1
|
Roa-Cordero MV, Arenas-Sepúlveda CA, Herrera-Plata MC, Leal-Pinto SM, Villota-Salazar NA, González-Prieto JM. Switching off the yeast-to-hyphae transition in Yarrowia lipolytica through histone deacetylase inhibitors. Res Microbiol 2025:104299. [PMID: 40306381 DOI: 10.1016/j.resmic.2025.104299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025]
Abstract
Fungi can develop a variety of morphotypes to survive, colonize, adapt and prevail under different environmental conditions. In general, two morphological shapes encompass the others: yeast (unicellular) and hyphae (multicellular). Under specific conditions, some fungi can adopt these two cellular morphologies, and for this reason, they are called dimorphic. Histone acetylation and deacetylation are well-known important mechanisms of chromatin remodelling that control cell differentiation processes as dimorphism. The reactions involved are catalysed by histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. In the present work, we used Yarrowia lipolytica as a dimorphic fungal model to investigate the effect of HDAC chemical inhibition on the growth and yeast-to-hyphae switch of fungi. For this purpose, we tested the compounds sodium butyrate (SB) and valproic acid (VPA) as epigenetic modulators. Our results indicated that Y. lipolytica tolerates high doses of these inhibitors due to its lipolytic nature. However, once the metabolic capability of the fungus is overcome, SB and VPA strongly suppress hyphal growth, suggesting that histone acetylation plays a pivotal role in the regulation of this process.
Collapse
Affiliation(s)
- Martha Viviana Roa-Cordero
- Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación Masira, 680006, Bucaramanga, Colombia.
| | | | - María Cristina Herrera-Plata
- Universidad de Santander, Facultad de Ciencias Exactas, Naturales y Agropecuarias, 680006, Bucaramanga, Colombia.
| | - Sandra Milena Leal-Pinto
- Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación Masira, 680006, Bucaramanga, Colombia.
| | - Nubia Andrea Villota-Salazar
- Biotecnología Vegetal, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Blvd. del Maestro. S/N esq. Elías Piña. Col. Narciso Mendoza, 88710, Reynosa, Tamaulipas, México; Vidarium-Nutrition, Health and Wellness Research Center, Grupo Empresarial Nutresa, Calle 8 sur # 50-67, 050023 Medellín, Colombia.
| | - Juan Manuel González-Prieto
- Biotecnología Vegetal, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Blvd. del Maestro. S/N esq. Elías Piña. Col. Narciso Mendoza, 88710, Reynosa, Tamaulipas, México.
| |
Collapse
|
2
|
Lortal L, Lyon CM, Sprague JL, Sonnberger J, Paulin OKA, Wickramasinghe DN, Richardson JP, Hube B, Naglik JR. Candidalysin biology and activation of host cells. mBio 2025:e0060324. [PMID: 40293285 DOI: 10.1128/mbio.00603-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can cause life-threatening systemic infections and distressing mucosal infections. A major breakthrough in understanding C. albicans pathogenicity was the discovery of candidalysin, the first cytolytic peptide toxin identified in a human pathogenic fungus. Secreted by C. albicans hyphae and encoded by the ECE1 gene, this 31-amino acid peptide integrates into and permeabilizes host cell membranes, causing damage across diverse cell types. Beyond its cytolytic activity, candidalysin can trigger potent innate immune responses in epithelial cells, macrophages, and neutrophils. Additionally, candidalysin plays a key role in nutrient acquisition during infection. This review explores the biology of candidalysin, its role in host cell activation, and extends the discussion to non-candidalysin Ece1p peptides, shedding light on their emerging significance.
Collapse
Affiliation(s)
- Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Claire M Lyon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Johannes Sonnberger
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Olivia K A Paulin
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Don N Wickramasinghe
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Vetsch G, Manoil D, Wulfman C, Parga A, Durual S, Srinivasan M. Candida albicans colonization on CAD-CAM denture resin surface. J Dent 2025; 157:105756. [PMID: 40222702 DOI: 10.1016/j.jdent.2025.105756] [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: 12/31/2024] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025] Open
Abstract
OBJECTIVE This study assessed the biofilm formation of C. albicans on milled and 3D-printed denture resin surfaces and compared it to a control group of conventional heat-polymerized polymethylmethacrylate (PMMA) resin group. METHODS Three groups of denture resin samples (n = 27) were fabricated: milled (Ivotion, Ivoclar Vivadent), 3D-printed (Saremco Print Denturetec), and heat-polymerized PMMA controls. Samples (8 mm × 3 mm) were CAD-designed, manufactured, quality-checked, and sterilized. C. albicans (GEGE1122.01) biofilms were grown on resin discs, incubated at 37 °C for 16 h, detached, and quantified. SEM analysis assessed biofilm morphology. Statistical analysis was conducted using non-parametric tests (p < 0.05). RESULTS The lowest median biofilm attachment (1.2 × 10⁶ CFU/biofilm, IQR: 3.4 × 10⁵ - 5.6 × 10⁶) with high variability was observed in the control group. The milled group displayed significantly higher biofilm formation (6.4 × 10⁶ CFU/biofilm, IQR: 5.9 × 10⁶ - 7.6 × 10⁶, p = 0.0051) with least variation. The biofilm attachment on 3D-printed discs (4.8 × 10⁶ CFU/biofilm, IQR: 3.6 × 10⁶ - 6.2 × 10⁶) was intermediate between the control and milled groups. SEM findings revealed sparse microcolonies with dense, multi-layered biofilms of yeast and pseudohyphal forms in the control and milled groups. 3D-printed group had moderately dense biofilms, where yeast and pseudohyphae were dominant, but true hyphae were also consistently observed. CONCLUSION This study reveals significant differences in the C. albicans biofilm formation across the resin types, with 3D-printed surfaces showing increased hyphal growth and potential for higher virulence. CLINICAL SIGNIFICANCE Understanding the impact of fabrication methods on microbial colonization is essential for improving denture hygiene and patient outcomes. Dentists and prosthodontists should consider these findings when selecting materials for patients at high risk of fungal infections, such as immunocompromised individuals or elderly denture wearers.
Collapse
Affiliation(s)
- Géraldine Vetsch
- Clinic of General, Special Care and Geriatric Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Daniel Manoil
- Division of cariology and endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Claudine Wulfman
- Innovative Dental Materials and Interfaces Research Unit (UR4462), Department of Prosthodontics, Faculty of Dentistry, University of Paris, France
| | - Ana Parga
- Division of cariology and endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stéphane Durual
- Biomaterials Laboratory, Division of Fixed Prosthodontics and Biomaterials, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Murali Srinivasan
- Clinic of General, Special Care and Geriatric Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland; Honorary Professor, Center of Excellence in Precision Medicine and Digital Health, Center of Excellance in Genomics and Precision Dentistry, Geriatric Dentistry and Special Patients Care International Program, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
4
|
Niitani Y, Ohta K, Yano K, Kaneyasu Y, Maehara T, Kitasaki H, Shigeishi H, Nishi H, Nishimura R, Naito M, Shiba F, Kawada-Matsuo M, Komatsuzawa H, Takemoto T. Effects of Nisin A Combined with Antifungal Drug Against Growth of Candida Species. Dent J (Basel) 2025; 13:160. [PMID: 40277490 PMCID: PMC12025931 DOI: 10.3390/dj13040160] [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: 08/19/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Nisin A, an antimicrobial peptide produced by Lactococcus lactis, primarily shows antimicrobial activity against Gram-positive bacteria, with efficacy increased when used in combination with an antimicrobial drug. On the other hand, oral candidiasis, caused by Candida, occurs in immunocompromised patients and requires antifungal therapy. However, antifungal drug-resistant Candida strains are increasing worldwide, leading to serious problems. Methods: To examine the effects of nisin A against Candida species, we investigated the combined effects of nisin A and antifungal drugs on the growth and viability of Candida strains. Results: While nisin A alone had no antifungal effect, together with amphotericin (AMPH), it showed synergistic effects towards C. albicans, as well as the non-albican strains C. glabrata, C tropicalis, and C. parapsilosis in checkerboard assay results. Furthermore, nisin A with miconazole (MCZ) or micafungin (MCFG) demonstrated a synergistic or additive effect on those strains. Cell viability assay results showed that nisin A enhanced the fungicidal activity of AMPH against both C. albicans and C. glabrata. Biofilm reduction assays showed that nisin A with AMPH, MCZ, or MCFG inhibited biofilm activity against C. albicans as compared with each antifungal drug alone. Finally, nisin A with AMPH, MCZ, or MCFG resulted in a reduced minimum inhibitory concentration of those antifungal drugs against clinically isolated C. albicans and C. glabrata.Conclusions: When used in combination with nisin A, the antifungal drug dosage can be lowered, thus helping to prevent adverse side effects and the emergence of drug-resistant oral Candida species.
Collapse
Affiliation(s)
- Yoshie Niitani
- Department of Oral Health Management, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan;
| | - Kouji Ohta
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan; (K.O.); (K.Y.); (Y.K.); (T.M.); (H.K.); (H.S.)
| | - Kanako Yano
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan; (K.O.); (K.Y.); (Y.K.); (T.M.); (H.K.); (H.S.)
| | - Yoshino Kaneyasu
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan; (K.O.); (K.Y.); (Y.K.); (T.M.); (H.K.); (H.S.)
| | - Tomoko Maehara
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan; (K.O.); (K.Y.); (Y.K.); (T.M.); (H.K.); (H.S.)
| | - Honami Kitasaki
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan; (K.O.); (K.Y.); (Y.K.); (T.M.); (H.K.); (H.S.)
| | - Hideo Shigeishi
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan; (K.O.); (K.Y.); (Y.K.); (T.M.); (H.K.); (H.S.)
| | - Hiromi Nishi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan;
| | - Rumi Nishimura
- Department of Oral Epidemiology, Graduate of School Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (R.N.); (M.N.)
| | - Mariko Naito
- Department of Oral Epidemiology, Graduate of School Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (R.N.); (M.N.)
| | - Fumie Shiba
- Collaborative Research Laboratory of Oral Inflammation Regulation, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan;
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.K.-M.); (H.K.)
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.K.-M.); (H.K.)
| | - Toshinobu Takemoto
- Department of Oral Health Management, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan;
| |
Collapse
|
5
|
Hasan SF, Sidkey NM, Elkhouly HI. Isolation and characterization of a phage against the multidrug-resistant Candida albicans ATCC 10231 strain from raw sewage. Microb Pathog 2025; 204:107562. [PMID: 40210137 DOI: 10.1016/j.micpath.2025.107562] [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: 12/27/2024] [Revised: 03/05/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
The financial cost of resistance to antibiotics is constantly increasing. Viruses are possible substitutes and effective treatments for diseases caused by multidrug-resistant pathogens. The aim of the present study is to isolate a Candida albicans virus from raw sewage and characterize it. Herein, direct and enrichment approaches were used for virus isolation from a raw sewage sample collected from the El-Rahmania wastewater treatment plant, El-Behiera Governorate, Egypt. Using a transmission electron microscope, the viral particle was found to have a hexagonal head with a diameter of approximately 160 nm and a short tail with a length of 87 nm. The virus was treated at different temperature ranges (30-90 °C) for 5, 15, 30, and 60 min. The virus titer was stable at 30 °C, 40 °C, and 50 °C for 5-60 min and partially stable at 60 °C, 70 °C, and 80 °C. The virus kept its activity at a wide range between pH 5-10, while it was completely inactivated in highly acidic (pH < 5) and alkaline (pH > 10) conditions. The effect of osmotic shock on the isolated virus showed 64 % survivability. The effects of five organic solvents on viral particles-chloroform, ethanol, methanol, dimethyl sulfoxide (DMSO), and ethyl acetate-showed 0.7, 1.1, 1.2, 1.4, and 1.7 log10 reductions in initial viral titer, respectively. This study provided new information about the characterization of the virus isolated against Candida albicans, which will be useful for future formulation of a successful therapeutic viral agent against Candida albicans.
Collapse
Affiliation(s)
- Seham F Hasan
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Yossuf Abbas st., P.O. 11754, Cairo, Nasr City, Egypt.
| | - Nagwa M Sidkey
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Yossuf Abbas st., P.O. 11754, Cairo, Nasr City, Egypt.
| | - Heba I Elkhouly
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Yossuf Abbas st., P.O. 11754, Cairo, Nasr City, Egypt.
| |
Collapse
|
6
|
Liu Z, Ma X, Zeng X, Li Z, Liu R, Luo R, Wang W, Tahir MS, Wang C, Gu Y. Identification of the ADH gene family in Trichosporon asahii and the role of TaADH_like in pathogenicity and fluconazole resistance. BMC Genomics 2025; 26:352. [PMID: 40197151 PMCID: PMC11978018 DOI: 10.1186/s12864-025-11546-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
Alcohol dehydrogenase has been studied in regulation of fungal growth and development, stress response and pathogenesis, but its function in T. asahii remains unexplored. In this study, we analyzed the ADH gene family in T. asahii for the first time, identifying six ADH genes and containing conserved ADH_N and ADH_Zinc_N domains. We constructed an overexpression strain of the most significantly differentially expressed gene TaADH_like and compared its phenotypes with those of the wild-type strain, focusing on colony morphology, biofilm biomass, stress response, drug resistance, and pathogenicity. The results showed that TaADH_like overexpression reduced sensitivity to hypoxic conditions, altered the hyphae-to-yeast transition, and led to slower growth, decreased colonization ability, reduced tissue damage, and lower lethality. Increased osmotic stress sensitivity and the involvement of the HOG MAPK pathway in the hyphae-to-yeast conversion contributed to the reduced colonization capacity of T. asahii. Furthermore, the overexpression of TaADH_like promoted biofilm formation and led to a slight enhancement in fluconazole resistance in T. asahii. This study is the first to elucidate the function of the alcohol dehydrogenase gene in T. asahii, providing a foundation for future genetic research on this pathogen.
Collapse
Affiliation(s)
- Zhen Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xiangwen Zeng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhiguo Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ruiguo Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Rongyan Luo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Weichen Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Muhammad Salman Tahir
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chengdong Wang
- China Conservation and Research Center for the Giant Panda, Chengdu, 611800, China
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
7
|
Schmidt S, Schwerk C, Schroten H, Ishikawa H, Schubert R, Lehrnbecher T, Rudolph H. Hyphal penetration is the major pathway of translocation of Candida albicans across the blood-cerebrospinal fluid barrier. Fluids Barriers CNS 2025; 22:34. [PMID: 40181464 PMCID: PMC11969880 DOI: 10.1186/s12987-025-00644-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Despite the availability of potent antifungal compounds, invasive fungal disease poses significant morbidity and mortality in immunocompromised patients. Candida albicans is one of the leading pathogens in this setting, and may affect the central nervous system (CNS), which is an extremely severe form of the infection. As the exact pathogenesis of Candida CNS infection is not clear, we investigated the mechanisms and effects of C. albicans transmigration into the CNS, which will be helpful for diagnosis, prevention and treatment. METHODS We used a human in vitro model of the Blood-Cerebrospinal Fluid Barrier (BCSFB), and we investigated the mechanisms of Candida albicans translocation into the CNS. Translocation was evaluated using immunofluorescence analysis focusing on tight and adherens junctions and the actin cytoskeleton. Barrier integrity was monitored via measurement of transepithelial resistance and the paracellular permeability of dextran. LIVE/DEAD assays were applied for viability controls and a cytometric bead array was performed to detect cytokine secretion of plexus epithelial cells. RESULTS Translocation at low doses occurs transcellularly in the absence of cytotoxicity or secretion of proinflammatory cytokines. This is accomplished by the formation of a tunnel-like structure exploiting the actin cytoskeleton. With higher infection doses of Candida albicans, a reduction in barrier integrity due to disruption of tight and adherens junctions was observed and cytotoxicity also increased. CONCLUSION Our findings reveal that Candida albicans can use transcellular translocation to invade into the CNS and is able to circumvent major host immune response, which may impact on diagnostic and preventive strategies.
Collapse
Affiliation(s)
- S Schmidt
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - C Schwerk
- Pediatric Infectious Diseases, Medical Faculty Mannheim, University Children's Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - H Schroten
- Pediatric Infectious Diseases, Medical Faculty Mannheim, University Children's Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - H Ishikawa
- Laboratory of Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - R Schubert
- Department of Pediatrics, Division of Pneumology, Allergology, Infectious Diseases and Gastroenterology, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - T Lehrnbecher
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - H Rudolph
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt/Main, Germany.
| |
Collapse
|
8
|
Aoki K, Ohkuma M, Sugita T, Kobayashi Y, Tanaka N, Takashima M. Analyses of hyphal diversity in Trichosporonales yeasts based on fluorescent microscopic observations. Microbiol Spectr 2025; 13:e0321024. [PMID: 39998240 PMCID: PMC11960055 DOI: 10.1128/spectrum.03210-24] [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: 12/08/2024] [Accepted: 02/09/2025] [Indexed: 02/26/2025] Open
Abstract
In dimorphic yeasts, hyphal growth is primarily associated with infection and mycosis progression, with Trichosporon asahii causing deep-seated mycosis and summer-type hypersensitivity pneumonitis. Magnesium accelerates hyphal growth in T. asahii, leading to multi-septation, vacuolar expansion, and decreased lipid droplet size. However, the commonality of these phenotypes has not been studied in Trichosporonales yeasts. Therefore, to explore whether similar magnesium-induced phenotypes occur across Trichosporonales yeasts, we examined hyphal growth, multi-septation, vacuolar extension, and lipid droplet size and number in 30 species. Cell length increased with magnesium treatment in 13 yeasts: 5 Trichosporon (T. asahii, Trichosporon aquatile, Trichosporon asteroides, Trichosporon coremiiforme, and Trichosporon ovoides), three Apiotrichum (Apiotrichum brassicae, Apiotrichum montevideense, and Apiotrichum veenhuisii), three Cutaneotrichosporon (Cutaneotrichosporon cavernicola, Cutaneotrichosporon cutaneum, and Cutaneotrichosporon dermatis), Pascua guehoae, and Takashimella koratensis. C. dermatis also underwent pseudo-hyphal growth. Multi-septation increased in seven dimorphic yeasts, including five Trichosporon spp., Trichosporon faecale, and C. dermatis. The vacuolar area was significantly extended in T. asahii, T. aquatile, T. ovoides, and C. dermatis. Lipid droplet size increased only in Trichosporon inkin; however, it decreased in T. asahii, T. coremiiforme, and T. faecale. Additionally, lipid droplet number was preferentially altered in Apiotrichum and Cutaneotrichosporon. These results suggested that magnesium-induced multi-septation and vacuolar area expansion phenotypically distinguish Trichosporon hyphae from Apiotrichum and Cutaneotrichosporon hyphae and distinguish C. dermatis pseudo-hyphae from Cutaneotrichosporon spp. Moreover, differences in lipid droplet size can discriminate species within Trichosporon. Our results suggest that phenotypic alteration via magnesium treatment can contribute to the characterization of Trichosporonales yeasts. These findings provide insights into fungal pathogenesis and may support new treatment strategies.IMPORTANCEMagnesium sulfate considerably affects hyphal growth and cellular organization in Trichosporon asahii. To examine the commonality of this phenotype in Trichosporonales, we treated 30 Trichosporonales yeasts with magnesium sulfate and observed genus-level phenotypic alterations. Using cell length measurement, lipid droplet staining, septum staining, and vacuole staining, considerable hyphal diversity was observed among Trichosporonales. Notably, differences in the multi-septation phenotype and vacuolar size distinguished Trichosporon hyphae from Apiotrichum and Cutaneotrichosporon hyphae and distinguished Cutaneotrichosporon dermatis from other Cutaneotrichosporon spp. Moreover, differences in lipid droplet phenotype divided Trichosporon hyphae into two groups. Our study revealed the relationship between hyphal morphology and phylogenetic systematics in Trichosporonales.
Collapse
Affiliation(s)
- Keita Aoki
- Laboratory of Yeast Systematics, Tokyo NODAI Research Institute, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| | - Moriya Ohkuma
- Japan Collection of Microorganisms, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Yuuki Kobayashi
- Laboratory of Yeast Systematics, Tokyo NODAI Research Institute, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| | - Naoto Tanaka
- Department of Molecular Microbiology, Faculty of Life Sciences, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| | - Masako Takashima
- Laboratory of Yeast Systematics, Tokyo NODAI Research Institute, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| |
Collapse
|
9
|
De Paiva Macedo J, Watanabe AAS, Machado ABF, Diniz CG, Da Silva VL, Dias VC. Unveiling antifungal resistance and biocide tolerance in clinical isolates of Candida spp. Future Microbiol 2025; 20:457-468. [PMID: 40119655 PMCID: PMC11980505 DOI: 10.1080/17460913.2025.2480924] [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: 01/06/2025] [Accepted: 03/14/2025] [Indexed: 03/24/2025] Open
Abstract
AIMS Candidiasis, caused by Candida spp. is an opportunistic infection with significant healthcare risks, worsened by trends in antifungal resistance. This study aimed to evaluate the antifungal susceptibility profile, investigate resistance mechanisms, assess efflux pump activity, and examine biocide tolerance in clinical Candida isolates. METHODS A total of 100 Candida isolates from hospitalized and outpatient individuals were analyzed for their antifungal susceptibility profile, molecular resistance mechanisms through PCR, efflux pump activity with the Cartwheel method, and biocide tolerance (sodium hypochlorite, hydrogen peroxide, and benzalkonium chloride), which was assessed by disk diffusion. RESULTS A high prevalence of resistance (87%) to at least one antifungal was observed, with 47.12% of isolates showing simultaneous multiple resistance to three azole derivatives. The highest antifungal agent resistance was observed for fluconazole (n = 70) and the highest susceptibility for amphotericin B (n = 1). The most common mutation was in the ERG11 gene (n = 38/43.7%). Efflux pump activity was detected in both C. albicans and non-albicans Candida species. Biocide testing revealed a higher tolerance for sodium hypochlorite, with an inhibition zone ranging from 18.25 (4.40) to 34.0 (4.00). CONCLUSIONS This study highlights significant antifungal resistance in Candida spp. particularly to azoles, stressing the need for improved infection control and novel therapeutic strategies.
Collapse
Affiliation(s)
- Jamile De Paiva Macedo
- Department of Biological Sciences, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Aripuana Aranha Sakurada Watanabe
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Alessandra Barbosa Ferreira Machado
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Cláudio Galuppo Diniz
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Vania Lucia Da Silva
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| | - Vanessa Cordeiro Dias
- Department of Parasitology, Microbiology, and Immunology, Federal University of Juiz de Fora – UFJF Rua José Lourenço Kelmer, Juiz de Fora, Brazil
| |
Collapse
|
10
|
Banda-Flores IA, Torres-Tirado D, Mora-Montes HM, Pérez-Flores G, Pérez-García LA. Resilience in Resistance: The Role of Cell Wall Integrity in Multidrug-Resistant Candida. J Fungi (Basel) 2025; 11:271. [PMID: 40278091 PMCID: PMC12028102 DOI: 10.3390/jof11040271] [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: 02/05/2025] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
The Candida species cell wall plays a pivotal role as a structural and functional barrier against external aggressors and as an intermediary in host-pathogen interactions. Candida species exhibit unique adaptations in their cell wall composition, with varying proportions of chitin, mannans, and β-glucans influenced by the environmental conditions and the morphological states. These components not only maintain cellular viability under osmotic, thermal, and chemical stress, but also serve as the key targets for novel antifungal strategies. MAPK signaling pathways, like the cell wall integrity pathway and the high-osmolarity glycerol pathway, play a crucial role in responding to cell wall stressors. Due to the rise of antifungal resistance and its clinical challenges, there is a need to identify new antifungal targets. This review discusses the recent advances in understanding the mechanisms underlying cell wall integrity, their impact on antifungal resistance and virulence, and their potential as therapeutic targets of C. albicans, N. glabratus, and C. auris.
Collapse
Affiliation(s)
- Iván A. Banda-Flores
- Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Romualdo del Campo 501, Fracc. Rafael Curiel, Ciudad Valles 79060, San Luis Potosi, Mexico; (I.A.B.-F.); (D.T.-T.); (G.P.-F.)
| | - David Torres-Tirado
- Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Romualdo del Campo 501, Fracc. Rafael Curiel, Ciudad Valles 79060, San Luis Potosi, Mexico; (I.A.B.-F.); (D.T.-T.); (G.P.-F.)
| | - Héctor M. Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, Col. Noria Alta, Guanajuato 36050, Guanajuato, Mexico;
| | - Gabriela Pérez-Flores
- Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Romualdo del Campo 501, Fracc. Rafael Curiel, Ciudad Valles 79060, San Luis Potosi, Mexico; (I.A.B.-F.); (D.T.-T.); (G.P.-F.)
| | - Luis A. Pérez-García
- Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Romualdo del Campo 501, Fracc. Rafael Curiel, Ciudad Valles 79060, San Luis Potosi, Mexico; (I.A.B.-F.); (D.T.-T.); (G.P.-F.)
| |
Collapse
|
11
|
Amann V, Kissmann AK, Firacative C, Rosenau F. Biofilm-Associated Candidiasis: Pathogenesis, Prevalence, Challenges and Therapeutic Options. Pharmaceuticals (Basel) 2025; 18:460. [PMID: 40283897 PMCID: PMC12030374 DOI: 10.3390/ph18040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The rising prevalence of fungal infections, especially those caused by Candida species, presents a major risk to global health. With approximately 1.5 million deaths annually, the urgency for effective treatment options has never been greater. Candida spp. are the leading cause of invasive infections, significantly impacting immunocompromised patients and those in healthcare settings. C. albicans, C. parapsilosis and the emerging species C. auris are categorized as highly dangerous species because of their pathogenic potential and increasing drug resistance. This review comparatively describes the formation of microbial biofilms of both bacterial and fungal origin, including major pathogens, thereby creating a novel focus. Biofilms can further complicate treatment, as these structures provide enhanced resistance to antifungal therapies. Traditional antifungal agents, including polyenes, azoles and echinocandins, have shown effectiveness, yet resistance development continues to rise, necessitating the exploration of novel therapeutic approaches. Antimicrobial peptides (AMPs) such as the anti-biofilm peptides Pom-1 and Cm-p5 originally isolated from snails represent promising candidates due to their unique mechanisms of action and neglectable cytotoxicity. This review article discusses the challenges posed by Candida infections, the characteristics of important species, the role of biofilms in virulence and the potential of new therapeutic options like AMPs.
Collapse
Affiliation(s)
- Valerie Amann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Ann-Kathrin Kissmann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Carolina Firacative
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogota 111221, Colombia;
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| |
Collapse
|
12
|
Horn S, Schmid M, Berest I, Piattini F, Zhang J, de Bock K, Devuyst O, Nlandu Khodo S, Kisielow J, Kopf M. IL-1 protects from fatal systemic candidiasis in mice by inhibiting oxidative phosphorylation and hypoxia. Nat Commun 2025; 16:2626. [PMID: 40097388 PMCID: PMC11914259 DOI: 10.1038/s41467-025-57797-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Invasive C. albicans infections result in high mortality rates. While IL-1 is important to combat C. albicans infections, the underlying mechanisms remain unclear. Using global and conditional Il1r1 knockouts in mice, here we show that IL-1R signaling in non-hematopoietic cells in the kidney and brain is crucial for a protective response. In the kidney, endothelial IL-1R contributes to fungal clearance independent of neutrophil recruitment, while IL-1R in hematopoietic cells is dispensable. IL-1R signaling indirectly recruits neutrophils and monocytes in the brain by regulating chemokines and adhesion molecules. Single-nucleus-RNA-sequencing data implicates excessive metabolic activity and oxidative phosphorylation across all cell types in the kidney of Il1r1-deficient mice within a few hours upon infection, with associated, localized hypoxia at infection foci. Lastly, we find that hypoxia promotes fungal growth and pathogenicity. In summary, our results show that IL-1R-signaling in non-hematopoietic cells is required to prevent fatal candidiasis by inhibiting a metabolic shift, including excessive oxidative phosphorylation and hypoxia.
Collapse
Affiliation(s)
- Sofia Horn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Mareike Schmid
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ivan Berest
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Federica Piattini
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Jing Zhang
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zurich, Zurich, Switzerland
| | - Katrien de Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zurich, Zurich, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Jan Kisielow
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Tosiano MA, Lanni F, Mitchell AP, McManus CJ. Roles of P-body factors in Candida albicans filamentation and stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.09.602714. [PMID: 40161774 PMCID: PMC11952329 DOI: 10.1101/2024.07.09.602714] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Hyphal growth is strongly associated with virulence in the human fungal pathogen Candida albicans. While hyphal transcriptional networks have been the subject of intense study, relatively little is known about post-transcriptional regulation. Previous work reported that P-Body (PB) factors Dhh1 and Edc3 were required for C. albicans virulence and filamentation, suggesting an essential role for post-transcriptional regulation of these processes. However, the molecular roles of these factors have not been determined. To further study the function of PB factors in filamentation, we generated homozygous deletions of DHH1 and EDC3 in diverse prototrophic clinical strains using transient CRISPR-Cas9. Homozygous DHH1 deletion strongly impaired growth, altered filamentation, and exhibited unusual colony morphology in response to heat stress in five strain backgrounds. Using RNA-seq, we found DHH1 deletion disrupts the regulation of thousands of genes under both yeast and hyphal growth conditions in SC5314 and P57055. This included upregulation of many stress response genes in the absence of external stress, similar to deletion of the S. cerevisiae DHH1 homolog. In contrast, we found EDC3 was not required for heat tolerance or filamentation in diverse strains. These results support a model in which DHH1, but not EDC3, represses hyphal stress response transcripts in yeast and remodels the transcriptome during filamentation. Our work supports distinct requirements for specific mRNA decay factors, bolstering evidence for post-transcriptional regulation of filamentation in C. albicans.
Collapse
Affiliation(s)
- Melissa A. Tosiano
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
14
|
Tosiano MA, Lanni F, Mitchell AP, McManus CJ. Roles of P-body factors in Candida albicans filamentation and stress response. PLoS Genet 2025; 21:e1011632. [PMID: 40096135 PMCID: PMC11975087 DOI: 10.1371/journal.pgen.1011632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 04/07/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
Hyphal growth is strongly associated with virulence in the human fungal pathogen Candida albicans. While hyphal transcriptional networks have been the subject of intense study, relatively little is known about post-transcriptional regulation. Previous work reported that P-Body (PB) factors Dhh1 and Edc3 were required for C. albicans virulence and filamentation, suggesting an essential role for post-transcriptional regulation of these processes. However, the molecular roles of these factors have not been determined. To further study the function of PB factors in filamentation, we generated homozygous deletions of DHH1 and EDC3 in diverse prototrophic clinical strains using transient CRISPR-Cas9. Homozygous DHH1 deletion strongly impaired growth, altered filamentation, and exhibited unusual colony morphology in response to heat stress in five strain backgrounds. Using RNA-seq, we found DHH1 deletion disrupts the regulation of thousands of genes under both yeast and hyphal growth conditions in SC5314 and P57055. This included upregulation of many stress response genes in the absence of external stress, similar to deletion of the S. cerevisiae DHH1 homolog. In contrast, we found EDC3 was not required for heat tolerance or filamentation in diverse strains. These results support a model in which DHH1, but not EDC3, represses hyphal stress response transcripts in yeast and remodels the transcriptome during filamentation. Our work supports distinct requirements for specific mRNA decay factors, bolstering evidence for post-transcriptional regulation of filamentation in C. albicans.
Collapse
Affiliation(s)
- Melissa A. Tosiano
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
15
|
Williams C, Carnahan BR, Hyland SN, DeMeester KE, Grimes CL. Bio-orthogonal Labeling of Chitin in Native Pathogenic Candida Species via the Chitin Scavenge Pathway. J Am Chem Soc 2025; 147:5632-5641. [PMID: 39925016 PMCID: PMC11849683 DOI: 10.1021/jacs.4c11554] [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: 08/21/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
The fungal cell wall is essential for the integrity of the cell, providing strength and shape, as well as protection against environmental stimuli. For pathogenic fungi, the cell wall is also the initial point of contact with the host. Specific cell wall features such as hypha tails and smaller glycan components modulate a wide range of fungal interactions with the immune defenses. Here, a bio-orthogonal labeling method utilizing N-acetyl-glucosamine (NAG) probes is developed to fluorescently label native, pathogenic yeast via the chitin scavenging pathway. A panel of NAG probes was assembled, synthesized, and characterized for the ability to label the chitin in pathogenic yeast. Enzymatic data show that the native scavenging biosynthetic enzyme, Hxk1, is promiscuous, permitting the labeling of the native chitin biopolymer. This chitin labeling method was validated via the development of mass spectrometry protocols. When compared to the current available labeling systems for chitin, the probes do not affect the integrity of the cell wall and do not interrupt cell growth. Furthermore, the NAG probes enabled multiple "click" platforms across pathogenic Candida species including Candida albicans and Candida tropicalis. Budding and filamentous hyphal states were observed. The results indicate the probes' utility for in vivo study of the morphological, pathogenic switch, and visualization of growth patterns. Thus, the use of these probes in pathogenic Candida strains is ideal for a variety of future applications including strain specific antifungals, diagnostic tools, and immunomodulators.
Collapse
Affiliation(s)
- Caroline Williams
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark, Delaware 19716, United States
| | - Bella R. Carnahan
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark, Delaware 19716, United States
| | - Stephen N. Hyland
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark, Delaware 19716, United States
| | - Kristen E. DeMeester
- Department
of Chemistry, Lafayette College, Easton, Pennsylvania 18042, United States
| | - Catherine L. Grimes
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark, Delaware 19716, United States
- Department
of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
16
|
Huang LZY, Penman R, Kariuki R, Vaillant PHA, Gharehgozlo S, Shaw ZL, Truong VK, Vongsvivut J, Elbourne A, Caruso RA. Graveyard effects of antimicrobial nanostructured titanium over prolonged exposure to drug resistant bacteria and fungi. NANOSCALE 2025; 17:3170-3188. [PMID: 39713977 DOI: 10.1039/d4nr03238a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Innovations in nanostructured surfaces have found a practical place in the medical area with use in implant materials for post-operative infection prevention. These textured surfaces should be dual purpose: (1) bactericidal on contact and (2) resistant to biofilm formation over prolonged periods. Here, hydrothermally etched titanium surfaces were tested against two highly antimicrobial resistant microbial species, methicillin-resistant Staphylococcus aureus and Candida albicans. Two surface types - unmodified titanium and nanostructured titanium - were incubated in a suspension of each microbial strain for 1 day and 7 days. Surface topography and cross-sectional information of the microbial cells adhered to the surfaces, along with biomass volume and live/dead rate, showed that while nanostructured titanium was able to kill microbes after 1 day of exposure, after 7 days, the rate of death becomes negligible when compared to the unmodified titanium. This suggests that as biofilms mature on a nanostructured surface, the cells that have lysed conceal the nanostructures and prime the surface for planktonic cells to adhere, decreasing the possibility of structure-induced lysis. Synchrotron macro-attenuated total reflection Fourier transform infrared (macro ATR-FTIR) micro-spectroscopy was used to elucidate the biochemical changes occurring following exposure to differing surface texture and incubation duration, providing further understanding into the effects of surface morphology on the biochemical molecules (lipids, proteins and polysaccharides) in an evolving and growing microbial colony.
Collapse
Affiliation(s)
- Louisa Z Y Huang
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Rowan Penman
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Rashad Kariuki
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Pierre H A Vaillant
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Soroosh Gharehgozlo
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Z L Shaw
- School of Engineering, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
| | - Vi Khanh Truong
- Healthcare Engineering Innovation Group, Department of Biomedical Engineering & Biotechnology, College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Jitraporn Vongsvivut
- Infrared Microspectroscopy (IRM) Beamline, ANSTO - Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Aaron Elbourne
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Rachel A Caruso
- School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
17
|
Matsumoto Y, Kurakado S, Yamada T, Sugita T. Strategy to Identify Virulence-Related Genes of the Pathogenic Fungus Trichosporon asahii Using an Efficient Gene-Targeting System. Microbiol Immunol 2025; 69:77-84. [PMID: 39660720 DOI: 10.1111/1348-0421.13192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Trichosporon asahii is a pathogenic fungus that causes severe deep-seated mycosis in immunocompromised patients with neutropenia. Understanding the molecular mechanisms of T. asahii infection will facilitate the development of new therapeutic and preventive strategies. Two main obstacles have prevented the identification of virulence-related genes in T. asahii using molecular genetic techniques: the lack of experimental animal infection models for easy evaluation of T. asahii virulence and the lack of genetic recombination technology for T. asahii. To address these issues, we developed a silkworm infection model to quantitatively evaluate T. asahii virulence and a genetic recombination method to generate gene-deficient T. asahii mutants, enabling the identification of virulence factors of T. asahii. In this review, we propose a strategy for identifying virulence-related factors in T. asahii using a silkworm infection model and an efficient gene-targeting system.
Collapse
Affiliation(s)
- Yasuhiko Matsumoto
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Sanae Kurakado
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Tsuyoshi Yamada
- Teikyo University Institute of Medical Mycology, Teikyo University, Hachioji, Tokyo, Japan
- Asia International Institute of Infectious Disease Control, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| |
Collapse
|
18
|
Praetorius JP, Hitzler SUJ, Gresnigt MS, Figge MT. Image-based quantification of Candida albicans filamentation and hyphal length using the open-source visual programming language JIPipe. FEMS Yeast Res 2025; 25:foaf011. [PMID: 40082735 PMCID: PMC11963753 DOI: 10.1093/femsyr/foaf011] [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: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/16/2025] Open
Abstract
The formation of hyphae is one of the most crucial virulence traits the human pathogenic fungus Candida albicans possesses. The assessment of hyphal length in response to various stimuli, such as exposure to human serum, provides valuable insights into the adaptation strategies of C. albicans to the host environment. Despite the increasing high-throughput capacity live-cell imaging and data generation, the accurate analysis of hyphal growth has remained a laborious, error-prone, and subjective manual process. We developed an analysis pipeline utilizing the open-source visual programming language Java Image Processing Pipeline (JIPipe) to overcome the limitations associated with manual analysis of hyphal growth. By comparing our automated approach with manual analysis, we refined the strategies to achieve accurate differentiation between yeast cells and hyphae. The automated method enables length measurements of individual hyphae, facilitating a time-efficient, high-throughput, and user-friendly analysis. By utilizing this JIPipe analysis approach, we obtained insights into the filamentation behavior of two C. albicans strains when exposed to human serum albumin (HSA), the most abundant protein in human serum. Our findings indicate that despite the known role of HSA in stimulating fungal growth, it reduces filamentous growth. The implementation of our automated JIPipe analysis approach for hyphal growth represents a long-awaited and time-efficient solution to meet the demand of high-throughput data generation. This tool can benefit different research areas investigating the virulence aspects of C. albicans.
Collapse
Affiliation(s)
- Jan-Philipp Praetorius
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), 07745 Jena, Germany
| | - Sophia U J Hitzler
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), 07745 Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, 07745 Jena, Germany
| |
Collapse
|
19
|
Chu F, Wu H, Li C, Qiu W, Zang L, Wu D, Shao J, Wang T, Wang C. Transcriptomics analysis reveals the effect of Pulsatilla decoction butanol extract on endoplasmic reticulum and peroxisome function of Candida albicans in hyphal state. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118826. [PMID: 39288826 DOI: 10.1016/j.jep.2024.118826] [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: 07/05/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional Chinese medicine formula known as Pulsatilla decoction was utilized to treat conditions such as bacterial dysentery, ulcerative colitis, and fungal infections like vulvovaginal candidiasis (VVC) caused by Candida albicans (C. albicans). In our prior research, it was shown that the n-butanol extract from Pulsatilla Decoction (BEPD) exhibited effective inhibition of C. albicans. Nevertheless, the exact mechanism by which BEPD hinders hyphal growth, a critical virulence factor of C. albicans, remains unclear. AIM OF THE STUDY In the present study, the inhibitory effect and mechanism of the BEPD on C. albicans hyphal growth was predicted by transcriptome analysis, and further verified by in vitro and in vivo experiments. MATERIALS AND METHODS The BEPD was prepared and C. albicans was cultured to induce the hyphal state. Transcriptome analysis was conducted to predict the significant difference in enrichment genes and signaling pathways in the inhibitory effect of BEPD on C. albicans hyphae. Various methods, such as spot assay, time-growth curve analysis, Confocal laser scanning microscope (CLSM), scanning electron microscope (SEM), transmission electron microscope (TEM), flow cytometry, and spectrophotometer, were used to assess the effect of BEPD on hyphal structure and growth activity, lipid peroxidation level, peroxidase (CAT) activity, superoxide dismutase (SOD) activity, and apoptosis of C. albicans. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was employed to examine the expression levels of genes associated with endoplasmic reticulum and peroxisome function. The VVC model was employed to evaluate the influence of BEPD on the growth of C. albicans hyphae in vivo. RESULT The growth of C. albicans hyphae on solid culture media was significantly inhibited by BEPD. CLSM showed that the length of C. albicans hyphae was decreased and their vitality was lowered. SEM indicated that the hyphae of C. albicans were fractured, while TEM revealed damage to the organelles within the cells. GO enrichment and KEGG pathways analysis from transcriptomic data demonstrated that BEPD effectively suppressed the functioning of the endoplasmic reticulum and peroxisomes in C. albicans hyphae. RT-qPCR verified the decreased expression of genes associated with endoplasmic reticulum and peroxisome function by BEPD. Investigation of the endoplasmic reticulum revealed that BEPD elevated levels of reactive oxygen species (ROS) and apoptosis, indicating endoplasmic reticulum stress, as well as malondialdehyde (MDA), a marker of oxidative stress. Additionally, BEPD was shown to lower the activities of catalase (CAT) and superoxide dismutase (SOD). In animal trials, BEPD effectively hindered the growth of C. albicans hyphae in the vaginas of mice with VVC, thus reducing immune inflammatory damage to the vaginal mucosa of these mice. CONCLUSION This study demonstrates that BEPD has an inhibitory effect on hyphae, which are an important virulence factor of C. albicans. This effect may be related to BEPD's inhibitory effect on endoplasmic reticulum (ER) and peroxisome function. The findings suggest that BEPD could potentially play a therapeutic role in C. albicans infectious diseases by inhibiting hyphae.
Collapse
Affiliation(s)
- Faqiong Chu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Hui Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Can Li
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Wei Qiu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Long Zang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Jing Shao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Tianming Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Changzhong Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.
| |
Collapse
|
20
|
Liao B, Zhang C, Shen J, Chen D, Wang J, Chen X, Zhou Y, Wei Y, Shi Y, Gou L, Guo Q, Zhou X, Xie H, Zhao L, Liao G, Zhu Z, Cheng L, Zhou X, Li Y, Ren B. Aloin remodels the cell wall of Candida albicans to reduce its hyphal virulence against oral candidiasis. Appl Microbiol Biotechnol 2025; 109:21. [PMID: 39853490 PMCID: PMC11761986 DOI: 10.1007/s00253-025-13411-7] [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: 11/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Aloe vera (L.) Burm.f. is a traditional Chinese medicine known for treating various ailments, including fungal infections. Aloin is one of the major components from A. vera, but its antifungal mechanism and therapeutic potential against oral candidiasis are not clear. This study aimed to examine the mechanism of aloin against Candida albicans and its inhibitory activity against oral candidiasis. In this study, we for the first time found that aloin could induce the formation of abnormal hyphae with smaller hyphal diameters and fewer branching points in C. albicans including 11 clinical isolates without growth inhibition. The transcriptome and further cell wall contents analysis indicated that aloin remodeled the cell wall to increase the contents of β-1,3-glucan and furtherly showed an antagonistic effect with micafungin. Aloin also significantly inhibited the cell damage of oral epithelial cells and oral candidiasis in mice infected by C. albicans due to its inhibitory actions on the hyphal development and expressions of virulence factors, including candidalysin (coded by ECE1). Our results suggest that aloin is a promising antifungal agent for controlling candidiasis and targeting hyphal development and pathogenesis represents a practical strategy for developing new antifungal drugs. KEY POINTS: • Aloin remodels the C. albicans cell wall to form avirulent hyphae. • Aloin inhibits C. albicans infections in oral epithelial cells and mouse mucosa without toxicity. • Aloin is a promising antifungal agent with therapeutic potential against C. albicans infections.
Collapse
Affiliation(s)
- Binyou Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chuanli Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Wei
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hongyu Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lin Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Information Management & Department of Stomatology Informatics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
21
|
Hassan MA, Noor S, Park J, Nabawy A, Dedhiya M, Patel R, Rotello VM. Gelatin Nanoemulsion-Based Co-Delivery of Terbinafine and Essential Oils for Treatment of Candida albicans Biofilms. Microorganisms 2025; 13:127. [PMID: 39858895 PMCID: PMC11767362 DOI: 10.3390/microorganisms13010127] [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: 12/19/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Fungal infections represent a significant global health challenge. Candida albicans is a particularly widespread pathogen, with both molecular and biofilm-based mechanisms making it resistant to or tolerant of available antifungal drugs. This study reports a combination therapy, active against C. albicans, utilizing terbinafine and essential oils incorporated into a gelatin-based nanoemulsion system (T-GNE). Eugenol and methyl eugenol/terbinafine T-GNEs had an additive efficacy, while carvacrol (CT-GNE) worked synergistically with terbinafine, providing effective antifungal treatment with minimal mammalian cell toxicity. Confocal microscopy demonstrated that CT-GNE penetrated the dense C. albicans biofilm and disrupted the fungal cell membrane. Overall, the combination of essential oils with terbinafine in GNE provided a promising treatment for fungal biofilms.
Collapse
Affiliation(s)
- Muhammad Aamir Hassan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA; (M.A.H.); (S.N.); (J.P.); (A.N.); (M.D.)
| | - Sadaf Noor
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA; (M.A.H.); (S.N.); (J.P.); (A.N.); (M.D.)
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA; (M.A.H.); (S.N.); (J.P.); (A.N.); (M.D.)
| | - Ahmed Nabawy
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA; (M.A.H.); (S.N.); (J.P.); (A.N.); (M.D.)
| | - Maitri Dedhiya
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA; (M.A.H.); (S.N.); (J.P.); (A.N.); (M.D.)
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA; (M.A.H.); (S.N.); (J.P.); (A.N.); (M.D.)
| |
Collapse
|
22
|
Singh SK, Pancholi SS. Current Updates on Pathogenesis, Systemic Therapy, and Treatment of Invasive Fungal Infections. Curr Drug Targets 2025; 26:203-220. [PMID: 39421988 DOI: 10.2174/0113894501337502241015121015] [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: 07/16/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024]
Abstract
Numerous health hazards are associated with fungal infections, ranging from asymptomatic cases to potentially fatal invasive diseases that are especially dangerous for those with impaired immune systems. The main causes behind these diseases are opportunistic fungi, namely Aspergillus, Candida, and Cryptococcus. Invasive fungal infections (IFIs) require a global response that includes the development of vaccines, standardized protocols for diagnosis, potent antifungal medications, and strategies to stop drug-resistant strains. Improving high-risk group diagnosis and treatment is essential to lowering death rates. This review highlights the substantial health concerns associated with fungal infections, especially in immunocompromised individuals, and identifies Aspergillus, Candida, and Cryptococcus as the main pathogens. It highlights the necessity of international efforts, such as the development of novel diagnostic instruments, imaging methods, and antifungal drugs, to combat these invasive infections. The review also addresses the increasing need for novel treatment approaches in light of the developing resistance to widely used antifungal medications. Furthermore, the significance of secretory proteins in fungal pathogenicity and the potential of combination therapy are investigated. It is also suggested that a multimodal strategy be used to fight these illnesses, given the promise of multivalent vaccinations. Overall, this study emphasizes how critical it is to develop better diagnostic and treatment strategies in order to successfully control and lessen the impact of invasive fungal diseases on the health of the world.
Collapse
Affiliation(s)
- Sushil Kumar Singh
- Shree S.K. Patel College of Pharmaceutical Education and Research, Department of Pharmaceutical Technology, Ganpat University, Kherva, Mehsana, Gujarat 384012, India
| | - Shyam Sunder Pancholi
- School of Pharmacy and Technology Management, Department of Pharmaceutics (SPTM) SVKM's NMIMS (Deemed to be University) Mukesh Patel Technology Park, Babulde, Shirpur 425405, Dist. Dhule Maharashtra, India
| |
Collapse
|
23
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
24
|
Yuan X, Cao D, Xiang Y, Jiang X, Liu J, Bi K, Dong X, Wu T, Zhang Y. Antifungal activity of essential oils and their potential synergistic effect with amphotericin B. Sci Rep 2024; 14:31125. [PMID: 39732745 DOI: 10.1038/s41598-024-82380-0] [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: 09/30/2024] [Accepted: 12/04/2024] [Indexed: 12/30/2024] Open
Abstract
Candida albicans is a common opportunistic pathogen, causing infections ranging from superficial to bloodstream infections. The limited antifungal options and rising drug resistance challenge clinical treatment. We screened 98 essential oils and identified 48 with antifungal activity against Candida albicans at 1% concentration, determining their minimum inhibitory concentrations (MIC). Of these, 14 maintained fungicidal activity at lower concentrations (0.25% and 0.125%). 5 essential oils (Cinnamon, Satureja montana, Palmarosa, Lemon eucalyptus, and Honey myrtle) showed the highest inhibitory effects on stationary-phase Candida albicans and inhibited hyphae elongation. Synergistic effects were observed when combining Palmarosa with amphotericin B (AmB) against growing-phase Candida albicans, while Cinnamon and Satureja montana with AmB showed superior efficacy against stationary-phase infections. We identified the active components of 5 essential oils using gas chromatography-mass spectrometry (GC-MS) and found the following main constituents: Cinnamon primarily contains benzyl benzoate and eugenol, Satureja montana is dominated by carvacrol and cymene, Palmarosa features geraniol and geranyl acetate, Lemon eucalyptus includes dl-Isopulegol and citronellal, and Honey myrtle is characterized by citral and neral. Our results may aid in developing more effective antifungal treatments.
Collapse
Affiliation(s)
- Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Dan Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yanghui Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiuzhi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jiaying Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Kefan Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xu Dong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tiantian Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ying Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| |
Collapse
|
25
|
Mendes AGG, Campos CDL, Pereira-Filho JL, Pereira APA, Reis GSA, Araújo ÁWDMS, Monteiro PDM, Vidal FCB, Monteiro SG, da Silva Figueiredo IF, Fernandes ES, Monteiro CDA, Monteiro-Neto V. Ellagic Acid Potentiates the Inhibitory Effects of Fluconazole Against Candida albicans. Antibiotics (Basel) 2024; 13:1174. [PMID: 39766564 PMCID: PMC11672414 DOI: 10.3390/antibiotics13121174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Antifungal resistance to azoles, coupled with the increasing prevalence of Candida albicans infections, represents a significant public health challenge and has driven the search for new natural compounds that can act as alternatives or adjuvants to the current antifungals. Ellagic acid (EA) has demonstrated antifungal activity; however, its effects are not fully understood. In this study, we investigated the in vitro anti-Candida activity of EA and its ability to potentiate the effects of fluconazole (FLZ) on C. albicans.Methods: The Minimum Inhibitory Concentration (MIC) of EA was determined by broth microdilution and its interaction with FLZ was assessed using a checkerboard assay. Additionally, we examined the effects of EA on yeast-to-hypha transition, inhibition of biofilm formation, time-kill kinetics, hemolytic activity, and cytotoxicity in HeLa ATCC® CCL-2™ cells. Results: EA exhibited MIC values ranging from 250 to 2000 µg/mL and showed synergistic and additive interactions with FLZ, resulting in a marked reduction in the MIC values of FLZ (up to 32-fold) and EA (up to 16-fold). In the time-kill assay, the most effective combinations were 4× EA MIC, 2× EA MIC, and FIC EA + FLZ, which showed fungicidal activity. Furthermore, EA did not show hemolytic activity and demonstrated low and dose-dependent cytotoxicity in HeLa cells, with no cytotoxic effects observed in combination with FLZ. EA and the synergistic combination of EA and FLZ interfered with both the yeast-to-hypha transition process in C. albicans cells and biofilm formation. In addition to its antifungal efficacy, EA demonstrated a favorable safety profile at the concentrations used. Conclusions: This study presents promising results regarding the potential use of EA in combination with FLZ for the treatment of C. albicans infections.
Collapse
Affiliation(s)
- Amanda Graziela Gonçalves Mendes
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Carmem Duarte Lima Campos
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - José Lima Pereira-Filho
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Aleania Polassa Almeida Pereira
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Gabriel Silva Abrantes Reis
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Árlon Wendel de Marinho Silva Araújo
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Pablo de Matos Monteiro
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Flávia Castello Branco Vidal
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Silvio Gomes Monteiro
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| | - Isabella Fernandes da Silva Figueiredo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (I.F.d.S.F.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Elizabeth Soares Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (I.F.d.S.F.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Cristina de Andrade Monteiro
- Departamento de Biologia, Instituto Federal do Maranhão, Av. Getúlio Vargas nº 2158/2159, São Luís 65080-805, MA, Brazil;
| | - Valério Monteiro-Neto
- Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil; (A.G.G.M.); (C.D.L.C.); (J.L.P.-F.); (A.P.A.P.); (G.S.A.R.); (Á.W.d.M.S.A.); (P.d.M.M.); (F.C.B.V.); (S.G.M.)
| |
Collapse
|
26
|
Liao Y, Gao IH, Kusakabe T, Lin WY, Grier A, Pan X, Morzhanaeva O, Shea TP, Yano H, Karo-Atar D, Olsen KA, Oh JH, Vandegrift KJ, King IL, Cuomo CA, Artis D, Rehermann B, Lipman N, Iliev ID. Fungal symbiont transmitted by free-living mice promotes type 2 immunity. Nature 2024; 636:697-704. [PMID: 39604728 PMCID: PMC11733984 DOI: 10.1038/s41586-024-08213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/14/2024] [Indexed: 11/29/2024]
Abstract
The gut mycobiota is crucial for intestinal homeostasis and immune function1. Yet its variability and inconsistent fungal colonization of laboratory mice hinders the study of the evolutionary and immune processes that underpin commensalism2,3. Here, we show that Kazachstania pintolopesii is a fungal commensal in wild urban and rural mice, with an exceptional ability to colonize the mouse gastrointestinal tract and dominate the gut mycobiome. Kazachstania pintolopesii colonization occurs in a bacteria-independent manner, results in enhanced colonization resistance to other fungi and is shielded from host immune surveillance, allowing commensal presence. Following changes in the mucosal environment, K. pintolopesii colonization triggers a type 2 immune response in mice and induces gastrointestinal eosinophilia. Mechanistically, we determined that K. pintolopesii activates type 2 immunity via the induction of epithelial IL-33 and downstream IL-33-ST2 signalling during mucus fluctuations. Kazachstania pintolopesii-induced type 2 immunity enhanced resistance to helminth infections or aggravated gastrointestinal allergy in a context-dependent manner. Our findings indicate that K. pintolopesii is a mouse commensal and serves as a valuable model organism for studying gut fungal commensalism and immunity in its native host. Its unnoticed presence in mouse facilities highlights the need to evaluate its influence on experimental outcomes and phenotypes.
Collapse
Affiliation(s)
- Yun Liao
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Iris H Gao
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Takato Kusakabe
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Woan-Yu Lin
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Alexander Grier
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Xiangyu Pan
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Olga Morzhanaeva
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Terrance P Shea
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hiroshi Yano
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Kaitlin A Olsen
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Ji Hoon Oh
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Kurt J Vandegrift
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - David Artis
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Neil Lipman
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
27
|
Xu T, Wang S, Ma T, Dong Y, Ashby CR, Hao GF. The identification of essential cellular genes is critical for validating drug targets. Drug Discov Today 2024; 29:104215. [PMID: 39428084 DOI: 10.1016/j.drudis.2024.104215] [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: 08/15/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Accurately identifying biological targets is crucial for advancing treatment options. Essential genes, vital for cell or organism survival, hold promise as potential drug targets in disease treatment. Although many studies have sought to identify essential genes as therapeutic targets in medicine and bioinformatics, systematic reviews on their relationship with drug targets are relatively rare. This work presents a comprehensive analysis to aid in identifying essential genes as potential targets for drug discovery, encompassing their relevance, identification methods, successful case studies, and challenges. This work will facilitate the identification of essential genes as therapeutic targets, thereby boosting new drug development.
Collapse
Affiliation(s)
- Ting Xu
- School of Pharmaceutical Sciences, Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang 550025, China
| | - Shuang Wang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, Guiyang 550025, China
| | - Tingting Ma
- School of Pharmaceutical Sciences, Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang 550025, China
| | - Yawen Dong
- School of Pharmaceutical Sciences, Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang 550025, China.
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, USA.
| | - Ge-Fei Hao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
28
|
Ikeda E, Yamaguchi M, Ono M, Kawabata S. In Vitro Acid Resistance of Pathogenic Candida Species in Simulated Gastric Fluid. GASTRO HEP ADVANCES 2024; 4:100591. [PMID: 39996247 PMCID: PMC11847298 DOI: 10.1016/j.gastha.2024.100591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/05/2024] [Indexed: 02/26/2025]
Abstract
Background and Aims Although species in the fungal genus Candida are often commensal residents of the gastrointestinal (GI) tract, they can also cause high-mortality systemic candidiasis. Most pathogenic Candida species are dimorphic fungi that exist predominantly in filamentous forms in the invading tissues. Candida albicans is the most prominent pathogen among Candida species, but nonalbicans Candida species have also emerged as important pathogens. The stomach is the most acidic niche in the GI tract and is maintained at pH 1-2 in healthy individuals. The aim of the present study was to determine whether Candida species can survive in gastric fluid and to observe their morphology under varied pH conditions. Methods We investigated the in vitro survival of the pathogenic Candida species C. albicans, Candida glabrata, Candida parapsilosis, and Candida tropicalis in simulated gastric fluid. Results We first described that a portion of the 4 Candida species can survive under highly acidic conditions. Moreover, dimorphic Candida species, namely, C. albicans, C. parapsilosis, and C. tropicalis, exhibited yeast-hyphal transition in simulated gastric fluid with elevated pH. Pathogenic filamentous cells had lower acid resistance than yeast cells. Conclusion These findings may illuminate the migration to the lower GI tract by commensal fungi of the oral cavity.
Collapse
Affiliation(s)
- Eri Ikeda
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan
- Bioinformatics Research Unit, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masayuki Ono
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Bioinformatics Research Unit, Graduates School of Dentistry, Osaka University, Osaka, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan
| |
Collapse
|
29
|
Prakash A, Holla SR, Selvaraj S, Nayak R, De S, Saquib M, Selvakumar M. Highly conducting Laser-Induced Graphene-Ag nanoparticle composite as an effective supercapacitor electrode with anti-fungal properties. Sci Rep 2024; 14:27849. [PMID: 39537725 PMCID: PMC11561331 DOI: 10.1038/s41598-024-79382-3] [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: 02/28/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
This study presents a simple and an environmentally friendly approach to make Laser-Induced Graphene (LIG) based supercapacitor electrodes anchored with abundant Silver Nanoparticles (AgNPs). LIG, was synthesized using a CO2 laser writing technique on polyimide substrate. The LIG-Ag composite was prepared using two techniques, drop-coating, and screen-printing. Ag nanoparticles prepared using the plant extract of Swietenia Macrophylla was utilized to drop-coat AgNP on LIG substrate. Screen-printing was done by using a commercial Ag-ink and a suitable mesh. The supercapacitor made from screen-printed electrodes and supercapacitor made form drop-coated electrodes showed a high specific capacitance of 118 mF/cm2, 38 mF/cm2, and a high energy density of 2.42 mWh/cm2, 0.05 mWh/cm2 respectively. The screen-printed composite of LIG and AgNP was further studied for its anti-fungal properties and proved to be effective against Candida sp.
Collapse
Affiliation(s)
- Abhishek Prakash
- Department of Electronics & Communication Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Sowmya R Holla
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India.
| | - Subbalaxmi Selvaraj
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India.
| | - Ramakrishna Nayak
- Department of Humanities and Management, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shounak De
- Department of Electronics & Communication Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Mohammad Saquib
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - M Selvakumar
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| |
Collapse
|
30
|
Alonso MF, Bain JM, Erwig LP, Brown AJP, Gow NAR. Hyphal swelling induced in the phagosome of macrophages. Fungal Biol 2024; 128:2148-2156. [PMID: 39384284 PMCID: PMC11482207 DOI: 10.1016/j.funbio.2024.08.011] [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: 04/22/2024] [Revised: 08/09/2024] [Accepted: 08/28/2024] [Indexed: 10/11/2024]
Abstract
Macrophages play critical protective roles as sentinels of the innate immune system against fungal infection. It is therefore important to understand the dynamics of the interaction between these phagocytes and their fungal prey. We show here that many of the hyphal apices formed by Candida albicans within the macrophage ceased elongating, and apical and sub-apical hyphal compartments became swollen. Swollen hyphal cell compartments assimilated less Lysotracker-Red than non-swollen compartments, suggesting they had enhanced viability. Staining with florescent dyes suggested that there were higher levels of β-glucan and chitin in internalized fungal filaments compared to non-internalized hyphae, suggesting active cell wall remodelling within macrophages. These observations suggest that the stresses imposed by macrophages upon the fungus lead to changes in cell wall composition, inhibition of polarised growth and the induction of swelling in hyphal compartments, and that this can prevent or delay loss of viability of hyphal cells within the phagocyte.
Collapse
Affiliation(s)
- María Fernanda Alonso
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Judith M Bain
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Lars P Erwig
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Alistair J P Brown
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK.
| | - Neil A R Gow
- Aberdeen Fungal Group, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
31
|
Shen JS, Wang ZJ, Duan Y, Mei LN, Zhu YY, Wei MZ, Wang XH, Luo XD. Antifungal bioactivity of Sarcococca hookeriana var. digyna Franch. against fluconazole-resistant Candida albicans in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118473. [PMID: 38897554 DOI: 10.1016/j.jep.2024.118473] [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: 05/20/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sarcococca hookeriana var. digyna Franch. has been widely utilized in folk medicine by the Miao people in the southwestern region of China for treating skin sores which may be associated with microbial infection. AIM OF THE STUDY To investigate the antifungal bioactivity of S. hookeriana var. digyna against fluconazole-resistant Candida albicans in vitro and in vivo, as well as its underlying mechanism and the key bioactive component. MATERIALS AND METHODS The antifungal bioactivity of 80% ethanol extract of S. hookeriana var. digyna (SHE80) was investigated in vitro using the broth microdilution method, time-growth curve, and time-kill assay. Its key functional component and antifungal mechanism were explored with combined approaches including UPLC-Q-TOF-MS, network pharmacology and metabolomics. The antifungal pathway was further supported via microscopic observation of fungal cell morphology and examination of its effects on fungal biofilm and cell membranes using fluorescent staining reagents. In vivo assessment of antifungal bioactivity was conducted using a mouse model infected with C. albicans on the skin. RESULTS S. hookeriana var. digyna suppressed fluconazole-resistant C. albicans efficiently (MIC = 16 μg/mL, MFC = 64 μg/mL). It removed fungal biofilm, increased cell membrane permeability, induced protein leakage, reduced membrane fluidity, disrupted mitochondrial membrane potential, induced the release of reactive oxygen species, promoted cell apoptosis, and inhibited the transformation of fungi from the yeast state to the hyphal state significantly. In terms of mechanism, it affected sphingolipid metabolism and signaling pathway. Moreover, the predicted bioactive component, sarcovagine D, was supported by antifungal bioactivity evaluation in vitro (MIC = 4 μg/mL, MFC = 16 μg/mL). Furthermore, S. hookeriana var. digyna promoted wound healing, reduced the number of colony-forming units, and reduced inflammation effectively in vivo. CONCLUSIONS The traditional use of S. hookeriana var. digyna for fungal skin infections was supported by antifungal bioactivity investigated in vitro and in vivo. Its mechanism and bioactive component were predicted and confirmed by experiments, which also provided a new antifungal agent for future research.
Collapse
Affiliation(s)
- Jia-Shan Shen
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yu Duan
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Li-Na Mei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Mei-Zheng Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Xin-Hui Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| |
Collapse
|
32
|
Kashyap B, Padala SR, Kaur G, Kullaa A. Candida albicans Induces Oral Microbial Dysbiosis and Promotes Oral Diseases. Microorganisms 2024; 12:2138. [PMID: 39597528 PMCID: PMC11596246 DOI: 10.3390/microorganisms12112138] [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: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Candida albicans are ubiquitous fungal organisms that colonize the oral cavity of healthy individuals without causing disease. C. albicans is an opportunistic microorganism with several virulent factors that influence the inflammatory process and allow it to invade tissues, evade host defense mechanisms, and release toxins, facilitating proliferation and degradation. At present, increasing emphasis is placed on polymicrobial interactions between C. albicans and various bacterial pathogens. Such interaction is mutually beneficial for both parties: it is competitive and antagonistic. Their complex interaction and colonization in the oral cavity serve as the basis for several oral diseases. The dispersion of C. albicans in saliva and the systemic circulation is noted in association with other bacterial populations, suggesting their virulence in causing disease. Hence, it is necessary to understand fungal-bacterial interactions for early detection and the development of novel therapeutic strategies to treat oral diseases. In this paper, we review the mutualistic interaction of C. albicans in oral biofilm formation and polymicrobial interactions in oral diseases. In addition, C. albicans virulence in causing biofilm-related oral diseases and its presence in saliva are discussed.
Collapse
Affiliation(s)
- Bina Kashyap
- Institute of Dentistry, University of Eastern Finland, 70211 Kuopio, Finland;
| | | | - Gaganjot Kaur
- Shaheed Kartar Singh Sarabha Dental College & Hospital, Ludhiana 141105, India;
| | - Arja Kullaa
- Institute of Dentistry, University of Eastern Finland, 70211 Kuopio, Finland;
| |
Collapse
|
33
|
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
|
34
|
Vandermeulen MD, Khaiwal S, Rubio G, Liti G, Cullen PJ. Gain- and loss-of-function alleles within signaling pathways lead to phenotypic diversity among individuals. iScience 2024; 27:110860. [PMID: 39381740 PMCID: PMC11460476 DOI: 10.1016/j.isci.2024.110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/29/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024] Open
Abstract
Understanding how phenotypic diversity is generated is an important question in biology. We explored phenotypic diversity among wild yeast isolates (Saccharomyces cerevisiae) and found variation in the activity of MAPK signaling pathways as a contributing mechanism. To uncover the genetic basis of this mechanism, we identified 1957 SNPs in 62 candidate genes encoding signaling proteins from a MAPK signaling module within a large collection of yeast (>1500 individuals). Follow-up testing identified functionally relevant variants in key signaling proteins. Loss-of-function (LOF) alleles in a PAK kinase impacted protein stability and pathway specificity decreasing filamentous growth and mating phenotypes. In contrast, gain-of-function (GOF) alleles in G-proteins that were hyperactivating induced filamentous growth. Similar amino acid substitutions in G-proteins were identified in metazoans that in some cases were fixed in multicellular lineages including humans, suggesting hyperactivating GOF alleles may play roles in generating phenotypic diversity across eukaryotes. A mucin signaler that regulates MAPK activity was also found to contain a prevalance of presumed GOF alleles amoung individuals based on changes in mucin repeat numbers. Thus, genetic variation in signaling pathways may act as a reservoir for generating phenotypic diversity across eukaryotes.
Collapse
Affiliation(s)
| | - Sakshi Khaiwal
- Université Côte d’Azur, CNRS, INSERM, IRCAN, Nice, France
| | - Gabriel Rubio
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA
| | - Gianni Liti
- Université Côte d’Azur, CNRS, INSERM, IRCAN, Nice, France
| | - Paul J. Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA
| |
Collapse
|
35
|
Li H, Shi Y, Chen H, Liang J, Zhang S, Li B, Chen J, Li M, Peng X, Zhou X, Ren B, Cheng L. A novel pH-responsive monomer inhibits Candida albicans via a dual antifungal mode of action. J Mater Chem B 2024; 12:10367-10382. [PMID: 39290132 DOI: 10.1039/d4tb00851k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The scarcity of the antifungal drug arsenal highlights an urgent need to develop alternative treatments for candidiasis caused by Candida albicans (C. albicans). As pH is closely associated with C. albicans infection, it could be an essential target in a novel approach for designing antifungal therapy. In this study, a novel intelligent antifungal monomer, dodecylmethylaminoethyl methacrylate (DMAEM), with a pH-responsive tertiary amine group and a methacrylate-derived CC double bond group is developed. It is uncovered that the two functional groups of DMAEM contribute to a dual mode of action. Under acidic pH, the tertiary amine of DMAEM protonates into a cationic fungicide, sharing similar structural and functional characteristics with quaternary ammonium salts, which exerts fungicidal activity by targeting the CHK1 two-component system in C. albicans. At neutral pH, the methacrylate-derived CC double bond group contributes to anti-virulence activity by blocking hyphal formation. In addition, it is also identified that DMAEM suppresses filamentation by altering the extracellular vesicles of C. albicans. These findings support that the novel intelligent pH-responsive monomer could be a therapeutic candidate for treating candidiasis.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hui Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jingou Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shiyong Zhang
- National Engineering Research Center for Biomaterials, and College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Bolei Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jing Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Xian Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China.
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
36
|
Vandermeulen MD, Lorenz MC, Cullen PJ. Conserved signaling modules regulate filamentous growth in fungi: a model for eukaryotic cell differentiation. Genetics 2024; 228:iyae122. [PMID: 39239926 PMCID: PMC11457945 DOI: 10.1093/genetics/iyae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/20/2024] [Indexed: 09/07/2024] Open
Abstract
Eukaryotic organisms are composed of different cell types with defined shapes and functions. Specific cell types are produced by the process of cell differentiation, which is regulated by signal transduction pathways. Signaling pathways regulate cell differentiation by sensing cues and controlling the expression of target genes whose products generate cell types with specific attributes. In studying how cells differentiate, fungi have proved valuable models because of their ease of genetic manipulation and striking cell morphologies. Many fungal species undergo filamentous growth-a specialized growth pattern where cells produce elongated tube-like projections. Filamentous growth promotes expansion into new environments, including invasion into plant and animal hosts by fungal pathogens. The same signaling pathways that regulate filamentous growth in fungi also control cell differentiation throughout eukaryotes and include highly conserved mitogen-activated protein kinase (MAPK) pathways, which is the focus of this review. In many fungal species, mucin-type sensors regulate MAPK pathways to control filamentous growth in response to diverse stimuli. Once activated, MAPK pathways reorganize cell polarity, induce changes in cell adhesion, and promote the secretion of degradative enzymes that mediate access to new environments. However, MAPK pathway regulation is complicated because related pathways can share components with each other yet induce unique responses (i.e. signal specificity). In addition, MAPK pathways function in highly integrated networks with other regulatory pathways (i.e. signal integration). Here, we discuss signal specificity and integration in several yeast models (mainly Saccharomyces cerevisiae and Candida albicans) by focusing on the filamentation MAPK pathway. Because of the strong evolutionary ties between species, a deeper understanding of the regulation of filamentous growth in established models and increasingly diverse fungal species can reveal fundamentally new mechanisms underlying eukaryotic cell differentiation.
Collapse
Affiliation(s)
| | - Michael C Lorenz
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Paul J Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA
| |
Collapse
|
37
|
Aguilar-Morales CM, Alejandre-Castañeda V, Contreras-Celedón C, Ramírez-Díaz MI, Islas-Jácome A, Meza-Carmen V, Chacón-García L, Cortés-García CJ. A one-pot five component reaction for the synthesis of tetrazol-benzofuran hybrids and their inhibitory activity against Mucor lusitanicus. Org Biomol Chem 2024; 22:7240-7244. [PMID: 39171544 DOI: 10.1039/d4ob00995a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
A synthetic strategy for obtaining a new series of 1,5-disubstituted tetrazole-benzofuran hybrid systems via a one-pot five-component reaction is described. This process involves a Ugi-azide multicomponent reaction coupled to an intramolecular cyclization catalyzed by Pd/Cu, resulting in low to moderate yields from 21 to 67%. This protocol allowed the synthesis of highly substituted benzofurans at the 2-position through an operationally simple process under mild reaction conditions and with high bond forming efficiency due to the formation of six new bonds (two C-C, two C-N, one N-N, and one C-O). Besides, to evaluate the antifungal activity of 1,5-disubstituted tetrazole-benzofurans 9a-n, in vitro studies against Mucor lusitanicus were performed, finding that compound 9b exhibits bioactivity comparable to the commercial antifungal drug Amphotericin B. These results suggest potential for use in controlling mucormycosis infections in animal models, highlighting the importance of these findings given the limited antifungal drug options and high mortality rates associated with this infection.
Collapse
Affiliation(s)
- Cesia M Aguilar-Morales
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58030, Morelia, Michoacán, Mexico.
| | - Viridiana Alejandre-Castañeda
- Laboratorio de Diferenciación Celular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ed. B-1, Ciudad Universitaria, Morelia, Michoacán 58030, Mexico
| | - Claudia Contreras-Celedón
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58030, Morelia, Michoacán, Mexico.
| | - Martha Isela Ramírez-Díaz
- Laboratorio de Diferenciación Celular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ed. B-1, Ciudad Universitaria, Morelia, Michoacán 58030, Mexico
| | - Alejandro Islas-Jácome
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Col. Leyes de Reforma 1A Sección, Iztapalapa, Ciudad de México, 09310, Mexico
| | - Victor Meza-Carmen
- Laboratorio de Diferenciación Celular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ed. B-1, Ciudad Universitaria, Morelia, Michoacán 58030, Mexico
| | - Luis Chacón-García
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58030, Morelia, Michoacán, Mexico.
| | - Carlos J Cortés-García
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58030, Morelia, Michoacán, Mexico.
| |
Collapse
|
38
|
Hao Y, Wang R, Ni T, Monk BC, Tyndall JDA, Bao J, Wang M, Chi X, Yu S, Jin Y, Zhang D, Yan L, Xie F. Synthesis and antifungal evaluation of novel triazole derivatives bearing a pyrazole-methoxyl moiety. Eur J Med Chem 2024; 275:116637. [PMID: 38959728 DOI: 10.1016/j.ejmech.2024.116637] [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: 05/15/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Life-threatening invasive fungal infections pose a serious threat to human health. A series of novel triazole derivatives bearing a pyrazole-methoxyl moiety were designed and synthesized in an effort to obtain antifungals with potent, broad-spectrum activity that are less susceptible to resistance. Most of these compounds exhibited moderate to excellent in vitro antifungal activities against Candida albicans SC5314 and 10,231, Cryptococcus neoformans 32,609, Candida glabrata 537 and Candida parapsilosis 22,019 with minimum inhibitory concentration (MIC) values of ≤0.125 μg/mL to 0.5 μg/mL. Use of recombinant Saccharomyces cerevisiae strains showed compounds 7 and 10 overcame the overexpression and resistant-related mutations in ERG11 of S. cerevisae and several pathogenic Candida spp. Despite being substrates of the C. albicans and Candida auris Cdr1 drug efflux pumps, compounds 7 and 10 showed moderate potency against five fluconazole (FCZ)-resistant fungi with MIC values from 2.0 μg/mL to 16.0 μg/mL. Growth kinetics confirmed compounds 7 and 10 had much stronger fungistatic activity than FCZ. For C. albicans, compounds 7 and 10 inhibited the yeast-to-hyphae transition, biofilm formation and destroyed mature biofilm more effectively than FCZ. Preliminary mechanism of action studies showed compounds 7 and 10 blocked the ergosterol biosynthesis pathway at Erg11, ultimately leading to cell membrane disruption. Further investigation of these novel triazole derivatives is also warranted by their predicted ADMET properties and low cytotoxicity.
Collapse
Affiliation(s)
- Yumeng Hao
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China
| | - Ruina Wang
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No.1239 Siping Road, Shanghai, 200072, China
| | - Brian C Monk
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, 9016, New Zealand
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand
| | - Junhe Bao
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China
| | - Mengyuan Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenyang, 110016, China
| | - Xiaochen Chi
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenyang, 110016, China
| | - Shichong Yu
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China
| | - Yongsheng Jin
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China
| | - Dazhi Zhang
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China; Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No.1239 Siping Road, Shanghai, 200072, China.
| | - Lan Yan
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China.
| | - Fei Xie
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
39
|
Sarkar R, Adhikary K, Banerjee A, Ganguly K, Sarkar R, Mohanty S, Dhua R, Bhattacharya K, Ahuja D, Pal S, Maiti R. Novel targets and improved immunotherapeutic techniques with an emphasis on antimycosal drug resistance for the treatment and management of mycosis. Heliyon 2024; 10:e35835. [PMID: 39224344 PMCID: PMC11367498 DOI: 10.1016/j.heliyon.2024.e35835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Infections due to pathogenic fungi are endemic in particular area with increased morbidity and mortality. More than a thousand people are infected per year and the way of treatment is of high demand having a significant impact on the population health. Medical practitioners confront various troublesome analytic and therapeutical challenges in the administration of immunosuppressed sufferer at high danger of expanding fungal infections. An upgraded antimycosal treatment is fundamental for a fruitful result while treating intrusive mycoses. A collection of antimycosal drugs keeps on developing with their specific antifungal targets including cell membrane, mitochondria, cell wall, and deoxyribonucleic acid (DNA)/ribonucleic acid (RNA) or protein biosynthesis. Some fundamental classes of ordinarily directed medications are the polyenes, amphotericin B, syringomycin, allylamines, honokiol, azoles, flucytosine, echinocandins etc. However, few immunotherapy processes and vaccinations are being developed to mark this need, although one presently can't seem to arrive at the conclusion. In this review article, there has been a trial to give details upgradation about the current immune therapeutic techniques and vaccination strategies against prevention or treatment of mycosis as well as the difficulties related with their turn of events. There has been also a visualization in the mentioned review paper about the various assorted drugs and their specific target analysis along with therapeutic interventions.
Collapse
Affiliation(s)
- Riya Sarkar
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Adhikary
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Arundhati Banerjee
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Ganguly
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Riya Sarkar
- Department of Medical Laboratory Technology, Dr. B. C. Roy Academy of Professional Courses, Durgapur, West Bengal, 713206, India
| | - Satyajit Mohanty
- Department of Advanced Pharmacology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rumpa Dhua
- Department of Nutrition, Bankura Sammilani College, Kenduadihi, Bankura, West Bengal, 722102, India
| | - Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Deepika Ahuja
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Suchandra Pal
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal, 713209, India
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, 722101, India
| |
Collapse
|
40
|
Cao X, Xiao N, Huang J, Li L, Zhong L, Zhang J, Wang F. Synergistic in vitro activity and mechanism of KBN lotion and miconazole nitrate against drug-resistant Candida albicans biofilms. Front Cell Infect Microbiol 2024; 14:1426791. [PMID: 39268490 PMCID: PMC11390680 DOI: 10.3389/fcimb.2024.1426791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/31/2024] [Indexed: 09/15/2024] Open
Abstract
Background In the face of increasing antifungal resistance among Candida albicans biofilms, this study explores the efficacy of a combined treatment using Kangbainian lotion (KBN) and miconazole nitrate (MN) to address this challenge. Methods Using UPLC-Q-TOF/MS Analysis for Identification of Active Compounds in KBN Lotion; FICI for synergy evaluation, XTT and ROS assays for biofilm viability and oxidative stress, fluorescence and confocal laser scanning microscopy (CLSM) for structural and viability analysis, and real-time fluorescence for gene expression. Conclusion Our study indicates that the combined application of KBN and MN somewhat impacts the structural integrity of Candida albicans biofilms and affects the expression of several key genes involved in biofilm formation, including ALS1, ALS3, HWP1, HSP90, and CSH1. These preliminary findings suggest that there may be a synergistic effect between KBN and MN, potentially influencing not only the structural aspects of fungal biofilms but also involving the modulation of genetic pathways during their formation.
Collapse
Affiliation(s)
- Xiaoyu Cao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ni Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingyi Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lian Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jun Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengyun Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
41
|
Williams C, Carnahan BR, Hyland SN, Grimes CL. Bioorthogonal labeling of chitin in pathogenic Candida species reveals biochemical mechanisms of hyphal growth and homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609898. [PMID: 39253419 PMCID: PMC11383299 DOI: 10.1101/2024.08.27.609898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Pathogenic fungi rely on the cell wall component, chitin, for critical structural and immunological functions. Here a chitin labeling method to visualize the hyphal pathogenic response was developed. The data show that filamentous fungi, Candida albicans , transport N -acetylglucosamine (NAG) bio-orthogonal probes and incorporate them into the cell wall, indicating the probes utility for in vivo study of the morphological, pathogenic switch. As yeast reside in complex microenvironments, The data show that the opportunistic microbe C. albicans , has developed processes to utilize surrounding bacterial cell wall fragments to initiate the morphogenic switch. The probes are utilized for visualization of growth patterns of pathogenic fungi, providing insights into novel mechanisms for the development of antifungals. Remodeling chitin in fungi using NAG derivatives will advance yeast pathogenic studies.
Collapse
|
42
|
Calvi GDS, Cartaxo GNJ, Carretoni QL, da Silva ALM, de Moraes DN, Pradella JGDC, Costa MS. Inhibition of Development and Metabolism of Dual-Species Biofilms of Candida albicans and Candida krusei ( Pichia kudriavzevii) by Organoselenium Compounds. Pharmaceuticals (Basel) 2024; 17:1078. [PMID: 39204183 PMCID: PMC11359205 DOI: 10.3390/ph17081078] [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: 06/28/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Although Candida albicans is the most frequently identified Candida species in clinical settings, a significant number of infections related to the non-albicans Candida (NAC) species, Candida krusei, has been reported. Both species are able to produce biofilms and have been an important resistance-related factor to antimicrobial resistance. In addition, the microbial relationship is common in the human body, contributing to the formation of polymicrobial biofilms. Considering the great number of reports showing the increase in cases of resistance to the available antifungal drugs, the development of new and effective antifungal agents is critical. The inhibitory effect of Organoselenium Compounds (OCs) on the development of Candida albicans and Candida krusei was recently demonstrated, supporting the potential of these compounds as efficient antifungal drugs. In addition, OCs were able to reduce the viability and the development of biofilms, a very important step in colonization and infection caused by fungi. Thus, the objective of this study was to investigate the effect of the Organoselenium Compounds (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2 on the development of dual-species biofilms of Candida albicans and Candida krusei produced using either RPMI-1640 or Sabouraud Dextrose Broth (SDB) media. The development of dual-species biofilms was evaluated by the determination of both metabolic activity, using a metabolic assay based on the reduction of XTT (2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide sodium salt) assay and identification of either Candida albicans and Candida krusei on CHROMagar Candida medium. Biofilm formation using RPMI-1640 was inhibited in 90, 55, and 20% by 30 µM (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2, respectively. However, biofilms produced using SDB presented an inhibition of 62, 30 and 15% in the presence of 30 µM (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2, respectively. The metabolic activity of 24 h biofilms was inhibited by 35, 30 and 20% by 30 µM (p-MeOPhSe)2, (PhSe)2, and (p-Cl-PhSe)2, respectively, with RPMI-1640; however, 24 h biofilms formed using SDB were not modified by the OCs. In addition, a great reduction in the number of CFUs of Candida albicans (93%) in biofilms produced using RPMI-1640 in the presence of 30 µM (p-MeOPhSe)2 was observed. However, biofilms formed using SDB and treated with 30 µM (p-MeOPhSe)2 presented a reduction of 97 and 69% in the number of CFUs of Candida albicans and Candida krusei, respectively. These results demonstrated that Organoselenium Compounds, mainly (p-MeOPhSe)2, are able to decrease the metabolic activity of dual-species biofilms by reducing both Candida albicans and Candida krusei cell number during biofilm formation using either RPMI-1640 or SDB. Taken together, these results demonstrated the potential of the OCs to inhibit the development of dual-species biofilms of Candida albicans and Candida krusei.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maricilia Silva Costa
- Instituto de Pesquisa & Desenvolvimento—IP&D, Universidade do Vale do Paraíba—UNIVAP, Av. Shishima Hifumi, 2911, São José dos Campos 12244-390, SP, Brazil; (G.d.S.C.); (G.N.J.C.); (Q.L.C.); (A.L.M.d.S.); (D.N.d.M.); (J.G.d.C.P.)
| |
Collapse
|
43
|
Spaggiari L, Ardizzoni A, Pedretti N, Iseppi R, Sabia C, Russo R, Kenno S, De Seta F, Pericolini E. Bacillus coagulans LMG S-24828 Impairs Candida Virulence and Protects Vaginal Epithelial Cells against Candida Infection In Vitro. Microorganisms 2024; 12:1634. [PMID: 39203476 PMCID: PMC11356316 DOI: 10.3390/microorganisms12081634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Probiotics are living microbes that provide benefits to the host. The growing data on health promotion, following probiotics administration, increased interest among researchers and pharmaceutical companies. Infections of the lower genital tract in females, caused by a wide range of pathogens, represent one of the main areas for the use of probiotics and postbiotics. Vulvovaginal candidiasis (VVC) affects 75% of women of reproductive age at least once during their lifetime, with 5-8% developing the recurrent form (RVVC). The disease is triggered by the overgrowth of Candida on the vaginal mucosa. Here, in order to establish its probiotic potential in the context of VVC, we evaluated the anti-fungal effects of the spore-producing Bacillus coagulans LMG S-24828 against C. albicans and C. parapsilosis as well as its beneficial effects in counteracting Candida vaginal infection in vitro. Our results show that both live B. coagulans and its Cell-Free Supernatant (CFS) exerted antifungal activity against both fungi. Moreover, live B. coagulans reduced hyphal formation, inhibited C. albicans adhesion to vaginal epithelial cells, showed co-aggregation capacity, and exerted a protective effect on vaginal epithelial cells infected with C. albicans. These data suggest that B. coagulans LMG S-24828 may provide benefits in the context of Candida vaginal infections.
Collapse
Affiliation(s)
- Luca Spaggiari
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Andrea Ardizzoni
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (A.A.); (S.K.)
| | - Natalia Pedretti
- Department of Medical Sciences, University of Trieste, 34129 Trieste, Italy;
| | - Ramona Iseppi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.I.); (C.S.)
| | - Carla Sabia
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.I.); (C.S.)
| | - Rosario Russo
- Giellepi S.p.A., Via G. Verdi, 41/Q, 20831 Seregno, Italy;
| | - Samyr Kenno
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (A.A.); (S.K.)
| | - Francesco De Seta
- Department of Obstetrics and Gynecology, IRCCS San Raffaele Scientific Institute, University Vita and Salute, 20132 Milan, Italy;
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (A.A.); (S.K.)
| |
Collapse
|
44
|
Thomas G, Kay WT, Fones HN. Life on a leaf: the epiphyte to pathogen continuum and interplay in the phyllosphere. BMC Biol 2024; 22:168. [PMID: 39113027 PMCID: PMC11304629 DOI: 10.1186/s12915-024-01967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/01/2024] [Indexed: 08/11/2024] Open
Abstract
Epiphytic microbes are those that live for some or all of their life cycle on the surface of plant leaves. Leaf surfaces are a topologically complex, physicochemically heterogeneous habitat that is home to extensive, mixed communities of resident and transient inhabitants from all three domains of life. In this review, we discuss the origins of leaf surface microbes and how different biotic and abiotic factors shape their communities. We discuss the leaf surface as a habitat and microbial adaptations which allow some species to thrive there, with particular emphasis on microbes that occupy the continuum between epiphytic specialists and phytopathogens, groups which have considerable overlap in terms of adapting to the leaf surface and between which a single virulence determinant can move a microbial strain. Finally, we discuss the recent findings that the wheat pathogenic fungus Zymoseptoria tritici spends a considerable amount of time on the leaf surface, and ask what insights other epiphytic organisms might provide into this pathogen, as well as how Z. tritici might serve as a model system for investigating plant-microbe-microbe interactions on the leaf surface.
Collapse
Affiliation(s)
| | - William T Kay
- Department of Plant Sciences, University of Oxford, Oxford, UK
| | | |
Collapse
|
45
|
Li J, Zhao Z, You D, Xie Y, Feng Y, Li X, Cui Z, Fuai L. Hemiprotonic ph-ph + with two targets inhibits metastatic breast cancer and concurrent candidiasis. Biochem Pharmacol 2024; 226:116394. [PMID: 38942090 DOI: 10.1016/j.bcp.2024.116394] [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: 03/26/2024] [Revised: 05/22/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Concurrent infection in breast cancer patients is the direct cause of the high mortality rate of the disease. However, there is no available method to increase the survival rate until now. To address the problem, we propose one drug with two target strategy to treat the refractory disease. A small chemical, ph-ph+, was attempted to be used in the study to explore the feasibility of the approach in anticancer and antifungus at the same time. The results showed that ph-ph+ could prevent the proliferation and metastasis of breast cancer cells, and kill C. albicans simultaneously. The molecular mechanism was associated with the activation of an evolutionarily conserved protease CLpP in the cancer and C. albicans cells. Also, the signaling pathway mediated by PLAGL2 that highly expressed in cancer cells participated in preventing cell metastasis and inducing apoptosis of ph-ph+. The one drug with dual targets inhibited the growth and metastasis of the cancer cells, and meanwhile eliminated C. albicans in tissues in the experimental animals. The results suggested that ph-ph+ with dual targets of CLpP and PLAGL2 would be a feasible approach to prolong the survival rate in patients with metastatic breast cancer and pathogenic infection.
Collapse
Affiliation(s)
- Jingli Li
- School of Pharmaceutical Sciences, Southwest University, China
| | - Zizhen Zhao
- School of Pharmaceutical Sciences, Southwest University, China
| | - Dongmei You
- School of Pharmaceutical Sciences, Southwest University, China
| | - Yafang Xie
- School of Pharmaceutical Sciences, Southwest University, China
| | - Yixiao Feng
- School of Pharmaceutical Sciences, Southwest University, China
| | - Xiaorong Li
- School of Pharmaceutical Sciences, Southwest University, China
| | - Zhihong Cui
- School of Pharmaceutical Sciences, Southwest University, China.
| | - Ling Fuai
- School of Pharmaceutical Sciences, Southwest University, China.
| |
Collapse
|
46
|
Soares AB, de Albuquerque MC, Rosa LM, Klein MI, Pavarina AC, Barbugli PA, Dovigo LN, Mima EGDO. Quantification methods of Candida albicans are independent irrespective of fungal morphology. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:265-277. [PMID: 39081907 PMCID: PMC11287054 DOI: 10.15698/mic2024.07.831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 08/02/2024]
Abstract
The ability of Candida albicans to switch its morphology from yeast to filaments, known as polymorphism, may bias the methods used in microbial quantification. Here, we compared the quantification methods [cell/mL, colony forming units (CFU)/mL, and the number of nuclei estimated by viability polymerase chain reaction (vPCR)] of three strains of C. albicans (one reference strain and two clinical isolates) grown as yeast, filaments, and biofilms. Metabolic activity (XTT assay) was also used for biofilms. Comparisons between the methods were evaluated by agreement analyses [Intraclass and Concordance Correlation Coefficients (ICC and CCC, respectively) and Bland-Altman Plot] and Pearson Correlation (α = 0.05). Principal Component Analysis (PCA) was employed to visualize the similarities and differences between the methods. Results demonstrated a lack of agreement between all methods irrespective of fungal morphology/growth, even when a strong correlation was observed. Bland-Altman plot also demonstrated proportional bias between all methods for all morphologies/growth, except between CFU/mL X vPCR for yeasts and biofilms. For all morphologies, the correlation between the methods were strong, but without linear relationship between them, except for yeast where vPCR showed weak correlation with cells/mL and CFU/mL. XTT moderately correlated with CFU/mL and vPCR and weakly correlated with cells/mL. For all morphologies/growth, PCA showed that CFU/mL was similar to cells/mL and vPCR was distinct from them, but for biofilms vPCR became more similar to CFU/mL and cells/mL while XTT was the most distinct method. As conclusions, our investigation demonstrated that CFU/mL underestimated cells/mL, while vPCR overestimated both cells/mL and CFU/mL, and that the methods had poor agreement and lack of linear relationship, irrespective of C. albicans morphology/growth.1.
Collapse
Affiliation(s)
- Amanda B Soares
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| | - Maria C de Albuquerque
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| | - Leticia M Rosa
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| | - Marlise I Klein
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP)Piracicaba, São PauloBrazil
| | - Ana C Pavarina
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| | - Paula A Barbugli
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| | - Livia N Dovigo
- Department of Social Dentistry, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| | - Ewerton G de O Mima
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP)Araraquara, Araraquara, São PauloBrazil
| |
Collapse
|
47
|
Zhou X, Chen X, Pan Q, Wang S, Li J, Yang Y. Exploring the role of candidalysin in the pathogenicity of Candida albicans by gene set enrichment analysis and evolutionary dynamics. Am J Transl Res 2024; 16:3191-3210. [PMID: 39114682 PMCID: PMC11301511 DOI: 10.62347/izym9087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/20/2024] [Indexed: 08/10/2024]
Abstract
AIMS To explore the pathogenic mechanisms of Candida albicans (C. albicans), focusing on its impact on human health, particularly through invasive infections in the gastrointestinal and respiratory tracts. METHODS In this study, we evaluated the demographic and clinical profiles of 7 pneumonia patients. Meanwhile, we used Gene Set Enrichment Analysis (GSEA) and Evolutionary Dynamics method to analyze the role of candidalysin in C. albicans pathogenicity. RESULTS By analyzing genomic data and conducting biomedical text mining, we identified novel mutation sites in the candidalysin coding gene ECE1-III, shedding light into the genetic diversity within C. albicans strains and their potential implications for antifungal resistance. Our results revealed significant associations between C. albicans and respiratory as well as gastrointestinal diseases, emphasizing the fungus's role in the pathogenesis of these diseases. Additionally, we identified a new mutation site in the C. albicans strain YF2-5, isolated from patients with pneumonia. This mutation may be associated with its heightened pathogenicity. CONCLUSION Our research advances the understanding of C. albicans pathogenicity and opens new avenues for developing targeted antifungal therapies. By focusing on the molecular basis of fungal virulence, we aim to contribute to the development of more effective treatment strategies, addressing the challenge of multidrug resistance in invasive fungal infections.
Collapse
Affiliation(s)
- Xingchen Zhou
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious DiseasesBeijing 100850, China
| | - Xiaolin Chen
- Sir Run Run Hospital, Nanjing Medical UniversityNanjing 210009, Jiangsu, China
| | - Qianglong Pan
- Sir Run Run Hospital, Nanjing Medical UniversityNanjing 210009, Jiangsu, China
| | - Shengqi Wang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious DiseasesBeijing 100850, China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical UniversityNanjing 210009, Jiangsu, China
| | - Ying Yang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious DiseasesBeijing 100850, China
| |
Collapse
|
48
|
Komath SS. To each its own: Mechanisms of cross-talk between GPI biosynthesis and cAMP-PKA signaling in Candida albicans versus Saccharomyces cerevisiae. J Biol Chem 2024; 300:107444. [PMID: 38838772 PMCID: PMC11294708 DOI: 10.1016/j.jbc.2024.107444] [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: 03/19/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can switch between yeast and hyphal morphologies depending on the environmental cues it receives. The switch to hyphal form is crucial for the establishment of invasive infections. The hyphal form is also characterized by the cell surface expression of hyphae-specific proteins, many of which are GPI-anchored and important determinants of its virulence. The coordination between hyphal morphogenesis and the expression of GPI-anchored proteins is made possible by an interesting cross-talk between GPI biosynthesis and the cAMP-PKA signaling cascade in the fungus; a parallel interaction is not found in its human host. On the other hand, in the nonpathogenic yeast, Saccharomyces cerevisiae, GPI biosynthesis is shut down when filamentation is activated and vice versa. This too is achieved by a cross-talk between GPI biosynthesis and cAMP-PKA signaling. How are diametrically opposite effects obtained from the cross-talk between two reasonably well-conserved pathways present ubiquitously across eukarya? This Review attempts to provide a model to explain these differences. In order to do so, it first provides an overview of the two pathways for the interested reader, highlighting the similarities and differences that are observed in C. albicans versus the well-studied S. cerevisiae model, before going on to explain how the different mechanisms of regulation are effected. While commonalities enable the development of generalized theories, it is hoped that a more nuanced approach, that takes into consideration species-specific differences, will enable organism-specific understanding of these processes and contribute to the development of targeted therapies.
Collapse
|
49
|
Doub JB, Siddiqui H. Propensity of Candida spp. prosthetic joint infection clinical isolates to form aggregates in synovial fluid and the clinical ramifications. Mycoses 2024; 67:e13769. [PMID: 39039764 DOI: 10.1111/myc.13769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Bacterial aggregation has been shown to occur in synovial fluid which are resistant to high concentrations of antibiotics. Yet the propensity of Candida spp. to form aggregates is unknown. OBJECTIVE To assess the ability of numerous Candida spp. to form synovial fluid aggregates and the clinical ramifications of the aggregates. METHODS Nine different Candidal prosthetic joint infection clinical isolates were evaluated for their ability to form aggregates at static and dynamic conditions and their resistance to high concentrations of amphotericin. Furthermore, the ability of tissue plasminogen activator (TPA) to disrupt the aggregates and enhance amphotericin activity was assessed. RESULTS The results show that all species of Candida spp. evaluated formed aggregates in synovial fluid under dynamic conditions that were resistant to amphotericin. Yet no aggregates formed in tryptic soy broth under any conditions or in synovial fluid under static conditions. As well, when TPA was combined with amphotericin there was a statistically significant decrease (p < .005) in the amount of colony forming units per mL for all Candidal species evaluated. Interestingly, for Candida krusei there was no colony forming units observed after exposure to TPA and amphotericin. CONCLUSION Our findings suggest that Candidal species form synovial fluid aggregates that are resistant to high dose amphotericin similar to those that occur with bacteria. However, the varying ability of the different Candida spp. to form hyphae and pseudohyphae compared to yeast cells may have direct impacts on the hardiness of the aggregates and thereby have clinical ramifications with respect to treatment durations.
Collapse
Affiliation(s)
- James B Doub
- The Doub Laboratory of Translational Bacterial Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hamda Siddiqui
- The Doub Laboratory of Translational Bacterial Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
50
|
do Socorro Costa M, da Silva ARP, Santos Araújo J, Dos Santos ATL, Fonseca VJA, Gonçalves Alencar G, Moura TF, Gonçalves SA, Filho JMB, Morais-Braga MFB, Andrade-Pinheiro JC, Coutinho HDM. In vitro Evaluation of Fungal Susceptibility and Inhibition of Virulence by Diosgenin. Chem Biodivers 2024; 21:e202400444. [PMID: 38670923 DOI: 10.1002/cbdv.202400444] [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: 02/22/2024] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 04/28/2024]
Abstract
Fungal infections are a public health problem that mainly affects immunosuppressed people, Candida spp. have been responsible for most sources of contamination and invasive fungal infections described around the world. The need arises to find new therapeutic approaches to combat growing infections. Plants and natural products have been considered a valuable source for discovering new molecules with active ingredients. Diosgenin is a sapogenin found in the families of Leguminosae and Dioscoreaceae, it is obtained mainly from the dioscin saponin through the hydrolysis method, it is a phytochemical that has been highlighted in the treatment of various diseases, as well as in combating microbial resistance. The present study aimed to evaluate the susceptibility of fungal strains to diosgenin, as well as verify the association with the reference drug and evaluate the inhibition of the virulence factor through morphological changes in the yeast state to the filamentous form of hyphae and pseudohyphae in strains of Candida albicans, Candida tropicalis and Candida krusei using the broth microdilution method and microculture technique. Antifungal assays revealed that diosgenin was not able to inhibit the growth of the tested strains. However, it was able to inhibit the fungal dimorphism of the strains evaluated, however further studies are recommended to verify its effectiveness against other virulence factors.
Collapse
Affiliation(s)
- Maria do Socorro Costa
- Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil
- Laboratory of Microbiology and Molecular Biology- LMBM, Regional University of Cariri, Crato, Ceará, Brazil
| | - Ana Raquel Pereira da Silva
- Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil
- Laboratory of Microbiology and Molecular Biology- LMBM, Regional University of Cariri, Crato, Ceará, Brazil
| | - Juliana Santos Araújo
- Laboratory of Applied Microbiology -, LAMAP, Federal University of Cariri, Barbalha, Ceará, Brazil
| | | | | | - Gabriel Gonçalves Alencar
- Laboratory of Microbiology and Molecular Biology- LMBM, Regional University of Cariri, Crato, Ceará, Brazil
| | - Talysson Felismino Moura
- Laboratory of Microbiology and Molecular Biology- LMBM, Regional University of Cariri, Crato, Ceará, Brazil
| | - Sheila Alves Gonçalves
- Laboratory of Microbiology and Molecular Biology- LMBM, Regional University of Cariri, Crato, Ceará, Brazil
| | - José Maria Barbosa Filho
- Laboratory Technology Pharmaceutical, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | | - Jacqueline Cosmo Andrade-Pinheiro
- Laboratory of Microbiology and Molecular Biology- LMBM, Regional University of Cariri, Crato, Ceará, Brazil
- Laboratory of Applied Microbiology -, LAMAP, Federal University of Cariri, Barbalha, Ceará, Brazil
| | | |
Collapse
|