1
|
Li M, Wang H, Zhao H, Jiang M, Cui M, Jia K, Lei D, Wang F. Effect of the Sho1 gene on the pathogenicity of Candida albicans and immune function in vivo. Heliyon 2024; 10:e38219. [PMID: 39397919 PMCID: PMC11467569 DOI: 10.1016/j.heliyon.2024.e38219] [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: 05/09/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Objectives Sho1, a ubiquitous membrane protein in fungi, plays a pivotal role in various physiological processes, such as osmotic stress, oxidative stress, temperature response, and virulence regulation across different fungal species. This study aimed to investigate the effect of the Sho1 gene on the pathogenicity of Candida albicans and its immune function in vivo. Materials and methods Ninety-nine clinical strains from various infection sites were collected to investigate the expression levels of the Sho1 gene compared to its levels in the standard strain (SC5314). Sho1-knockout strains (Sho1Δ/Δ) were constructed to investigate the impact of the Sho1 gene deletion on the biofilm formation, adhesion, and flocculation abilities of C. albicans. A mouse model of systemic infection was established to evaluate the impact of Sho1 deletion on survival, organ pathology, and immune cell function, as assessed by flow cytometry. Results The expression level of the Sho1 gene was found to be higher in clinical strains derived from sterile fluids, sputum, and secretions compared to that in the standard strains. Deletion of the Sho1 gene diminished the biofilm-formation capacity of C. albicans, leading to a sparse structure and reduced thickness, as well as diminished adhesion and flocculation abilities. Deletion of the Sho1 gene prolonged mouse survival; decreased the fungal load in the liver, kidney, and spleen; and reduced inflammatory cell infiltration into the kidney. In the spleens of mice injected with the Sho1Δ/Δ strain, a decrease was observed in the percentage of M1-type macrophages and an increase in M2-type macrophages, resulting in a decreased M1/M2 macrophage ratio. Additionally, an increase was observed in the number of Th1 cells and a decrease in the number of Th2 and Th17 cells, leading to an increased Th1/Th2 ratio. Conclusion The Sho1 gene significantly contributes to the pathogenesis of C. albicans by influencing its biological behaviour and immune response in vivo.
Collapse
Affiliation(s)
| | | | - Huihai Zhao
- Clinical Laboratory, The 980Th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), 398 Zhongshan Road, Shijiazhuang, Hebei, 050082, China
| | - Mengyu Jiang
- Clinical Laboratory, The 980Th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), 398 Zhongshan Road, Shijiazhuang, Hebei, 050082, China
| | - Mengge Cui
- Clinical Laboratory, The 980Th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), 398 Zhongshan Road, Shijiazhuang, Hebei, 050082, China
| | - Keran Jia
- Clinical Laboratory, The 980Th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), 398 Zhongshan Road, Shijiazhuang, Hebei, 050082, China
| | - Daxin Lei
- Clinical Laboratory, The 980Th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), 398 Zhongshan Road, Shijiazhuang, Hebei, 050082, China
| | - Fukun Wang
- Clinical Laboratory, The 980Th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), 398 Zhongshan Road, Shijiazhuang, Hebei, 050082, China
| |
Collapse
|
2
|
Xiong J, Lu H, Jiang Y. A Causal Relationship between Type 2 Diabetes and Candidiasis through Two-Sample Mendelian Randomization Analysis. Microorganisms 2024; 12:1984. [PMID: 39458293 PMCID: PMC11509823 DOI: 10.3390/microorganisms12101984] [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: 08/16/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
The potential relationship between type 2 diabetes (T2D) and candidiasis is of concern due to the respective characteristics of these conditions, yet the exact causal link between the two remains uncertain and requires further investigation. In this study, the inverse-variance-weighted (IVW) analysis indicated a significant genetic causal relationship between T2D and candidiasis (p = 0.0264, Odds Ratio [OR], 95% confidence interval [CI] = 1.1046 [0.9096-1.2996]), T2D (wide definition) and candidiasis (p = 0.0031, OR 95% [CI] = 1.1562 [0.8718-1.4406]), and severe autoimmune T2D and candidiasis (p = 0.0041, OR 95% [CI] = 1.0559 [0.9493-1.1625]). Additionally, the MR-Egger analyses showed a significant genetic causal relationship between T2D (wide definition) and candidiasis (p = 0.0154, OR 95% [CI] = 1.3197 [0.7760-1.8634]). The weighted median analyses showed a significant genetic causal relationship between severe autoimmune T2D and candidiasis (p = 0.0285, OR 95% [CI] = 1.0554 [0.9498-1.1610]). This Mendelian randomization (MR) study provides evidence for a genetic correlation between T2D and candidiasis.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
3
|
Cheng M, Liu S, Zhu M, Li M, Yu Q. Adhesin Antibody-Grafted Mesoporous Silica Nanoparticles Suppress Immune Escape for Treatment of Fungal Systemic Infection. Molecules 2024; 29:4547. [PMID: 39407477 PMCID: PMC11478059 DOI: 10.3390/molecules29194547] [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: 08/19/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Life-threatening systemic fungal infections caused by Candida albicans are significant contributors to clinical mortality, particularly among cancer patients and immunosuppressed individuals. The evasion of the immune response facilitated by fungal surface components enables fungal pathogens to evade macrophage attacks and to establish successful infections. This study developed a mesoporous silica nanoplatform, i.e., MSNP-EAP1Ab, which is composed of mesoporous silica nanoparticles grafted with the antibody of C. albicans surface adhesin Eap1. The activity of MSNP-EAP1Ab against C. albicans immune escape and infection was then evaluated by using the cell interaction model and the mouse systemic infection model. During interaction between C. albicans cells and macrophages, MSNP-EAP1Ab significantly inhibited fungal immune escape, leading to the enhanced phagocytosis of fungal cells by macrophages, with phagocytosis rates increasing from less than 8% to 14%. Furthermore, after treatment of the C. albicans-infected mice, MSNP-EAP1Ab drastically prolonged the mouse survival time and decreased the kidney fungal burden from >30,0000 CFU/g kidney to <100 CFU/g kidney, indicating the rapid recognition and killing of the pathogens by immune cells. Moreover, MSNP-EAP1Ab attenuated kidney tissue inflammation, with remarkable attenuation of renal immune cell accumulation. This study presents an innovative nanoplatform that targets the C. albicans adhesin, offering a promising approach for combatting systemic fungal infections.
Collapse
Affiliation(s)
- Mengjuan Cheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Suke Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
| | - Mengsen Zhu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
| | - Mingchun Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
| | - Qilin Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
4
|
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 2024:118826. [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] [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
|
5
|
Zhen C, Wang L, Feng Y, Whiteway M, Hang S, Yu J, Lu H, Jiang Y. Otilonium Bromide Exhibits Potent Antifungal Effects by Blocking Ergosterol Plasma Membrane Localization and Triggering Cytotoxic Autophagy in Candida Albicans. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406473. [PMID: 38995235 PMCID: PMC11425263 DOI: 10.1002/advs.202406473] [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: 06/11/2024] [Indexed: 07/13/2024]
Abstract
Candidiasis, which presents a substantial risk to human well-being, is frequently treated with azoles. However, drug-drug interactions caused by azoles inhibiting the human CYP3A4 enzyme, together with increasing resistance of Candida species to azoles, represent serious issues with this class of drug, making it imperative to develop innovative antifungal drugs to tackle this growing clinical challenge. A drug repurposing approach is used to examine a library of Food and Drug Administration (FDA)-approved drugs, ultimately identifying otilonium bromide (OTB) as an exceptionally encouraging antifungal agent. Mechanistically, OTB impairs vesicle-mediated trafficking by targeting Sec31, thereby impeding the plasma membrane (PM) localization of the ergosterol transporters, such as Sip3. Consequently, OTB obstructs the movement of ergosterol across membranes and triggers cytotoxic autophagy. It is noteworthy that C. albicans encounters challenges in developing resistance to OTB because it is not a substrate for drug transporters. This study opens a new door for antifungal therapy, wherein OTB disrupts ergosterol subcellular distribution and induces cytotoxic autophagy. Additionally, it circumvents the hepatotoxicity associated with azole-mediated liver enzyme inhibition and avoids export-mediated drug resistance in C. albicans.
Collapse
Affiliation(s)
- Cheng Zhen
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Li Wang
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Malcolm Whiteway
- Department of BiologyConcordia UniversityMontrealQCH4B 1R6Canada
| | - Sijin Hang
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Jinhua Yu
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| |
Collapse
|
6
|
Lin J, Peng CX, Huang WM. Comparative efficacy of antifungal drugs for the treatment of oral candidiasis in HIV-positive patients: A Bayesian network meta-analysis. Med Clin (Barc) 2024:S0025-7753(24)00393-2. [PMID: 39214731 DOI: 10.1016/j.medcli.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 09/04/2024]
Abstract
Oral candidiasis infection is particularly prevalent among individuals in HIV-positive patients. Antifungal drugs have shown promising therapeutic effects in treating oral candidiasis in HIV-positive patients. However, the selection of specific antifungal drugs for the treatment of oral candidiasis in HIV-positive patients lacks evidence-based guidelines. This study aims to address this gap by conducting a comprehensive review of relevant randomized controlled trials (RCTs) and performing a network meta-analysis to assess the efficacy of different antifungal drugs in treating oral candidiasis in HIV-positive patients. A systematic search was conducted in databases including EMBASE, Web of Science, Medline, and Cochrane databases to identify relevant articles. Additionally, key pertinent sources in the literatures were also reviewed. All studies published prior to August 2023 were eligible for inclusion. Two researchers independently conducted the screening of literature, extraction of data, and evaluation of quality. Pairwise and network meta-analysis were then performed to assess the primary outcomes of the randomized controlled trials (RCTs) included. The protocol was registered on the PROSPERO database (CRD42024513912). Twenty-six RCTs were included in this meta-analysis, involving a total of 3145 patients and evaluating seven interventions (placebo, fluconazole, itraconazole, nystatin, clotrimazole, ketoconazole, miconazole). Pairwise meta-analysis and network meta-analysis showed fluconazole was significantly efficacy in increasing mycological cure rates when compared with placebo, clotrimazole, and nystatin. Ketoconazole and miconazole were significantly efficacy in increasing mycological cure rates when compared with nystatin. Network meta-analysis also suggested the efficacy of the seven interventions in increasing mycological cure rates was ranked as follows: placebo (35.3%), fluconazole (95.2%), itraconazole (61.6%), nystatin (17.0%), clotrimazole (52.7%), ketoconazole (69.2%), miconazole (69.1%). The available evidence indicates that fluconazole had the greatest possibility to increase mycological cure rates in HIV-positive patients, while, nystatin was the least effective antifungal drug in increasing mycological cure rates in HIV-positive patients.
Collapse
Affiliation(s)
- Jiong Lin
- Department of Stomatology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518100, Shenzhen, China.
| | - Cai-Xia Peng
- Department of Stomatology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518100, Shenzhen, China
| | - Wei-Man Huang
- Department of Stomatology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518100, Shenzhen, China
| |
Collapse
|
7
|
Hu Y, Zeng G, Wang Y, Yang D. Nanorobots to Treat Candida albicans Infection. RESEARCH (WASHINGTON, D.C.) 2024; 10:0455. [PMID: 39148662 PMCID: PMC11324951 DOI: 10.34133/research.0455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Candida albicans is an opportunistic fungal pathogen of humans. It causes a variety of infections ranging from superficial mucocutaneous conditions to severe systemic diseases that result in substantial morbidity and mortality. This pathogen frequently forms biofilms resistant to antifungal drugs and the host immune system, leading to treatment failures. Recent research has demonstrated the potential of nanorobots to penetrate biological barriers and disrupt fungal biofilms. In this perspective paper, we provide a brief overview of recent breakthroughs in nanorobots for candidiasis treatment and discuss current challenges and prospects.
Collapse
Affiliation(s)
- Yanling Hu
- College of Life and Health, Nanjing Polytechnic Institute, Nanjing 210048, China
| | - Guisheng Zeng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
| | - Yue Wang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
| |
Collapse
|
8
|
Wu H, Ji Z, Huang X, Li L, Hang S, Yu J, Lu H, Jiang Y. Isobavachalcone Exhibits Potent Antifungal Efficacy by Inhibiting Enolase Activity and Glycolysis in Candida albicans. ACS Infect Dis 2024; 10:3059-3070. [PMID: 38995732 DOI: 10.1021/acsinfecdis.4c00399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Invasive fungal diseases (IFDs) are becoming increasingly acknowledged as a significant concern linked to heightened rates of morbidity and mortality. Regrettably, the available antifungal therapies for managing IFDs are constrained. Emerging evidence indicates that enolase holds promise as a potential target protein for combating IFDs; however, there is currently a deficiency in antifungal medications specifically targeting enolase. This study establishes that isobavachalcone (IBC) exhibits noteworthy antifungal efficacy both in vitro and in vivo. Moreover, our study has demonstrated that IBC effectively targets Eno1 in Candida albicans (CaEno1), resulting in the suppression of the glycolytic pathway. Additionally, our research has indicated that IBC exhibits a higher affinity for CaEno1 compared to human Eno1 (hEno1), with the presence of isoprenoid in the side chain of IBC playing a crucial role in its ability to inhibit enolase activity. These findings contribute to the comprehension of antifungal approaches that target Eno1, identifying IBC as a potential inhibitor of Eno1 in human pathogenic fungi.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhe Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xin Huang
- Department of Dermatology, Hair Medical Center of Shanghai Tongji Hospital, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Sijin Hang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jinhua Yu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
9
|
Jiang C, Li F, Song P, Wen M, Yang S, Tian G, Shao D, Shi J, Shang L. Multifunctional Gold Nanozyme-Engineered Amphotericin B for Enhanced Antifungal Infection Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312253. [PMID: 38501846 DOI: 10.1002/smll.202312253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Chronic wounds of significant severity and acute injuries are highly vulnerable to fungal infections, drastically impeding the expected wound healing trajectory. The clinical use of antifungal therapeutic drug is hampered by poor solubility, high toxicity and adverse reactions, thereby necessitating the urgent development of novel antifungal therapy strategy. Herein, this study proposes a new strategy to enhance the bioactivity of small-molecule antifungal drugs based on multifunctional metal nanozyme engineering, using amphotericin B (AmB) as an example. AmB-decorated gold nanoparticles (AmB@AuNPs) are synthesized by a facile one-pot reaction strategy, and the AmB@AuNPs exhibit superior peroxidase (POD)-like enzyme activity, with maximal reaction rates (Vmax) 3.4 times higher than that of AuNPs for the catalytic reaction of H2O2. Importantly, the enzyme-like activity of AuNPs significantly enhanced the antifungal properties of AmB, and the minimum inhibitory concentrations of AmB@AuNPs against Candida albicans (C. albicans) and Saccharomyces cerevisiae (S. cerevisiae) W303 are reduced by 1.6-fold and 50-fold, respectively, as compared with AmB alone. Concurrent in vivo studies conducted on fungal-infected wounds in mice underscored the fundamentally superior antifungal ability and biosafety of AmB@AuNPs. The proposed strategy of engineering antifungal drugs with nanozymes has great potential for enhanced therapy of fungal infections and related diseases.
Collapse
Affiliation(s)
- Chunmei Jiang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Fangping Li
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Pei Song
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Mengyao Wen
- State Key Laboratory of Solidification Processing, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Saixue Yang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Geng Tian
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Li Shang
- State Key Laboratory of Solidification Processing, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| |
Collapse
|
10
|
Xiong J, Wang L, Feng Y, Zhen C, Hang S, Yu J, Lu H, Jiang Y. Geldanamycin confers fungicidal properties to azole by triggering the activation of succinate dehydrogenase. Life Sci 2024; 348:122699. [PMID: 38718854 DOI: 10.1016/j.lfs.2024.122699] [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: 03/06/2024] [Revised: 04/17/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
AIMS Azoles have been widely employed for the treatment of invasive fungal diseases; however, their efficacy is diminished as pathogenic fungi tolerate them due to their fungistatic properties. Geldanamycin (GdA) can render azoles fungicidal by inhibiting the ATPase and molecular chaperone activities of heat shock protein 90 (Hsp90). Nonetheless, the clinical applicability of GdA is restricted due to its cytotoxic ansamycin scaffold structure, its induction of cytoprotective heat shock responses, and the conservative nature of Hsp90. Hence, it is imperative to elucidate the mechanism of action of GdA to confer fungicidal properties to azoles and mitigate the toxic adverse effects associated with GdA. MATERIALS AND METHODS Through various experimental methods, including the construction of gene-deleted Candida albicans mutants, in vitro drug sensitivity experiments, Western blot analysis, reactive oxygen species (ROS) assays, and succinate dehydrogenase activity assays, we identified Hsp90 client proteins associated with the tolerance of C. albicans to azoles. KEY FINDINGS It was observed that GdA effectively hindered the entry of Hsp90 into mitochondria, resulting in the alleviation of inhibitory effect of Hsp90 on succinate dehydrogenase. Consequently, the activation of succinate dehydrogenase led to an increased production of ROS. within the mitochondria, thereby facilitating the antifungal effects of azoles against C. albicans. SIGNIFICANCE This research presents a novel approach for conferring fungicidal properties to azoles, which involves specifically disrupting the interaction of between Hsp90 and succinate dehydrogenase rather than employing a non-specific inhibition of ATPase activity of Hsp90.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Li Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Zhen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Sijin Hang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jinhua Yu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
11
|
Schaefer KG, Russell CM, Pyron RJ, Conley EA, Barrera FN, King GM. Polymerization mechanism of the Candida albicans virulence factor candidalysin. J Biol Chem 2024; 300:107370. [PMID: 38750794 PMCID: PMC11193009 DOI: 10.1016/j.jbc.2024.107370] [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: 03/05/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Candida albicans is a commensal fungus that can cause epithelial infections and life-threatening invasive candidiasis. The fungus secretes candidalysin (CL), a peptide that causes cell damage and immune activation by permeation of epithelial membranes. The mechanism of CL action involves strong peptide assembly into polymers in solution. The free ends of linear CL polymers can join, forming loops that become pores upon binding to membranes. CL polymers constitute a therapeutic target for candidiasis, but little is known about CL self-assembly in solution. Here, we examine the assembly mechanism of CL in the absence of membranes using complementary biophysical tools, including a new fluorescence polymerization assay, mass photometry, and atomic force microscopy. We observed that CL assembly is slow, as tracked with the fluorescent marker C-laurdan. Single-molecule methods showed that CL polymerization involves a convolution of four processes. Self-assembly begins with the formation of a basic subunit, thought to be a CL octamer that is the polymer seed. Polymerization proceeds via the addition of octamers, and as polymers grow they can curve and form loops. Alternatively, secondary polymerization can occur and cause branching. Interplay between the different rates determines the distribution of CL particle types, indicating a kinetic control mechanism. This work elucidates key physical attributes underlying CL self-assembly which may eventually evoke pharmaceutical development.
Collapse
Affiliation(s)
| | - Charles M Russell
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | - Robert J Pyron
- Genome Science and Technology, University of Tennessee, Knoxville, Tennessee
| | - Elizabeth A Conley
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee.
| | - Gavin M King
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri; Department of Biochemistry, University of Missouri, Columbia, Missouri.
| |
Collapse
|
12
|
Li W, Feng Y, Feng Z, Wang L, Whiteway M, Lu H, Jiang Y. Pitavastatin Calcium Confers Fungicidal Properties to Fluconazole by Inhibiting Ubiquinone Biosynthesis and Generating Reactive Oxygen Species. Antioxidants (Basel) 2024; 13:667. [PMID: 38929106 PMCID: PMC11200976 DOI: 10.3390/antiox13060667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Fluconazole (FLC) is extensively employed for the prophylaxis and treatment of invasive fungal infections (IFIs). However, the fungistatic nature of FLC renders pathogenic fungi capable of developing tolerance towards it. Consequently, converting FLC into a fungicidal agent using adjuvants assumes significance to circumvent FLC resistance and the perpetuation of fungal infections. This drug repurposing study has successfully identified pitavastatin calcium (PIT) as a promising adjuvant for enhancing the fungicidal activity of FLC from a comprehensive library of 2372 FDA-approved drugs. PIT could render FLC fungicidal even at concentrations as low as 1 μM. The median lethal dose (LD50) of PIT was determined to be 103.6 mg/kg. We have discovered that PIT achieves its synergistic effect by inhibiting the activity of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, thereby impeding ubiquinone biosynthesis, inducing reactive oxygen species (ROS) generation, triggering apoptosis, and disrupting Golgi function. We employed a Candida albicans strain that demonstrated a notable tolerance to FLC to infect mice and found that PIT effectively augmented the antifungal efficacy of FLC against IFIs. This study is an illustrative example of how FDA-approved drugs can effectively eliminate fungal tolerance to FLC.
Collapse
Affiliation(s)
- Wanqian Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yanru Feng
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Zhe Feng
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Li Wang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
13
|
Hang S, Lu H, Jiang Y. Marine-Derived Metabolites Act as Promising Antifungal Agents. Mar Drugs 2024; 22:180. [PMID: 38667797 PMCID: PMC11051449 DOI: 10.3390/md22040180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
The incidence of invasive fungal diseases (IFDs) is on the rise globally, particularly among immunocompromised patients, leading to significant morbidity and mortality. Current clinical antifungal agents, such as polyenes, azoles, and echinocandins, face increasing resistance from pathogenic fungi. Therefore, there is a pressing need for the development of novel antifungal drugs. Marine-derived secondary metabolites represent valuable resources that are characterized by varied chemical structures and pharmacological activities. While numerous compounds exhibiting promising antifungal activity have been identified, a comprehensive review elucidating their specific underlying mechanisms remains lacking. In this review, we have compiled a summary of antifungal compounds derived from marine organisms, highlighting their diverse mechanisms of action targeting various fungal cellular components, including the cell wall, cell membrane, mitochondria, chromosomes, drug efflux pumps, and several biological processes, including vesicular trafficking and the growth of hyphae and biofilms. This review is helpful for the subsequent development of antifungal drugs due to its summary of the antifungal mechanisms of secondary metabolites from marine organisms.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 200092 Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 200092 Shanghai, China
| |
Collapse
|
14
|
Maggini V, Semenzato G, Gallo E, Nunziata A, Fani R, Firenzuoli F. Antimicrobial Activity of Syzygium aromaticum Essential Oil in Human Health Treatment. Molecules 2024; 29:999. [PMID: 38474510 DOI: 10.3390/molecules29050999] [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: 01/12/2024] [Revised: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The use of natural compounds to prevent and treat infective diseases is increasing its importance, especially in the case of multidrug-resistant (MDR) microorganisms-mediated infections. The drug resistance phenomenon is today a global problem, so it is important to have available substances able to counteract MDR infections. Syzygium aromaticum (L.) Merr. & L.M. Perry (commonly called clove) is a spice characterized by several biological properties. Clove essential oil (EO) consists of numerous active molecules, being eugenol as the principal component; however, other compounds that synergize with each other are responsible for the biological properties of the EO. S. aromaticum is traditionally used for bowel and stomach disorders, cold and flu, oral hygiene, tooth decay, and for its analgesic action. Its EO has shown antioxidant, antimicrobial, anti-inflammatory, neuro-protective, anti-stress, anticancer, and anti-nociceptive activities. This review aims to investigate the role of E. S. aromaticum EO in the counteraction of MDR microorganisms responsible for human disorders, diseases, or infections, such as Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, Salmonella typhi, Candida albicans, Giardia lamblia, Streptococcus mutans, Porphyromonas gingivalis, and Klebsiella pneumoniae. This study might orient clinical researchers on future therapeutic uses of S. aromaticum EO in the prevention and treatment of infectious diseases.
Collapse
Affiliation(s)
- Valentina Maggini
- Center for Integrative Medicine, Careggi University Hospital, University of Florence, 50124 Florence, Italy
| | - Giulia Semenzato
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Eugenia Gallo
- Center for Integrative Medicine, Careggi University Hospital, University of Florence, 50124 Florence, Italy
| | - Alessia Nunziata
- Center for Integrative Medicine, Careggi University Hospital, University of Florence, 50124 Florence, Italy
| | - Renato Fani
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Fabio Firenzuoli
- Center for Integrative Medicine, Careggi University Hospital, University of Florence, 50124 Florence, Italy
| |
Collapse
|
15
|
Xiong J, Wang L, Feng Z, Hang S, Yu J, Feng Y, Lu H, Jiang Y. Halofantrine Hydrochloride Acts as an Antioxidant Ability Inhibitor That Enhances Oxidative Stress Damage to Candida albicans. Antioxidants (Basel) 2024; 13:223. [PMID: 38397821 PMCID: PMC10886025 DOI: 10.3390/antiox13020223] [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: 12/20/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Candida albicans, a prominent opportunistic pathogenic fungus in the human population, possesses the capacity to induce life-threatening invasive candidiasis in individuals with compromised immune systems despite the existence of antifungal medications. When faced with macrophages or neutrophils, C. albicans demonstrates its capability to endure oxidative stress through the utilization of antioxidant enzymes. Therefore, the enhancement of oxidative stress in innate immune cells against C. albicans presents a promising therapeutic approach for the treatment of invasive candidiasis. In this study, we conducted a comprehensive analysis of a library of drugs approved by the Food and Drug Administration (FDA). We discovered that halofantrine hydrochloride (HAL) can augment the antifungal properties of oxidative damage agents (plumbagin, menadione, and H2O2) by suppressing the response of C. albicans to reactive oxygen species (ROS). Furthermore, our investigation revealed that the inhibitory mechanism of HAL on the oxidative response is dependent on Cap1. In addition, the antifungal activity of HAL has been observed in the Galleria mellonella infection model. These findings provide evidence that targeting the oxidative stress response of C. albicans and augmenting the fungicidal capacity of oxidative damage agents hold promise as effective antifungal strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
16
|
Feng Z, Lu H, Jiang Y. Promising immunotherapeutic targets for treating candidiasis. Front Cell Infect Microbiol 2024; 14:1339501. [PMID: 38404288 PMCID: PMC10884116 DOI: 10.3389/fcimb.2024.1339501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
In the last twenty years, there has been a significant increase in invasive fungal infections, which has corresponded with the expanding population of individuals with compromised immune systems. As a result, the mortality rate linked to these infections remains unacceptably high. The currently available antifungal drugs, such as azoles, polyenes, and echinocandins, face limitations in terms of their diversity, the escalating resistance of fungi and the occurrence of significant adverse effects. Consequently, there is an urgent need to develop new antifungal medications. Vaccines and antibodies present a promising avenue for addressing fungal infections due to their targeted antifungal properties and ability to modulate the immune response. This review investigates the structure and function of cell wall proteins, secreted proteins, and functional proteins within C. albicans. Furthermore, it seeks to analyze the current advancements and challenges in macromolecular drugs to identify new targets for the effective management of candidiasis.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
17
|
Chen T, Yao J, Quan K, Xu J, Hang X, Tong Q, Liu G, Luo P, Zeng L, Feng G, Bi H. Repurposing a human acetyl-CoA carboxylase inhibitor firsocostat to treat fungal candidiasis alone and in combination. Antimicrob Agents Chemother 2024; 68:e0113123. [PMID: 38018962 PMCID: PMC10777831 DOI: 10.1128/aac.01131-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/15/2023] [Indexed: 11/30/2023] Open
Abstract
Opportunistic fungal infections, particularly caused by Candida albicans, remain a common cause of high morbidity and mortality in immunocompromised patients. The escalating prevalence of antifungal drug resistance necessitates the immediate exploration of alternative treatment strategies to combat these life-threatening fungal diseases. In this study, we investigated the antifungal efficacy of firsocostat, a human acetyl-CoA carboxylase (ACC) inhibitor, against C. albicans. Firsocostat alone displayed moderate antifungal activity, while combining it with voriconazole, itraconazole, or amphotericin B exhibited synergistic effects across almost all drug-sensitive and drug-resistant C. albicans strains tested. These observed synergies were further validated in two mouse models of oropharyngeal and systemic candidiasis, where the combination therapies demonstrated superior fungicidal effects compared to monotherapy. Moreover, firsocostat was shown to directly bind to C. albicans ACC and inhibit its enzymatic activity. Sequencing spontaneous firsocostat-resistant mutants revealed mutations mapping to C. albicans ACC, confirming that firsocostat has retained its target in C. albicans. Overall, our findings suggest that repurposing firsocostat, either alone or in combination with other antifungal agents, holds promising potential in the development of antifungal drugs and the treatment of candidiasis.
Collapse
Affiliation(s)
- Tianyu Chen
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jing Yao
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Keao Quan
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingchen Xu
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xudong Hang
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Tong
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Genyan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peipei Luo
- Department of Gastroenterology, Wujin People’s Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu, China
| | - Liping Zeng
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ganzhu Feng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongkai Bi
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Seiser S, Arzani H, Ayub T, Phan-Canh T, Staud C, Worda C, Kuchler K, Elbe-Bürger A. Native human and mouse skin infection models to study Candida auris-host interactions. Microbes Infect 2024; 26:105234. [PMID: 37813159 DOI: 10.1016/j.micinf.2023.105234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
The World Health Organization (WHO) declared certain fungal pathogens as global health threats for the next decade. Candida auris (C. auris) is a newly emerging skin-tropic multidrug-resistant fungal pathogen that can cause life-threatening infections of high mortality in hospitals and healthcare settings. Here, we address an unmet need and present novel native ex vivo skin models, thus extending previous C. auris-host interaction studies. We exploit histology and immunofluorescence analysis of ex vivo skin biopsies of human adult and fetal, as well as mouse origin infected with C. auris via distinct routes. We demonstrate that an intact skin barrier efficiently protects from C. auris penetration and invasion. Although C. auris readily grows on native human skin, it can reach deeper layers only upon physical disruption of the barrier by needling or through otherwise damaged skin. By contrast, a barrier disruption is not necessary for C. auris penetration of native mouse skin. Importantly, we show that C. auris undergoes morphogenetic changes upon skin penetration, as it acquires pseudohyphal growth phenotypes in deeper human and mouse dermis. Taken together, this new human and mouse skin model toolset yields new insights into C. auris colonization, adhesion, growth and invasion properties of native versus damaged human skin. The results form a crucial basis for future studies on skin immune defense to colonizing pathogens, and offer new options for testing the action and efficacy of topical antimicrobial compound formulations.
Collapse
Affiliation(s)
- Saskia Seiser
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Hossein Arzani
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Tanya Ayub
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Trinh Phan-Canh
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Clement Staud
- Medical University of Vienna, Department of Plastic and Reconstructive Surgery, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Christof Worda
- Medical University of Vienna, Department of Obstetrics and Gynecology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria.
| | - Adelheid Elbe-Bürger
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
19
|
Cruz Sánchez E, García MT, Gracia I, Fernández-Bermejo SI, Rodríguez JF, García-Vargas JM, Vidal Roig D. Antibacterial Activity Assessment of Chitosan/Alginate Lavender Essential Oil Membranes for Biomedical Applications. MEMBRANES 2024; 14:12. [PMID: 38248702 PMCID: PMC10818663 DOI: 10.3390/membranes14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024]
Abstract
The demand for natural products in the treatment of dermatological pathologies has boosted the use of bioactive substances such as lavender essential oil (LEO), which stands out for its anti-inflammatory and antioxidant properties and its antimicrobial potential. Biopolymers such as chitosan (CHT) and alginate (ALG) are biodegradable and biocompatible and have proven their viability in biomedical applications such as skin regeneration. The inhibitory effect of LEO on the growth of skin-related bacterial species Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa and the fungus Candida albicans was studied by incorporating 1% v/v LEO encapsulated in CHT, ALG, and CHT/ALG membranes. Despite the verification of the antimicrobial effect of all type of membranes, no synergistic effect was observed following the addition of LEO. S. aureus and P. aeruginosa showed the most growth on the different substrates and C. albicans demonstrated the highest inhibition. This is a first approach using microorganisms isolated from clinical samples or skin microbiota. Further investigation would be advisable using more clinical strains for each microorganism to validate their biomedical applicability.
Collapse
Affiliation(s)
- Encarnación Cruz Sánchez
- Department of Chemical Engineering, Facultad de Ciencias y Tecnologías Químicas, University of Castilla-La Mancha, Avda. Camilo José Cela 12, 13071 Ciudad Real, Spain; (E.C.S.); (M.T.G.); (I.G.); (J.F.R.); (J.M.G.-V.)
| | - María Teresa García
- Department of Chemical Engineering, Facultad de Ciencias y Tecnologías Químicas, University of Castilla-La Mancha, Avda. Camilo José Cela 12, 13071 Ciudad Real, Spain; (E.C.S.); (M.T.G.); (I.G.); (J.F.R.); (J.M.G.-V.)
| | - Ignacio Gracia
- Department of Chemical Engineering, Facultad de Ciencias y Tecnologías Químicas, University of Castilla-La Mancha, Avda. Camilo José Cela 12, 13071 Ciudad Real, Spain; (E.C.S.); (M.T.G.); (I.G.); (J.F.R.); (J.M.G.-V.)
| | - Soledad Illescas Fernández-Bermejo
- Department of Medical Sciences, Microbiology Area, Facultad de Medicina, University of Castilla-La Mancha, Paseo de Moledores s/n, 13071 Ciudad Real, Spain;
- Department of Microbiology, Hospital General Universitario de Ciudad Real, Obispo Rafael Torija s/n, 13005 Ciudad Real, Spain
| | - Juan Francisco Rodríguez
- Department of Chemical Engineering, Facultad de Ciencias y Tecnologías Químicas, University of Castilla-La Mancha, Avda. Camilo José Cela 12, 13071 Ciudad Real, Spain; (E.C.S.); (M.T.G.); (I.G.); (J.F.R.); (J.M.G.-V.)
| | - Jesús Manuel García-Vargas
- Department of Chemical Engineering, Facultad de Ciencias y Tecnologías Químicas, University of Castilla-La Mancha, Avda. Camilo José Cela 12, 13071 Ciudad Real, Spain; (E.C.S.); (M.T.G.); (I.G.); (J.F.R.); (J.M.G.-V.)
| | - Dolors Vidal Roig
- Department of Medical Sciences, Microbiology Area, Facultad de Medicina, University of Castilla-La Mancha, Paseo de Moledores s/n, 13071 Ciudad Real, Spain;
| |
Collapse
|
20
|
Wu M, Xu X, Hu R, Chen Q, Chen L, Yuan Y, Li J, Zhou L, Feng S, Wang L, Chen S, Gu M. A Membrane-Targeted Photosensitizer Prevents Drug Resistance and Induces Immune Response in Treating Candidiasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207736. [PMID: 37875397 PMCID: PMC10724446 DOI: 10.1002/advs.202207736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Candida albicans (C. albicans), a ubiquitous polymorphic fungus in humans, causes different types of candidiasis, including oral candidiasis (OC) and vulvovaginal candidiasis (VVC), which are physically and mentally concerning and financially costly. Thus, developing alternative antifungals that prevent drug resistance and induce immunity to eliminate Candida biofilms is crucial. Herein, a novel membrane-targeted aggregation-induced emission (AIE) photosensitizer (PS), TBTCP-QY, is developed for highly efficient photodynamic therapy (PDT) of candidiasis. TBTCP-QY has a high molar absorption coefficient and an excellent ability to generate 1 O2 and •OH, entering the interior of biofilms due to its high permeability. Furthermore, TBTCP-QY can efficiently inhibit biofilm formation by suppressing the expression of genes related to the adhesion (ALS3, EAP1, and HWP1), invasion (SAP1 and SAP2), and drug resistance (MDR1) of C. albicans, which is also advantageous for eliminating potential fungal resistance to treat clinical infectious diseases. TBTCP-QY-mediated PDT efficiently targets OC and VVC in vivo in a mouse model, induces immune response, relieves inflammation, and accelerates the healing of mucosal defects to combat infections caused by clinically isolated fluconazole-resistant strains. Moreover, TBTCP-QY demonstrates excellent biocompatibility, suggesting its potential applications in the clinical treatment of OC and VVC.
Collapse
Affiliation(s)
- Ming‐Yu Wu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Xiaoyu Xu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Rui Hu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Qingrong Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Luojia Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Yuncong Yuan
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Jie Li
- Department of Medical Intensive Care UnitMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430070China
| | - Li Zhou
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Lianrong Wang
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Shi Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Meijia Gu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| |
Collapse
|
21
|
Fang T, Xiong J, Wang L, Feng Z, Hang S, Yu J, Li W, Feng Y, Lu H, Jiang Y. Unexpected Inhibitory Effect of Octenidine Dihydrochloride on Candida albicans Filamentation by Impairing Ergosterol Biosynthesis and Disrupting Cell Membrane Integrity. Antibiotics (Basel) 2023; 12:1675. [PMID: 38136708 PMCID: PMC10741164 DOI: 10.3390/antibiotics12121675] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Candida albicans filamentation plays a significant role in developing both mucosal and invasive candidiasis, making it a crucial virulence factor. Consequently, exploring and identifying inhibitors that impede fungal hyphal formation presents an intriguing approach toward antifungal strategies. In line with this anti-filamentation strategy, we conducted a comprehensive screening of a library of FDA-approved drugs to identify compounds that possess inhibitory properties against hyphal growth. The compound octenidine dihydrochloride (OCT) exhibits potent inhibition of hyphal growth in C. albicans across different hyphae-inducing media at concentrations below or equal to 3.125 μM. This remarkable inhibitory effect extends to biofilm formation and the disruption of mature biofilm. The mechanism underlying OCT's inhibition of hyphal growth is likely attributed to its capacity to impede ergosterol biosynthesis and induce the generation of reactive oxygen species (ROS), compromising the integrity of the cell membrane. Furthermore, it has been observed that OCT demonstrates protective attributes against invasive candidiasis in Galleria mellonella larvae through its proficient eradication of C. albicans colonization in infected G. mellonella larvae by impeding hyphal formation. Although additional investigation is required to mitigate the toxicity of OCT in mammals, it possesses considerable promise as a potent filamentation inhibitor against invasive candidiasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
22
|
Menu E, Filori Q, Dufour JC, Ranque S, L’Ollivier C. A Repertoire of the Less Common Clinical Yeasts. J Fungi (Basel) 2023; 9:1099. [PMID: 37998905 PMCID: PMC10671991 DOI: 10.3390/jof9111099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Invasive fungal diseases are a public health problem. They affect a constantly increasing number of at-risk patients, and their incidence has risen in recent years. These opportunistic infections are mainly due to Candida sp. but less common or rare yeast infections should not be underestimated. These so-called "less common" yeasts include Ascomycota of the genera Candida (excluding the five major Candida species), Magnusiomyces/Saprochaete, Malassezia, and Saccharomyces, and Basidiomycota of the genera Cryptococcus (excluding the Cryptococcus neoformans/gattii complex members), Rhodotorula, and Trichosporon. The aim of this review is to (i) inventory the less common yeasts isolated in humans, (ii) provide details regarding the specific anatomical locations where they have been detected and the clinical characteristics of the resulting infections, and (iii) provide an update on yeast taxonomy. Of the total of 239,890 fungal taxa and their associated synonyms sourced from the MycoBank and NCBI Taxonomy databases, we successfully identified 192 yeasts, including 127 Ascomycota and 65 Basidiomycota. This repertoire allows us to highlight rare yeasts and their tropism for certain anatomical sites and will provide an additional tool for diagnostic management.
Collapse
Affiliation(s)
- Estelle Menu
- Laboratoire de Parasitologie-Mycologie, IHU Méditerranée Infection, 13385 Marseille, France; (S.R.); (C.L.)
- Institut de Recherche pour le Développement, Assistance Publique-Hôpitaux de Marseille, Service de Santé des Armées, VITROME: Vecteurs-Infections Tropicales et Méditerranéennes, Aix Marseille Université, 13385 Marseille, France
| | - Quentin Filori
- INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l’Information Médicale, ISSPAM, Aix Marseille University, 13385 Marseille, France; (Q.F.); (J.-C.D.)
| | - Jean-Charles Dufour
- INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l’Information Médicale, ISSPAM, Aix Marseille University, 13385 Marseille, France; (Q.F.); (J.-C.D.)
- APHM, Hôpital de la Timone, Service Biostatistique et Technologies de l’Information et de la Communication, 13385 Marseille, France
| | - Stéphane Ranque
- Laboratoire de Parasitologie-Mycologie, IHU Méditerranée Infection, 13385 Marseille, France; (S.R.); (C.L.)
- Institut de Recherche pour le Développement, Assistance Publique-Hôpitaux de Marseille, Service de Santé des Armées, VITROME: Vecteurs-Infections Tropicales et Méditerranéennes, Aix Marseille Université, 13385 Marseille, France
| | - Coralie L’Ollivier
- Laboratoire de Parasitologie-Mycologie, IHU Méditerranée Infection, 13385 Marseille, France; (S.R.); (C.L.)
- Institut de Recherche pour le Développement, Assistance Publique-Hôpitaux de Marseille, Service de Santé des Armées, VITROME: Vecteurs-Infections Tropicales et Méditerranéennes, Aix Marseille Université, 13385 Marseille, France
| |
Collapse
|
23
|
Kaur G, Chawla S, Kumar P, Singh R. Advancing Vaccine Strategies against Candida Infections: Exploring New Frontiers. Vaccines (Basel) 2023; 11:1658. [PMID: 38005990 PMCID: PMC10674196 DOI: 10.3390/vaccines11111658] [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: 08/28/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Candida albicans, along with several non-albicans Candida species, comprise a prominent fungal pathogen in humans, leading to candidiasis in various organs. The global impact of candidiasis in terms of disease burden, suffering, and fatalities is alarmingly high, making it a pressing global healthcare concern. Current treatment options rely on antifungal drugs such as azoles, polyenes, and echinocandins but are delimited due to the emergence of drug-resistant strains and associated adverse effects. The current review highlights the striking absence of a licensed antifungal vaccine for human use and the urgent need to shift our focus toward developing an anti-Candida vaccine. A number of factors affect the development of vaccines against fungal infections, including the host, intraspecies and interspecies antigenic variations, and hence, a lack of commercial interest. In addition, individuals with a high risk of fungal infection tend to be immunocompromised, so they are less likely to respond to inactivated or subunit whole organisms. Therefore, it is pertinent to discover newer and novel alternative strategies to develop safe and effective vaccines against fungal infections. This review article provides an overview of current vaccination strategies (live attenuated, whole-cell killed, subunit, conjugate, and oral vaccine), including their preclinical and clinical data on efficacy and safety. We also discuss the mechanisms of immune protection against candidiasis, including the role of innate and adaptive immunity and potential biomarkers of protection. Challenges, solutions, and future directions in vaccine development, namely, exploring novel adjuvants, harnessing the trained immunity, and utilizing immunoinformatics approaches for vaccine design and development, are also discussed. This review concludes with a summary of key findings, their implications for clinical practice and public health, and a call to action for continued investment in candidiasis vaccine research.
Collapse
Affiliation(s)
- Gurpreet Kaur
- Department of Biotechnology, Chandigarh College of Technology (CCT), Chandigarh Group of Colleges (CGC), Landran, Mohali 140307, India
| | - Sonam Chawla
- Department of Biotechnology, Jaypee Institute of Information Technology, Sector 62, Noida 201309, India; (S.C.)
| | - Piyush Kumar
- Department of Biotechnology, Jaypee Institute of Information Technology, Sector 62, Noida 201309, India; (S.C.)
| | - Ritu Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Sector 62, Noida 201309, India; (S.C.)
| |
Collapse
|
24
|
Wang L, Lu H, Jiang Y. Natural Polyketides Act as Promising Antifungal Agents. Biomolecules 2023; 13:1572. [PMID: 38002254 PMCID: PMC10669366 DOI: 10.3390/biom13111572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
Invasive fungal infections present a significant risk to human health. The current arsenal of antifungal drugs is hindered by drug resistance, limited antifungal range, inadequate safety profiles, and low oral bioavailability. Consequently, there is an urgent imperative to develop novel antifungal medications for clinical application. This comprehensive review provides a summary of the antifungal properties and mechanisms exhibited by natural polyketides, encompassing macrolide polyethers, polyether polyketides, xanthone polyketides, linear polyketides, hybrid polyketide non-ribosomal peptides, and pyridine derivatives. Investigating natural polyketide compounds and their derivatives has demonstrated their remarkable efficacy and promising clinical application as antifungal agents.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China;
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China;
| |
Collapse
|