of fungal pathogenesis such as adhesion, hyphal development, biofilm formation, host invasion and immune evasion can potentially be disrupted. Developing therapies that target SAPs could provide an effective treatment option for a wide range of fungal infections.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, India
| |
Collapse
|
31
|
Verma M, Singh V, Mishra V. Moving towards the enhancement of extracellular electron transfer in electrogens. World J Microbiol Biotechnol 2023; 39:130. [PMID: 36959310 DOI: 10.1007/s11274-023-03582-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Electrogens are very common in nature and becoming a contemporary theme for research as they can be exploited for extracellular electron transfer. Extracellular electron transfer is the key mechanism behind bioelectricity generation and bioremediation of pollutants via microbes. Extracellular electron transfer mechanisms for electrogens other than Shewanella and Geobacter are less explored. An efficient extracellular electron transfer system is crucial for the sustainable future of bioelectrochemical systems. At present, the poor extracellular electron transfer efficiency remains a decisive factor in limiting the development of efficient bioelectrochemical systems. In this review article, the EET mechanisms in different electrogens (bacteria and yeast) have been focused. Apart from the well-known electron transfer mechanisms of Shewanella oneidensis and Geobacter metallireducens, a brief introduction of the EET pathway in Rhodopseudomonas palustris TIE-1, Sideroxydans lithotrophicus ES-1, Thermincola potens JR, Lysinibacillus varians GY32, Carboxydothermus ferrireducens, Enterococcus faecalis and Saccharomyces cerevisiae have been included. In addition to this, the article discusses the several approaches to anode modification and genetic engineering that may be used in order to increase the rate of extracellular electron transfer. In the side lines, this review includes the engagement of the electrogens for different applications followed by the future perspective of efficient extracellular electron transfer.
Collapse
Affiliation(s)
- Manisha Verma
- School of Biochemical Engineering, IIT (BHU), 221005, Varanasi, India
| | - Vishal Singh
- School of Biochemical Engineering, IIT (BHU), 221005, Varanasi, India
| | - Vishal Mishra
- School of Biochemical Engineering, IIT (BHU), 221005, Varanasi, India.
| |
Collapse
|
32
|
do Rosário Esteves Guimarães C, de Freitas HF, Barros TF. Candida albicans antibiofilm molecules: analysis based on inhibition and eradication studies. Braz J Microbiol 2023; 54:37-52. [PMID: 36576671 PMCID: PMC9944165 DOI: 10.1007/s42770-022-00876-1] [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: 09/22/2022] [Accepted: 11/14/2022] [Indexed: 12/29/2022] Open
Abstract
Biofilms are communities of microbial cells surrounded by an extracellular polysaccharide matrix, recognized as a fungal source for local and systemic infections and less susceptible to antifungal drugs. Thus, treatment of biofilm-related Candida spp. infections with popular antifungals such as fluconazole is limited and species-dependent and alternatively demands the use of expensive and high toxic drugs. In this sense, molecules with antibiofilm activity have been studied but without care regarding the use of important criteria such as antibiofilm concentration lower than antifungal concentration when considering the process of inhibition of formation and concentrations equal to or lower than 300 µM. Therefore, this review tries to gather the most promising molecules regarding the activity against the C. albicans biofilm described in the last 10 years, considering the activity of inhibition and eradication. From January 2011 to July 2021, articles were searched on Scopus, PubMed, and Science Direct, combining the keywords "antibiofilm," "candida albicans," "compound," and "molecule" with AND and OR operators. After 3 phases of selection, 21 articles describing 42 molecules were discussed in the review. Most of them were more promising for the inhibition of biofilm formation, with SM21 (24) being an interesting molecule for presenting inhibitory and eradication activity in biofilms with 24 and 48 h, as well as alizarin (26) and chrysazine (27), with concentrations well below the antifungal concentration. Despite the detection of these molecules and the attempts to determine the mechanisms of action by microscopic analysis and gene expression, no specific target has been determined. Thus, a gap is signaled, requiring further studies such as proteomic analyses to clarify it.
Collapse
Affiliation(s)
- Carolina do Rosário Esteves Guimarães
- Post-Graduation Program in Pharmacy, Pharmacy College, Federal University of Bahia, Barão de Geremoabo Street, 147, Ondina, Salvador, Bahia CEP, 40170115, Brazil
| | - Humberto Fonseca de Freitas
- Post-Graduation Program in Pharmacy, Pharmacy College, Federal University of Bahia, Barão de Geremoabo Street, 147, Ondina, Salvador, Bahia CEP, 40170115, Brazil
| | - Tânia Fraga Barros
- Post-Graduation Program in Pharmacy, Pharmacy College, Federal University of Bahia, Barão de Geremoabo Street, 147, Ondina, Salvador, Bahia CEP, 40170115, Brazil.
| |
Collapse
|
33
|
Hassan N, Farooq U, Das AK, Sharma K, Mirza MA, Fatima S, Singh O, Ansari MJ, Ali A, Iqbal Z. In Silico Guided Nanoformulation Strategy for Circumvention of Candida albicans Biofilm for Effective Therapy of Candidal Vulvovaginitis. ACS OMEGA 2023; 8:6918-6930. [PMID: 36844532 PMCID: PMC9947946 DOI: 10.1021/acsomega.2c07718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Candidal vulvovaginitis involving multispecies of Candida and epithelium-bound biofilm poses a drug-resistant pharmacotherapeutic challenge. The present study aims for a disease-specific predominant causative organism resolution for the development of a tailored vaginal drug delivery system. The proposed work fabricates a luliconazole-loaded nanostructured lipid carrier-based transvaginal gel for combating Candida albicans biofilm and disease amelioration. The interaction and binding affinity of luliconazole against the proteins of C. albicans and biofilm were assessed using in silico tools. A systematic QbD analysis was followed to prepare the proposed nanogel using a modified melt emulsification-ultrasonication-gelling method. The DoE optimization was logically implemented to ascertain the effect of independent process variables (excipients concentration; sonication time) on dependent formulation responses (particle size; polydispersity index; entrapment efficiency). The optimized formulation was characterized for final product suitability. The surface morphology and dimensions were spherical and ≤300 nm, respectively. The flow behavior of an optimized nanogel (semisolid) was non-Newtonian similar to marketed preparation. The texture pattern of a nanogel was firm, consistent, and cohesive. The release kinetic model followed was Higuchi (nanogel) with a % cumulative drug release of 83.97 ± 0.69% in 48 h. The % cumulative drug permeated across a goat vaginal membrane was found to be 53.148 ± 0.62% in 8 h. The skin-safety profile was examined using a vaginal irritation model (in vivo) and histological assessments. The drug and proposed formulation(s) were checked against the pathogenic strains of C. albicans (vaginal clinical isolates) and in vitro established biofilms. The visualization of biofilms was done under a fluorescence microscope revealing mature, inhibited, and eradicated biofilm structures.
Collapse
Affiliation(s)
- Nazia Hassan
- Department
of Pharmaceutics, School of Pharmaceutical Education & Research
(SPER), Jamia Hamdard, New Delhi 110062, India
| | - Uzma Farooq
- Department
of Pharmaceutics, School of Pharmaceutical Education & Research
(SPER), Jamia Hamdard, New Delhi 110062, India
| | - Ayan Kumar Das
- Hamdard
Institute of Medical Sciences & Research, Jamia Hamdard, New Delhi 110062, India
| | - Kalicharan Sharma
- Department
of Pharmaceutical Chemistry, DPSRU, New Delhi 110017, India
| | - Mohd. Aamir Mirza
- Department
of Pharmaceutics, School of Pharmaceutical Education & Research
(SPER), Jamia Hamdard, New Delhi 110062, India
| | - Suhail Fatima
- Department
of Amraz-E-Niswan Wa Qabalat, School of Unani Medical Education &
Research (SUMER), Jamia Hamdard, New Delhi 110062, India
| | - Omana Singh
- Department
of Pharmaceutics, School of Pharmaceutical Education & Research
(SPER), Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Javed Ansari
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-Kharj 16278, Saudi Arabia
| | - Asgar Ali
- Department
of Pharmaceutics, School of Pharmaceutical Education & Research
(SPER), Jamia Hamdard, New Delhi 110062, India
| | - Zeenat Iqbal
- Department
of Pharmaceutics, School of Pharmaceutical Education & Research
(SPER), Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
34
|
Sprague JL, Kasper L, Hube B. From intestinal colonization to systemic infections: Candida albicans translocation and dissemination. Gut Microbes 2022; 14:2154548. [PMID: 36503341 PMCID: PMC9746630 DOI: 10.1080/19490976.2022.2154548] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Candida species are the most prevalent cause of invasive fungal infections, of which Candida albicans is the most common. Translocation across the epithelial barrier into the bloodstream by intestinal-colonizing C. albicans cells serves as the main source for systemic infections. Understanding the fungal mechanisms behind this process will give valuable insights on how to prevent such infections and keep C. albicans in the commensal state in patients with predisposing conditions. This review will focus on recent developments in characterizing fungal translocation mechanisms, compare what we know about enteric bacterial pathogens with C. albicans, and discuss the different proposed hypotheses for how C. albicans enters and disseminates through the bloodstream immediately following translocation.
Collapse
Affiliation(s)
- Jakob L. Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany,Contact: Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Beutenbergstrasse 11a, 07745Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany,Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
35
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 203] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
36
|
Cattò C, Corte L, Roscini L, Cardinali G, Villa F, Cappitelli F. Metabolomic and Proteomic Changes in Candida albicans Biofilm in Response to Zosteric Acid Treatment. Int J Mol Sci 2022; 23:ijms232214067. [PMID: 36430545 PMCID: PMC9697788 DOI: 10.3390/ijms232214067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Zosteric acid (ZA) is a secondary metabolite of the seagrass Zostera marina, with antibiofilm activity against fungi. Information concerning its mechanisms of action is lacking and this limits the development of more potent derivatives based on the same target and activity structure. The aim of this work was to investigate the ZA mode of action by analyzing the metabolic status of Candida albicans biofilm and its protein expression profile upon ZA treatment. Fourier-Transform Infrared Spectroscopy confirmed that ZA modified the metabolomic response of treated cells, showing changes in the spectral regions, mainly related to the protein compartment. Nano Liquid Chromatography-High-Resolution Mass Spectrometry highlighted that 10 proteins were differentially expressed in the C. albicans proteome upon ZA treatment. Proteins involved in the biogenesis, structure and integrity of cell walls as well as adhesion and stable attachment of hyphae were found downregulated, whereas some proteins involved in the stress response were found overexpressed. Additionally, ZA was involved in the modulation of non-DNA-based epigenetic regulatory mechanisms triggered by reactive oxygen species. These results partially clarified the ZA mechanism of action against fungi and provided insight into the major C. albicans pathways responsible for biofilm formation.
Collapse
Affiliation(s)
- Cristina Cattò
- Department of Food Environmental and Nutritional Sciences, Università degli Studi di Milano, 20133 Milano, Italy
- Correspondence: ; Tel.: +39-02-503-19121
| | - Laura Corte
- Department of Pharmaceutical Sciences-Microbiology, Università di Perugia, 06121 Perugia, Italy
| | - Luca Roscini
- Department of Pharmaceutical Sciences-Microbiology, Università di Perugia, 06121 Perugia, Italy
| | - Gianluigi Cardinali
- Department of Pharmaceutical Sciences-Microbiology, Università di Perugia, 06121 Perugia, Italy
| | - Federica Villa
- Department of Food Environmental and Nutritional Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Francesca Cappitelli
- Department of Food Environmental and Nutritional Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| |
Collapse
|
37
|
Light-driven single-cell rotational adhesion frequency assay. ELIGHT 2022; 2:13. [PMID: 35965781 PMCID: PMC9358104 DOI: 10.1186/s43593-022-00020-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/28/2022] [Accepted: 07/07/2022] [Indexed: 01/13/2023]
Abstract
The interaction between cell surface receptors and extracellular ligands is highly related to many physiological processes in living systems. Many techniques have been developed to measure the ligand-receptor binding kinetics at the single-cell level. However, few techniques can measure the physiologically relevant shear binding affinity over a single cell in the clinical environment. Here, we develop a new optical technique, termed single-cell rotational adhesion frequency assay (scRAFA), that mimics in vivo cell adhesion to achieve label-free determination of both homogeneous and heterogeneous binding kinetics of targeted cells at the subcellular level. Moreover, the scRAFA is also applicable to analyze the binding affinities on a single cell in native human biofluids. With its superior performance and general applicability, scRAFA is expected to find applications in study of the spatial organization of cell surface receptors and diagnosis of infectious diseases.
Collapse
|
38
|
Liu Y, Ding H, Li J, Lou X, Yang M, Zheng Y. Light-driven single-cell rotational adhesion frequency assay. ELIGHT 2022; 2:13. [PMID: 35965781 DOI: 10.1186/s43593-022-00013-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/28/2022] [Accepted: 07/07/2022] [Indexed: 05/23/2023]
Abstract
UNLABELLED The interaction between cell surface receptors and extracellular ligands is highly related to many physiological processes in living systems. Many techniques have been developed to measure the ligand-receptor binding kinetics at the single-cell level. However, few techniques can measure the physiologically relevant shear binding affinity over a single cell in the clinical environment. Here, we develop a new optical technique, termed single-cell rotational adhesion frequency assay (scRAFA), that mimics in vivo cell adhesion to achieve label-free determination of both homogeneous and heterogeneous binding kinetics of targeted cells at the subcellular level. Moreover, the scRAFA is also applicable to analyze the binding affinities on a single cell in native human biofluids. With its superior performance and general applicability, scRAFA is expected to find applications in study of the spatial organization of cell surface receptors and diagnosis of infectious diseases. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1186/s43593-022-00020-4.
Collapse
Affiliation(s)
- Yaoran Liu
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX 78712 USA
| | - Hongru Ding
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
| | - Jingang Li
- Materials Science & Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, TX 78712 USA
| | - Xin Lou
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Mingcheng Yang
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190 China
- Songshan Lake Materials Laboratory, Dongguan, 523808 Guangdong China
| | - Yuebing Zheng
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX 78712 USA
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
- Materials Science & Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, TX 78712 USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
| |
Collapse
|
39
|
Farmand M, Jahanpeyma F, Gholaminejad A, Azimzadeh M, Malaei F, Shoaie N. Carbon nanostructures: a comprehensive review of potential applications and toxic effects. 3 Biotech 2022; 12:159. [PMID: 35814038 PMCID: PMC9259781 DOI: 10.1007/s13205-022-03175-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/25/2022] [Indexed: 12/17/2022] Open
Abstract
There is no doubt that nanotechnology has revolutionized our life since the 1970s when it was first introduced. Nanomaterials have helped us to improve the current products and services we use. Among the different types of nanomaterials, the application of carbon-based nanomaterials in every aspect of our lives has rapidly grown over recent decades. This review discusses recent advances of those applications in distinct categories, including medical, industrial, and environmental applications. The first main section introduces nanomaterials, especially carbon-based nanomaterials. In the first section, we discussed medical applications, including medical biosensors, drug and gene delivery, cell and tissue labeling and imaging, tissue engineering, and the fight against bacterial and fungal infections. The next section discusses industrial applications, including agriculture, plastic, electronic, energy, and food industries. In addition, the environmental applications, including detection of air and water pollutions and removal of environmental pollutants, were vastly reviewed in the last section. In the conclusion section, we discussed challenges and future perspectives.
Collapse
Affiliation(s)
- Maryam Farmand
- Department of Biology, Tehran University, PO Box: 14155-6619, Tehran, Iran
| | - Fatemeh Jahanpeyma
- Department of Medical Biotechnology, Faculty of Medical Science, Tarbiat Modares University, P.O. Box: 14115-111, Tehran, Iran
| | - Alieh Gholaminejad
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, PO Box: 73461-81746, Isfahan, Iran
| | - Mostafa Azimzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, PO Box: 89195-999, Yazd, Iran
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, PO Box: 89195-999, Yazd, Iran
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, PO Box: 8916188635, Yazd, Iran
| | - Fatemeh Malaei
- Department of Medical Biotechnology, Faculty of Medical Science, Tarbiat Modares University, P.O. Box: 14115-111, Tehran, Iran
| | - Nahid Shoaie
- Department of Medical Biotechnology, Faculty of Medical Science, Tarbiat Modares University, P.O. Box: 14115-111, Tehran, Iran
| |
Collapse
|
40
|
Molecular Mapping of Antifungal Mechanisms Accessing Biomaterials and New Agents to Target Oral Candidiasis. Int J Mol Sci 2022; 23:ijms23147520. [PMID: 35886869 PMCID: PMC9320712 DOI: 10.3390/ijms23147520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Oral candidiasis has a high rate of development, especially in immunocompromised patients. Immunosuppressive and cytotoxic therapies in hospitalized HIV and cancer patients are known to induce the poor management of adverse reactions, where local and systemic candidiasis become highly resistant to conventional antifungal therapy. The development of oral candidiasis is triggered by several mechanisms that determine oral epithelium imbalances, resulting in poor local defense and a delayed immune system response. As a result, pathogenic fungi colonies disseminate and form resistant biofilms, promoting serious challenges in initiating a proper therapeutic protocol. Hence, this study of the literature aimed to discuss possibilities and new trends through antifungal therapy for buccal drug administration. A large number of studies explored the antifungal activity of new agents or synergic components that may enhance the effect of classic drugs. It was of significant interest to find connections between smart biomaterials and their activity, to find molecular responses and mechanisms that can conquer the multidrug resistance of fungi strains, and to transpose them into a molecular map. Overall, attention is focused on the nanocolloids domain, nanoparticles, nanocomposite synthesis, and the design of polymeric platforms to satisfy sustained antifungal activity and high biocompatibility with the oral mucosa.
Collapse
|
41
|
Khadke SK, Lee JH, Kim YG, Raj V, Lee J. Appraisal of Cinnamaldehyde Analogs as Dual-Acting Antibiofilm and Anthelmintic Agents. Front Microbiol 2022; 13:818165. [PMID: 35369516 PMCID: PMC8966877 DOI: 10.3389/fmicb.2022.818165] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
Cinnamaldehyde has a broad range of biological activities, which include antibiofilm and anthelmintic activities. The ever-growing problem of drug resistance and limited treatment options have created an urgent demand for natural molecules with antibiofilm and anthelmintic properties. Hence, we hypothesized that molecules with a scaffold structurally similar to that of cinnamaldehyde might act as dual inhibitors against fungal biofilms and helminths. In this regard, eleven cinnamaldehyde analogs were tested to determine their effects on fungal Candida albicans biofilm and nematode Caenorhabditis elegans. α-Methyl and trans-4-methyl cinnamaldehydes efficiently inhibited C. albicans biofilm formation (>90% inhibition at 50 μg/mL) with minimum inhibitory concentrations (MICs) of ≥ 200 μg/mL and 4-bromo and 4-chloro cinnamaldehydes exhibited anthelmintic property at 20 μg/mL against C. elegans. α-Methyl and trans-4-methyl cinnamaldehydes inhibited hyphal growth and cell aggregation. Scanning electron microscopy was employed to determine the surface architecture of C. albicans biofilm and cuticle of C. elegans, and confocal laser scanning microscopy was used to determine biofilm characteristics. The perturbation in gene expression of C. albicans was investigated using qRT-PCR analysis and α-methyl and trans-4-methyl cinnamaldehydes exhibited down-regulation of ECE1, IFD6, RBT5, UCF1, and UME6 and up-regulation of CHT4 and YWP1. Additionally, molecular interaction of these two molecules with UCF1 and YWP1 were revealed by molecular docking simulation. Our observations collectively suggest α-methyl and trans-4-methyl cinnamaldehydes are potent biofilm inhibitors and that 4-bromo and 4-chloro cinnamaldehydes are anthelmintic agents. Efforts are required to determine the range of potential therapeutic applications of cinnamaldehyde analogs.
Collapse
Affiliation(s)
- Sagar Kiran Khadke
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Vinit Raj
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
42
|
Hassan MN, Nabi F, Khan AN, Hussain M, Siddiqui WA, Uversky VN, Khan RH. The amyloid state of proteins: A boon or bane? Int J Biol Macromol 2022; 200:593-617. [PMID: 35074333 DOI: 10.1016/j.ijbiomac.2022.01.115] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/05/2022]
Abstract
Proteins and their aggregation is significant field of research due to their association with various conformational maladies including well-known neurodegenerative diseases like Alzheimer's (AD), Parkinson's (PD), and Huntington's (HD) diseases. Amyloids despite being given negative role for decades are also believed to play a functional role in bacteria to humans. In this review, we discuss both facets of amyloid. We have shed light on AD, which is one of the most common age-related neurodegenerative disease caused by accumulation of Aβ fibrils as extracellular senile plagues. We also discuss PD caused by the aggregation and deposition of α-synuclein in form of Lewy bodies and neurites. Other amyloid-associated diseases such as HD and amyotrophic lateral sclerosis (ALS) are also discussed. We have also reviewed functional amyloids that have various biological roles in both prokaryotes and eukaryotes that includes formation of biofilm and cell attachment in bacteria to hormone storage in humans, We discuss in detail the role of Curli fibrils' in biofilm formation, chaplins in cell attachment to peptide hormones, and Pre-Melansomal Protein (PMEL) roles. The disease-related and functional amyloids are compared with regard to their structural integrity, variation in regulation, and speed of forming aggregates and elucidate how amyloids have turned from foe to friend.
Collapse
Affiliation(s)
- Md Nadir Hassan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Faisal Nabi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Asra Nasir Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Murtaza Hussain
- Department of Biochemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Waseem A Siddiqui
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Vladimir N Uversky
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, 10 Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy 11 of Sciences", Pushchino, Moscow Region 142290, Russia; Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College 13 of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
43
|
Cid-Chevecich C, Müller-Sepúlveda A, Jara JA, López-Muñoz R, Santander R, Budini M, Escobar A, Quijada R, Criollo A, Díaz-Dosque M, Molina-Berríos A. Origanum vulgare L. essential oil inhibits virulence patterns of Candida spp. and potentiates the effects of fluconazole and nystatin in vitro. BMC Complement Med Ther 2022; 22:39. [PMID: 35139827 PMCID: PMC8827202 DOI: 10.1186/s12906-022-03518-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Background Recurrence and resistance of Candida spp. infections is associated with the ability of these microorganisms to present several virulence patterns such as morphogenesis, adhesion, and biofilm formation. In the search for agents with antivirulence activity, essential oils could represent a strategy to act against biofilms and to potentiate antifungal drugs. Objective To evaluate the antivirulence effect of Origanum vulgare L. essential oil (O-EO) against Candida spp. and to potentiate the effect of fluconazole and nystatin. Methods The effect of O-EO was evaluated on ATCC reference strains of C. albicans and non-albicans Candida species. Minimum inhibitory concentration (MIC) was determined through broth microdilution assay. Adhesion to microplates was determined by crystal violet (CV) assay. An adapted scratch assay in 24-well was used to determine the effect of essential oil on biofilms proliferation. Viability of biofilms was evaluated by MTT reduction assay and through a checkerboard assay we determined if O-EO could act synergistically with fluconazole and nystatin. Results MIC for C. albicans ATCC-90029 and ATCC-10231 was 0.01 mg/L and 0.97 mg/L, respectively. For non-albicans Candida strains MIC values were 2.6 mg/L for C. dubliniensis ATCC-CD36 and 5.3 mg/L for C. krusei ATCC-6258. By using these concentrations, O-EO inhibited morphogenesis, adhesion, and proliferation at least by 50% for the strains assayed. In formed biofilms O-EO decreased viability in ATCC 90029 and ATCC 10231 strains (IC50 7.4 and 2.8 mg/L respectively). Finally, we show that O-EO interacted synergistically with fluconazole and nystatin. Conclusions This study demonstrate that O-EO could be considered to improve the antifungal treatment against Candida spp. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03518-z.
Collapse
Affiliation(s)
- Camila Cid-Chevecich
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Andrea Müller-Sepúlveda
- Institute of Agrifood, Animals and Environmental Sciences, Universidad de O'Higgins, San Fernando, Chile
| | - José Antonio Jara
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Rodrigo López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Rocío Santander
- Department of Environmental Sciences, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Mauricio Budini
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Alejandro Escobar
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Raúl Quijada
- Faculty of Physical and Mathematical Sciences, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Mario Díaz-Dosque
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile.
| | - Alfredo Molina-Berríos
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile.
| |
Collapse
|
44
|
Abreu-Pereira CA, Klein MI, Vitorino Lobo CI, Gorayb Pereira AL, Jordão CC, Pavarina AC. DNase enhances photodynamic therapy against fluconazole-resistant Candida albicans biofilms. Oral Dis 2022; 29:1855-1867. [PMID: 35133698 DOI: 10.1111/odi.14149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/19/2022] [Accepted: 02/03/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE This study evaluated the effectiveness of DNase I combined with antimicrobial photodynamic therapy, mediated by Photodithazine® and light-emitting diode light, against biofilms formed by a fluconazole-resistant Candida albicans strain (ATCC 96901) and two clinical isolates (R14 and R70). MATERIALS AND METHODS Biofilms were grown for 48 h and exposed to DNase for 5 min, followed by application of a photosensitizer (P) and light (L), either singly or combined (P+L+, P-L+, P+L-, P-L-, P-L-DNase, P+L+DNase, P+L-DNase, and P-L+DNase; n = 12). Biofilm analysis included quantification of extracellular matrix components (water-soluble and insoluble proteins and polysaccharides, and extracellular DNA), and biomass (total and insoluble), as well as enumeration of colony-forming units. The data were analyzed using three-way analysis of variance with Bonferroni's post-hoc test. RESULTS The DNase treatment combined with aPDT showed a reduction of 1.92, 1.65, and 1.29 log10 of cell viability compared with untreated controls for ATCC 96901, R14, and R70 strains, respectively. It also reduced extracellular matrix contents of water-soluble polysaccharides (36.3%) and extracellular DNA (72.3%), as well as insoluble biomass content (43.3%). CONCLUSION The three strains showed similar behavior when treated with DNase, and the extracellular matrix components were affected, improving the effectiveness of antimicrobial photodynamic therapy.
Collapse
Affiliation(s)
- César Augusto Abreu-Pereira
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, Univ Estadual Paulista - UNESP, Rua Humaitá, 1680, 14801-903, Araraquara, SP, Brazil
| | - Marlise Inêz Klein
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, Univ Estadual Paulista - UNESP, Rua Humaitá, 1680, 14801-903, Araraquara, SP, Brazil
| | - Carmélia Isabel Vitorino Lobo
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, Univ Estadual Paulista - UNESP, Rua Humaitá, 1680, 14801-903, Araraquara, SP, Brazil
| | - Ana Luiza Gorayb Pereira
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, Univ Estadual Paulista - UNESP, Rua Humaitá, 1680, 14801-903, Araraquara, SP, Brazil
| | - Cláudia Carolina Jordão
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, Univ Estadual Paulista - UNESP, Rua Humaitá, 1680, 14801-903, Araraquara, SP, Brazil
| | - Ana Claudia Pavarina
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, Univ Estadual Paulista - UNESP, Rua Humaitá, 1680, 14801-903, Araraquara, SP, Brazil
| |
Collapse
|
45
|
Inhibitory effect of 405-nm blue LED light on the growth of Candida albicans and Streptococcus mutans dual-species biofilms on denture base resin. Lasers Med Sci 2022; 37:2311-2319. [PMID: 35034224 DOI: 10.1007/s10103-022-03507-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
Abstract
We investigated whether irradiation with 405-nm blue LED light could inhibit the growth of not only single- but dual-species biofilms formed by Candida albicans and Streptococcus mutans on denture base resin and cause the alteration in gene expression related to adhesion and biofilm formation. C. albicans and S. mutans single-/dual-species biofilms were formed on the denture base specimens. The biofilms were irradiated with 405-nm blue LED light (power density output: 280 mW/cm2) for 0 (control) and 40 min. Dual-species biofilms were analyzed using CFU assay and fluorescence microscopy, and single-/dual-species biofilms were analyzed using alamarBlue assays and gene expression analysis. To assess the inhibitory effect of irradiation on dual-species biofilms, specimens after irradiation were aerobically incubated for 12 h. After incubation, the inhibition of growth was assessed using CFU assays and fluorescence microscopy. Data were analyzed using the Mann-Whitney U or Student's t test (p < 0.05). Irradiation produced a significant inhibitory effect on biofilms. Fluorescence microscopy revealed that almost all C. albicans and S. mutans cells were killed by irradiation, and there was no notable difference in biofilm thickness immediately after irradiation and after irradiation and incubation for 12 h. alamarBlue assays indicated the growth of the biofilms was inhibited for 12-13 h. The expression of genes associated with adhesion and biofilm formation-als1 in C. albicans and ftf, gtfC, and gtfB in S. mutans-significantly reduced by irradiation. Irradiation with 405-nm blue LED light effectively inhibited the growth of C. albicans and S. mutans dual-species biofilms for 12 h.
Collapse
|
46
|
Subroto E, van Neer J, Valdes I, de Cock H. Growth of Aspergillus fumigatus in Biofilms in Comparison to Candida albicans. J Fungi (Basel) 2022; 8:48. [PMID: 35049988 PMCID: PMC8779434 DOI: 10.3390/jof8010048] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 12/18/2022] Open
Abstract
Biofilm formation during infections with the opportunistic pathogen Aspergillus fumigatus can be very problematic in clinical settings, since it provides the fungal cells with a protective environment. Resistance against drug treatments, immune recognition as well as adaptation to the host environment allows fungal survival in the host. The exact molecular mechanisms behind most processes in the formation of biofilms are unclear. In general, the formation of biofilms can be categorized roughly in a few stages; adhesion, conidial germination and development of hyphae, biofilm maturation and cell dispersion. Fungi in biofilms can adapt to the in-host environment. These adaptations can occur on a level of phenotypic plasticity via gene regulation. However, also more substantial genetic changes of the genome can result in increased resistance and adaptation in the host, enhancing the survival chances of fungi in biofilms. Most research has focused on the development of biofilms. However, to tackle developing microbial resistance and adaptation in biofilms, more insight in mechanisms behind genetic adaptations is required to predict which defense mechanisms can be expected. This can be helpful in the development of novel and more targeted antifungal treatments to combat fungal infections.
Collapse
Affiliation(s)
| | | | | | - Hans de Cock
- Molecular Microbiology Laboratory, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; (E.S.); (J.v.N.); (I.V.)
| |
Collapse
|
47
|
Esposito A, Migliaccio A, Iula VD, Zarrilli R, Guaragna A, De Gregorio E. The Glucocorticoid PYED-1 Disrupts Mature Biofilms of Candida spp. and Inhibits Hyphal Development in Candida albicans. Antibiotics (Basel) 2021; 10:1396. [PMID: 34827334 PMCID: PMC8614962 DOI: 10.3390/antibiotics10111396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Invasive Candida infections have become a global public health problem due to the increase of Candida species resistant against antifungal therapeutics. The glucocorticoid PYED-1 (pregnadiene-11-hydroxy-16α,17α-epoxy-3,20-dione-1) has antimicrobial activity against various bacterial taxa. Consequently, it might be considered for the treatment of Candida infections. The antifungal activity of PYED-1 was evaluated against several fungal strains that were representative of the five species that causes the majority of Candida infections-namely, Candida albicans, Candida glabrata, Candida tropicalis, Candida parapsilosis and Candida krusei. PYED-1 exhibited a weak antifungal activity and a fungistatic effect on all five Candida species. On the other hand, PYED-1 exhibited a good anti-biofilm activity, and was able to eradicate the preformed biofilms of all Candida species analyzed. Moreover, PYED-1 inhibited germ tube and hyphae formation of C. albicans and reduced adhesion of C. albicans to abiotic surfaces by up to 30%.
Collapse
Affiliation(s)
- Anna Esposito
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80126 Naples, Italy; (A.E.); (A.G.)
| | - Antonella Migliaccio
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (A.M.); (R.Z.)
| | - Vita Dora Iula
- Complex Operative Unit of Clinical Pathology, Ospedale del Mare-ASL NA1 Centro, 80145 Naples, Italy;
| | - Raffaele Zarrilli
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (A.M.); (R.Z.)
| | - Annalisa Guaragna
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80126 Naples, Italy; (A.E.); (A.G.)
| | - Eliana De Gregorio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
48
|
Tonprasong W, Inokoshi M, Tamura M, Uo M, Wada T, Takahashi R, Hatano K, Shimizubata M, Minakuchi S. Tissue Conditioner Incorporating a Nano-Sized Surface Pre-Reacted Glass-Ionomer (S-PRG) Filler. MATERIALS 2021; 14:ma14216648. [PMID: 34772173 PMCID: PMC8588282 DOI: 10.3390/ma14216648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022]
Abstract
We aimed to evaluate the properties of a novel tissue conditioner containing a surface pre-reacted glass-ionomer (S-PRG) nanofiller. Tissue conditioners containing 0 (control), 2.5, 5, 10, 20, or 30 wt% S-PRG nanofiller or 10 or 20 wt% S-PRG microfiller were prepared. The S-PRG nanofillers and microfillers were observed using scanning electron microscopy. The ion release, acid buffering capacity, detail reproduction, consistency, Shore A0 hardness, surface roughness, and Candida albicans adhesion of the tissue conditioners were examined. The results indicated that the nanofiller particles were smaller and more homogeneous in size than the microfiller particles. In addition, Al, B, F, and Sr ions eluted from S-PRG were generally found to decrease after 1 day. Acid neutralization was confirmed in a concentration-dependent manner. The mechanical properties of tissue conditioners containing S-PRG nanofiller were clinically acceptable according to ISO standard 10139-1:2018, although the surface roughness increased with increasing filler content. Conditioners with 5-30 wt% nanofiller had a sublethal effect on C. albicans and reduced fungal adhesion in vitro. In summary, tissue conditioner containing at least 5 wt% S-PRG nanofiller can reduce C. albicans adhesion and has potential as an alternative soft lining material.
Collapse
Affiliation(s)
- Watcharapong Tonprasong
- Department of Gerodontology and Oral Rehabilitation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (W.T.); (K.H.); (M.S.); (S.M.)
| | - Masanao Inokoshi
- Department of Gerodontology and Oral Rehabilitation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (W.T.); (K.H.); (M.S.); (S.M.)
- Correspondence:
| | - Muneaki Tamura
- Department of Microbiology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda, Tokyo 101-8310, Japan;
| | - Motohiro Uo
- Department of Advanced Biomaterials, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.U.); (T.W.)
| | - Takahiro Wada
- Department of Advanced Biomaterials, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.U.); (T.W.)
| | - Rena Takahashi
- Department of Cariology and Operative Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan;
| | - Keita Hatano
- Department of Gerodontology and Oral Rehabilitation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (W.T.); (K.H.); (M.S.); (S.M.)
| | - Makoto Shimizubata
- Department of Gerodontology and Oral Rehabilitation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (W.T.); (K.H.); (M.S.); (S.M.)
| | - Shunsuke Minakuchi
- Department of Gerodontology and Oral Rehabilitation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (W.T.); (K.H.); (M.S.); (S.M.)
| |
Collapse
|
49
|
Antifungal activity of menthol alone and in combination on growth inhibition and biofilm formation of Candida albicans. J Herb Med 2021. [DOI: 10.1016/j.hermed.2021.100495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Candida albicans Sfp1 Is Involved in the Cell Wall and Endoplasmic Reticulum Stress Responses Induced by Human Antimicrobial Peptide LL-37. Int J Mol Sci 2021; 22:ijms221910633. [PMID: 34638975 PMCID: PMC8508991 DOI: 10.3390/ijms221910633] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Candida albicans is a commensal fungus of humans but can cause infections, particularly in immunocompromised individuals, ranging from superficial to life-threatening systemic infections. The cell wall is the outermost layer of C. albicans that interacts with the host environment. Moreover, antimicrobial peptides (AMPs) are important components in innate immunity and play crucial roles in host defense. Our previous studies showed that the human AMP LL-37 binds to the cell wall of C. albicans, alters the cell wall integrity (CWI) and affects cell adhesion of this pathogen. In this study, we aimed to further investigate the molecular mechanisms underlying the C. albicans response to LL-37. We found that LL-37 causes cell wall stress, activates unfolded protein response (UPR) signaling related to the endoplasmic reticulum (ER), induces ER-derived reactive oxygen species and affects protein secretion. Interestingly, the deletion of the SFP1 gene encoding a transcription factor reduced C. albicans susceptibility to LL-37, which is cell wall-associated. Moreover, in the presence of LL-37, deletion of SFP1 attenuated the UPR pathway, upregulated oxidative stress responsive (OSR) genes and affected bovine serum albumin (BSA) degradation by secreted proteases. Therefore, these findings suggested that Sfp1 positively regulates cell wall integrity and ER homeostasis upon treatment with LL-37 and shed light on pathogen-host interactions.
Collapse
|