1
|
Kordana N, Johnson A, Quinn K, Obar JJ, Cramer RA. Recent developments in Aspergillus fumigatus research: diversity, drugs, and disease. Microbiol Mol Biol Rev 2025; 89:e0001123. [PMID: 39927770 PMCID: PMC11948498 DOI: 10.1128/mmbr.00011-23] [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] [Indexed: 02/11/2025] Open
Abstract
SUMMARYAdvances in modern medical therapies for many previously intractable human diseases have improved patient outcomes. However, successful disease treatment outcomes are often prevented due to invasive fungal infections caused by the environmental mold Aspergillus fumigatus. As contemporary antifungal therapies have not experienced the same robust advances as other medical therapies, defining mechanisms of A. fumigatus disease initiation and progression remains a critical research priority. To this end, the World Health Organization recently identified A. fumigatus as a research priority human fungal pathogen and the Centers for Disease Control has highlighted the emergence of triazole-resistant A. fumigatus isolates. The expansion in the diversity of host populations susceptible to aspergillosis and the complex and dynamic A. fumigatus genotypic and phenotypic diversity call for a reinvigorated assessment of aspergillosis pathobiological and drug-susceptibility mechanisms. Here, we summarize recent advancements in the field and discuss challenges in our understanding of A. fumigatus heterogeneity and its pathogenesis in diverse host populations.
Collapse
Affiliation(s)
- Nicole Kordana
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Angus Johnson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Katherine Quinn
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Joshua J. Obar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
2
|
Vicente F, Reyes F, Genilloud O. Fungerps: discovery of the glucan synthase inhibitor enfumafungin and development of a new class of antifungal triterpene glycosides. Nat Prod Rep 2024; 41:1835-1845. [PMID: 39552282 DOI: 10.1039/d4np00044g] [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: 11/19/2024]
Abstract
Covering: up to 2024Fungal pathogens are a major threat to public health, with emerging resistance to all three classes of antifungals that are currently available and increased incidence of invasive fungal infections among hospitalized patients. Ibrexafungerp is a semi-synthetic analog of enfumafungin and the first antifungal agent approved in more than 20 years since the launch of caspofungin, the first of echinocandins. This new drug approval was made possible after a long arduous journey lasting 25 years by dedicated and talented medicinal chemists from two companies that undertook tedious atom-by-atom chemical modification of the natural product enfumafungin, a glycosylated fernane-type triterpenoid isolated from the fungus Hormonema carpetanum. This highlight will cover the discovery of enfumafungin, its biosynthesis and the characterisation of its antifungal profile and mode of action that led to the development of ibrexafungerp. We will discuss the challenges encountered during this long preclinical program and the clinical trial validation of this first-in-class oral antifungal approved to treat vulvovaginal candidiasis with an enormous therapeutic potential to treat future major threatening drug-resistant fungal pathogens.
Collapse
Affiliation(s)
| | - Fernando Reyes
- Fundación MEDINA, Avda Conocimiento 34, 18016 Granada, Spain.
| | - Olga Genilloud
- Fundación MEDINA, Avda Conocimiento 34, 18016 Granada, Spain.
| |
Collapse
|
3
|
Amin H, Kantroo HA, Mubarak MM, Bhat SA, Ahmad Z, Bhat KA. Design and synthesis of betulinic acid-dithiocarbamate conjugates as potential antifungal agents against Candida albicans. RSC Adv 2024; 14:38293-38301. [PMID: 39628461 PMCID: PMC11613976 DOI: 10.1039/d4ra05020g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/03/2024] [Indexed: 12/06/2024] Open
Abstract
Diverse betulinic acid-dithiocarbamate conjugates were designed and synthesized via a two-step reaction at room temperature. Among the fourteen dithiocarbamate analogs of betulinic acid, DTC2 demonstrated the best antifungal activity against Candida albicans, with a minimum inhibitory concentration (MIC) of 4 μg mL-1, achieving 99% fungicidal activity at the same concentration. These compounds were found to be ineffective against common Gram-negative and Gram-positive pathogens, suggesting their specificity to fungi. Furthermore, DTC2 exhibited synergistic effects with the antifungal drugs fluconazole and nystatin, resulting in a significant decrease in MIC by 64 and 16 folds, respectively, when co-administered. Notably, the molecule also hindered hyphae formation in Candida albicans, thereby reducing its pathogenicity. Furthermore, it displayed time- and concentration-dependent kill kinetics, sterilizing C. albicans within 8 hours at 8× MIC. Additionally, DTC2 exhibits greater efficacy against β-carbonic anhydrase with better docking scores and binding patterns than ethoxyzolamide, a well-known inhibitor of β-carbonic anhydrase.
Collapse
Affiliation(s)
- Henna Amin
- Bioorganic Chemistry Division, Indian Institute of Integrative Medicine (CSIR) Srinagar J&K 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Hadiya Amin Kantroo
- Clinical Microbiology and PK-PD Division, CSIR Indian Institute of Integrative Medicine Srinagar J&K 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Mohamad Mosa Mubarak
- Clinical Microbiology and PK-PD Division, CSIR Indian Institute of Integrative Medicine Srinagar J&K 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Showkat Ahmad Bhat
- Bioorganic Chemistry Division, Indian Institute of Integrative Medicine (CSIR) Srinagar J&K 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Zahoor Ahmad
- Clinical Microbiology and PK-PD Division, CSIR Indian Institute of Integrative Medicine Srinagar J&K 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Khursheed Ahmad Bhat
- Bioorganic Chemistry Division, Indian Institute of Integrative Medicine (CSIR) Srinagar J&K 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| |
Collapse
|
4
|
Antunes D, Domingues R, Cruz-Almeida M, Rodrigues L, Borges O, Carvalho A, Casadevall A, Fernandes C, Gonçalves T. Cell wall nanoparticles from hyphae of Alternaria infectoria grown with caspofungin, nikkomycin, or pyroquilon trigger different activation profiles in macrophages. Microbiol Spectr 2024; 12:e0064524. [PMID: 39329485 PMCID: PMC11537108 DOI: 10.1128/spectrum.00645-24] [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: 03/11/2024] [Accepted: 08/02/2024] [Indexed: 09/28/2024] Open
Abstract
Alternaria infectoria causes opportunistic human infections and is a source of allergens leading to respiratory allergies. In this work, we prepared cell wall nanoparticles (CWNPs) as a novel approach to study macrophage immunomodulation by fungal hyphal cell walls. A. infectoria was grown in the presence of caspofungin, an inhibitor of β(1,3)-glucan synthesis; nikkomycin Z, an inhibitor of chitin synthases; and pyroquilon, an inhibitor of dihydroxynaphthalene (DHN)-melanin synthesis. Distinct CWNPs were obtained from these cultures, referred to as casCWNPs, nkCWNPs, and pyrCWNPs, respectively. CWNPs are round-shaped particles with a diameter of 70-200 nm diameter particles that when added to macrophages are taken up by membrane ruffling. CWNPs with no DHN-melanin and more glucan (pyrCWNPs) caused early macrophage activation and lowest viability, with the cells exhibiting ultrastructural modifications such as higher vacuolization and formation of autophagy-like structures. CasCWNPs promoted the highest tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β) increase, also resulting in the release of partially degraded chitin, an aspect never observed in macrophage-like cells and fungi. After 6 h of interaction with CWNPs, only half were viable, except with control CWNPs. Overall, this work indicates that compounds that modify the fungal cell wall led to CWNPs with new properties that may have implications for the effects of drugs during antifungal therapy. CWNPs provide a new tool to study the interaction of hyphal fungal cell wall components with phagocytic cells and enable to show how the modification of cell wall components in A. infectoria can modulate the response by macrophages.IMPORTANCEAlternaria species are ubiquitous environmental fungi to which the human host can continuously be exposed, through the inhalation of fungal spores but also of fragments of hyphae, from desegregated mycelia. These fungi are involved in hypersensitization and severe respiratory allergies, such as asthma, and can cause opportunistic infections in immunodepressed human host leading to severe disease. The first fungal structures to interact with the host cells are the cell wall components, and their modulation leads to differential immune responses. Here, we show that fungal cells grown with cell wall inhibitors led to cell wall nanoparticles with new properties in their interaction with macrophages. With this strategy, we overcame the limitation of in vitro assays interacting with filamentous fungi and showed that the absence of DNH-melanin leads to higher virulence, while caspofungin leads to cells walls that trigger higher hydrolysis of chitin and higher production of cytokines.
Collapse
Affiliation(s)
- Daniela Antunes
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rita Domingues
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana Cruz-Almeida
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Olga Borges
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Univ Coimbra, FFUC—Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Chantal Fernandes
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Teresa Gonçalves
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Univ Coimbra, FMUC—Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Sun C, Li Y, Kidd JM, Han J, Ding L, May AE, Zhou L, Liu Q. Characterization of a New Hsp110 Inhibitor as a Potential Antifungal. J Fungi (Basel) 2024; 10:732. [PMID: 39590652 PMCID: PMC11595998 DOI: 10.3390/jof10110732] [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: 08/21/2024] [Revised: 10/03/2024] [Accepted: 10/13/2024] [Indexed: 11/28/2024] Open
Abstract
Fungal infections present a significant global health challenge, prompting ongoing research to discover innovative antifungal agents. The 110 kDa heat shock proteins (Hsp110s) are molecular chaperones essential for maintaining cellular protein homeostasis in eukaryotes. Fungal Hsp110s have emerged as a promising target for innovative antifungal strategies. Notably, 2H stands out as a promising candidate in the endeavor to target Hsp110s and combat fungal infections. Our study reveals that 2H exhibits broad-spectrum antifungal activity, effectively disrupting the in vitro chaperone activity of Hsp110 from Candida auris and inhibiting the growth of Cryptococcus neoformans. Pharmacokinetic analysis indicates that oral administration of 2H may offer enhanced efficacy compared to intravenous delivery, emphasizing the importance of optimizing the AUC/MIC ratio for advancing its clinical therapy.
Collapse
Affiliation(s)
- Cancan Sun
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Yi Li
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Justin M. Kidd
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jizhong Han
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Liangliang Ding
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Aaron E. May
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Lei Zhou
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Qinglian Liu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
6
|
Guilloux K, Hegde P, Wong SSW, Aimanianda V, Bayry J, Latgé JP. Comparative Analysis of the Aspergillus fumigatus Cell Wall Modification and Ensuing Human Dendritic Cell Responses by β-(1,3)-Glucan Synthase Inhibitors-Caspofungin and Enfumafungin. Mycopathologia 2024; 189:86. [PMID: 39302505 DOI: 10.1007/s11046-024-00894-7] [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: 07/04/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Caspofungin, a lipopeptide, is an antifungal drug that belong to the class of echinocandin. It inhibits fungal cell wall β-(1,3)-glucan synthase activity and is the second-line of drug for invasive aspergillosis, a fatal infection caused mainly by Aspergillus fumigatus. On the other hand, Enfumafungin is a natural triterpene glycoside also with a β-(1,3)-glucan synthase inhibitory activity and reported to have antifungal potential. In the present study, we compared the growth as well as modifications in the A. fumigatus cell wall upon treatment with Caspofungin or Enfumafungin, consequentially their immunomodulatory capacity on human dendritic cells. Caspofungin initially inhibited the growth of A. fumigatus, but the effect was lost over time. By contrast, Enfumafungin inhibited this fungal growth for the duration investigated. Both Caspofungin and Enfumafungin caused a decrease in the cell wall β-(1,3)-glucan content with a compensatory increase in the chitin, and to a minor extent they also affected cell wall galactose content. Treatment with these two antifungals did not result in the exposure of β-(1,3)-glucan on A. fumigatus mycelial surface. Enzymatic digestion suggested a modification of β-(1,3)-glucan structure, specifically its branching, upon Enfumafungin treatment. While there was no difference in the immunostimulatory capacity of antifungal treated A. fumigatus conidia, alkali soluble-fractions from Caspofungin treated mycelia weakly stimulated the dendritic cells, possibly due to an increased content of immunosuppressive polysaccharide galactosaminogalactan. Overall, we demonstrate a novel mechanism that Enfumafungin not only inhibits β-(1,3)-glucan synthase activity, but also causes modifications in the structure of β-(1,3)-glucan in the A. fumigatus cell wall.
Collapse
Affiliation(s)
- Karine Guilloux
- Unité des Aspergillus, Institut Pasteur, 75015, Paris, France
| | - Pushpa Hegde
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université Paris Cité, 75006, Paris, France
| | - Sarah Sze Wah Wong
- Unité des Aspergillus, Institut Pasteur, 75015, Paris, France
- Immunobiologie d'Aspergillus, Institut Pasteur, Paris, France
| | - Vishukumar Aimanianda
- Unité des Aspergillus, Institut Pasteur, 75015, Paris, France
- Immunobiologie d'Aspergillus, Institut Pasteur, Paris, France
| | - Jagadeesh Bayry
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université Paris Cité, 75006, Paris, France.
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, 678623, India.
| | - Jean-Paul Latgé
- Unité des Aspergillus, Institut Pasteur, 75015, Paris, France.
- IMBB-FORTH, Heraklion, Greece.
| |
Collapse
|
7
|
Gao Y, Cao Q, Xiao Y, Wu Y, Ding L, Huang H, Li Y, Yang J, Meng L. The progress and future of the treatment of Candida albicans infections based on nanotechnology. J Nanobiotechnology 2024; 22:568. [PMID: 39285480 PMCID: PMC11406819 DOI: 10.1186/s12951-024-02841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024] Open
Abstract
Systemic infection with Candida albicans poses a significant risk for people with weakened immune systems and carries a mortality rate of up to 60%. However, current therapeutic options have several limitations, including increasing drug tolerance, notable off-target effects, and severe adverse reactions. Over the past four decades, the progress in developing drugs to treat Candida albicans infections has been sluggish. This comprehensive review addresses the limitations of existing drugs and summarizes the efforts made toward redesigning and innovating existing or novel drugs through nanotechnology. The discussion explores the potential applications of nanomedicine in Candida albicans infections from four perspectives: nano-preparations for anti-biofilm therapy, innovative formulations of "old drugs" targeting the cell membrane and cell wall, reverse drug resistance therapy targeting subcellular organelles, and virulence deprivation therapy leveraging the unique polymorphism of Candida albicans. These therapeutic approaches are promising to address the above challenges and enhance the efficiency of drug development for Candida albicans infections. By harnessing nano-preparation technology to transform existing and preclinical drugs, novel therapeutic targets will be uncovered, providing effective solutions and broader horizons to improve patient survival rates.
Collapse
Affiliation(s)
- Yang Gao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Qinyan Cao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yuyang Xiao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yue Wu
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Liang Ding
- Nanjing Stomatological Hospital, Nanjing, 210008, China
| | - He Huang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yanan Li
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Jingpeng Yang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Lingtong Meng
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
8
|
Liu CY, Zhang L, Liu SX, Lu YF, Li C, Pei YH. A review of the fernane-type triterpenoids as anti-fungal drugs. Front Pharmacol 2024; 15:1447450. [PMID: 39234110 PMCID: PMC11371599 DOI: 10.3389/fphar.2024.1447450] [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: 06/11/2024] [Accepted: 08/08/2024] [Indexed: 09/06/2024] Open
Abstract
Human fungal pathogens could cause a broad plethora of infections in both the immunocompetent and immunocompromised host. Fungal infections have become important causes of morbidity and mortality in recent years, the current arsenal of anti-fungal therapies was restricted. Ibrexafungerp was a novel, highly bioavailable glucan synthase inhibitor formulated for both intravenous and oral administration being developed by Scynexis; it was also the first novel anti-fungal drug class approved in more than 20 years. Ibrexafungerp was one semi-synthetic derivative of enfumafungin, a natural product isolated from fungi. This review reported the discovery of enfumafungin and ibrexafungerp, their anti-fungal mechanism, summed up 63 fernane-type triterpenoids from natural products, including 49 from plants, 9 from fungi and 5 from lichen. In addition, the review summarized the progress of enzymes responsible for the biosynthesis of type II fernane triterpenoid (enfumafungin skeleton) and type I fernane triterpenoid (polytolypin skeleton). The good example kept our confidence up for searching for new leading compounds and discovering drugs from fungi.
Collapse
Affiliation(s)
- Chun-Yue Liu
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
- Department of Chemistry, College of Pharmacy, Harbin Medical University, Daqing, China
| | - Lu Zhang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Si-Xuan Liu
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yong-Fu Lu
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chang Li
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yue-Hu Pei
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Khalifa HO, Oreiby A, Abdelhamid MAA, Ki MR, Pack SP. Biomimetic Antifungal Materials: Countering the Challenge of Multidrug-Resistant Fungi. Biomimetics (Basel) 2024; 9:425. [PMID: 39056866 PMCID: PMC11274442 DOI: 10.3390/biomimetics9070425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
In light of rising public health threats like antifungal and antimicrobial resistance, alongside the slowdown in new antimicrobial development, biomimetics have shown promise as therapeutic agents. Multidrug-resistant fungi pose significant challenges as they quickly develop resistance, making traditional antifungals less effective. Developing new antifungals is also complicated by the need to target eukaryotic cells without harming the host. This review examines biomimetic antifungal materials that mimic natural biological mechanisms for targeted and efficient action. It covers a range of agents, including antifungal peptides, alginate-based antifungals, chitosan derivatives, nanoparticles, plant-derived polyphenols, and probiotic bacteria. These agents work through mechanisms such as disrupting cell membranes, generating reactive oxygen species, and inhibiting essential fungal processes. Despite their potential, challenges remain in terms of ensuring biocompatibility, optimizing delivery, and overcoming potential resistance. Production scalability and economic viability are also concerns. Future research should enhance the stability and efficacy of these materials, integrate multifunctional approaches, and develop sophisticated delivery systems. Interdisciplinary efforts are needed to understand interactions between these materials, fungal cells, and the host environment. Long-term health and environmental impacts, fungal resistance mechanisms, and standardized testing protocols require further study. In conclusion, while biomimetic antifungal materials represent a revolutionary approach to combating multidrug-resistant fungi, extensive research and development are needed to fully realize their potential.
Collapse
Affiliation(s)
- Hazim O. Khalifa
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 1555, United Arab Emirates
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Atef Oreiby
- Department of Animal Medicine, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Mohamed A. A. Abdelhamid
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea; (M.A.A.A.); (M.-R.K.)
- Department of Botany and Microbiology, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Mi-Ran Ki
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea; (M.A.A.A.); (M.-R.K.)
- Institute of Industrial Technology, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea; (M.A.A.A.); (M.-R.K.)
| |
Collapse
|
10
|
Manzano JAH, Brogi S, Calderone V, Macabeo APG, Austriaco N. Globospiramine Exhibits Inhibitory and Fungicidal Effects against Candida albicans via Apoptotic Mechanisms. Biomolecules 2024; 14:610. [PMID: 38927014 PMCID: PMC11201426 DOI: 10.3390/biom14060610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Candidiasis is considered an emerging public health concern because of the occurrence of drug-resistant Candida strains and the lack of an available structurally diverse antifungal drug armamentarium. The indole alkaloid globospiramine from the anticandidal Philippine medicinal plant Voacanga globosa exhibits a variety of biological activities; however, its antifungal properties remain to be explored. In this study, we report the in vitro anticandidal activities of globospiramine against two clinically relevant Candida species (C. albicans and C. tropicalis) and the exploration of its possible target proteins using in silico methods. Thus, the colony-forming unit (CFU) viability assay revealed time- and concentration-dependent anticandidal effects of the alkaloid along with a decrease in the number of viable CFUs by almost 50% at 60 min after treatment. The results of the MIC and MFC assays indicated inhibitory and fungicidal effects of globospiramine against C. albicans (MIC = 8 µg/mL; MFC = 8 µg/mL) and potential fungistatic effects against C. tropicalis at lower concentrations (MIC = 4 µg/mL; MFC > 64 µg/mL). The FAM-FLICA poly-caspase assay showed metacaspase activation in C. albicans cells at concentrations of 16 and 8 µg/mL, which agreed well with the MIC and MFC values. Molecular docking and molecular dynamics simulation experiments suggested globospiramine to bind strongly with 1,3-β-glucan synthase and Als3 adhesin-enzymes indirectly involved in apoptosis-driven candidal inhibition.
Collapse
Affiliation(s)
- Joe Anthony H. Manzano
- The Graduate School, University of Santo Tomas, España Blvd., Manila 1015, Philippines;
- UST Laboratories for Vaccine Science, Molecular Biology and Biotechnology, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines;
- Laboratory for Organic Reactivity, Discovery, and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Allan Patrick G. Macabeo
- Laboratory for Organic Reactivity, Discovery, and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines
- Department of Chemistry, College of Science, University of Santo Tomas, España Blvd., Manila 1015, Philippines
| | - Nicanor Austriaco
- UST Laboratories for Vaccine Science, Molecular Biology and Biotechnology, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines;
- Department of Biological Sciences, College of Science, University of Santo Tomas, España Blvd., Manila 1015, Philippines
| |
Collapse
|
11
|
Kim Y. Fisetin-Mediated Perturbations of Membrane Permeability and Intracellular pH in Candida albicans. J Microbiol Biotechnol 2024; 34:783-794. [PMID: 38213272 PMCID: PMC11091701 DOI: 10.4014/jmb.2311.11027] [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: 11/20/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024]
Abstract
The antifungal activity of fisetin against Candida albicans is explored, elucidating a mechanism centered on membrane permeabilization and ensuing disruption of pH homeostasis. The Minimum Inhibitory Concentration (MIC) of fisetin, indicative of its interaction with the fungal membrane, increases in the presence of ergosterol. Hoechst 33342 and propidium-iodide staining reveal substantial propidium-iodide accumulation in fisetin-treated C. albicans cells at their MIC, with crystal violet uptake assays confirming fisetin-induced membrane permeabilization. Leakage analysis demonstrates a significant release of DNA and proteins in fisetin-treated cells compared to controls, underscoring the antifungal effect through membrane disruption. Green fluorescence, evident in both the cytoplasm and vacuoles of fisetin-treated cells under BCECF, AM staining, stands in contrast to controls where only acidic vacuoles exhibit staining. Ratiometric pH measurements using BCECF, AM reveal a noteworthy reduction in intracellular pH in fisetin-treated cells, emphasizing its impact on pH homeostasis. DiBAC4(3) uptake assays demonstrate membrane hyperpolarization in fisetin-treated cells, suggesting potential disruptions in ion flux and cellular homeostasis. These results provide comprehensive insights into the antifungal mechanisms of fisetin, positioning it as a promising therapeutic agent against Candida infections.
Collapse
Affiliation(s)
- Younhee Kim
- Department of Korean Medicine, Semyung University, Jecheon 27136, Republic of Korea
| |
Collapse
|
12
|
Kumar V, Huang J, Dong Y, Hao GF. Targeting Fks1 proteins for novel antifungal drug discovery. Trends Pharmacol Sci 2024; 45:366-384. [PMID: 38493014 DOI: 10.1016/j.tips.2024.02.007] [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: 01/09/2024] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
Fungal infections are a major threat to human health. The limited availability of antifungal drugs, the emergence of drug resistance, and a growing susceptible population highlight the critical need for novel antifungal agents. The enzymes involved in fungal cell wall synthesis offer potential targets for antifungal drug development. Recent studies have enhanced our focus on the enzyme Fks1, which synthesizes β-1,3-glucan, a critical component of the cell wall. These studies provide a deeper understanding of Fks1's function in cell wall biosynthesis, pathogenicity, structural biology, evolutionary conservation across fungi, and interaction with current antifungal drugs. Here, we discuss the role of Fks1 in the survival and adaptation of fungi, guided by insights from evolutionary and structural analyses. Furthermore, we delve into the dynamics of Fks1 modulation with novel antifungal strategies and assess its potential as an antifungal drug target.
Collapse
Affiliation(s)
- Vinit Kumar
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China; BMLT, Markham College of Commerce, Vinoba Bhave University, Hazaribagh, Jharkhand 825301, India
| | - Juan Huang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Yawen Dong
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China.
| | - Ge-Fei Hao
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China; National Key Laboratory of Green Pesticide, Central China Normal University, Wuhan 430079, PR China.
| |
Collapse
|
13
|
Yadav A, Sah SK, Perlin DS, Rustchenko E. Candida albicans Genes Modulating Echinocandin Susceptibility of Caspofungin-Adapted Mutants Are Constitutively Expressed in Clinical Isolates with Intermediate or Full Resistance to Echinocandins. J Fungi (Basel) 2024; 10:224. [PMID: 38535232 PMCID: PMC10971431 DOI: 10.3390/jof10030224] [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: 02/07/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 11/12/2024] Open
Abstract
The opportunistic fungus Candida albicans is the leading cause of invasive candidiasis in immune-compromised individuals. Drugs from the echinocandin (ECN) class, including caspofungin, are used as a first line of therapy against invasive candidiasis. The only known mechanism of clinical resistance to ECNs is point mutations in the FKS1 gene, which encodes the drug target. However, many clinical isolates developed decreased ECN susceptibilities in the absence of resistance-associated FKS1 mutations. We have identified 15 C. albicans genes that contribute to decreased drug susceptibility. We explored the expression of these 15 genes in clinical isolates with different levels of ECN susceptibility. We found that these 15 genes are expressed in clinical isolates with or without FKS1 mutations, including those strains that are less susceptible to ECNs. In addition, FKS1 expression was increased in such less susceptible isolates compared to highly susceptible isolates. Similarities of gene expression patterns between isolates with decreased ECN susceptibilities in the absence of FKS1 mutations and clinically resistant isolates with mutations in FKS1 suggest that clinical isolates with decreased ECN susceptibilities may be a precursor to development of resistance.
Collapse
Affiliation(s)
- Anshuman Yadav
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.Y.); (S.K.S.)
| | - Sudisht K. Sah
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.Y.); (S.K.S.)
| | - David S. Perlin
- Hackensack Meridian School of Medicine, Center for Discovery and Innovation, Nutley, NJ 07110, USA;
| | - Elena Rustchenko
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.Y.); (S.K.S.)
| |
Collapse
|
14
|
Boakye-Yiadom E, Odoom A, Osman AH, Ntim OK, Kotey FCN, Ocansey BK, Donkor ES. Fungal Infections, Treatment and Antifungal Resistance: The Sub-Saharan African Context. Ther Adv Infect Dis 2024; 11:20499361241297525. [PMID: 39544852 PMCID: PMC11562003 DOI: 10.1177/20499361241297525] [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: 01/17/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024] Open
Abstract
Fungal pathogens cause a wide range of infections in humans, from superficial to disfiguring, allergic syndromes, and life-threatening invasive infections, affecting over a billion individuals globally. With an estimated 1.5 million deaths annually attributable to them, fungal pathogens are a major cause of mortality in humans, especially people with underlying immunosuppression. The continuous increase in the population of individuals at risk of fungal infections in sub-Saharan Africa, such as HIV patients, tuberculosis patients, intensive care patients, patients with haematological malignancies, transplant (haematopoietic stem cell and organ) recipients and the growing global threat of multidrug-resistant fungal strains, raise the need for an appreciation of the region's perspective on antifungal usage and resistance. In addition, the unavailability of recently introduced novel antifungal drugs in sub-Saharan Africa further calls for regular evaluation of resistance to antifungal agents in these settings. This is critical for ensuring appropriate and optimal use of the limited available arsenal to minimise antifungal resistance. This review, therefore, elaborates on the multifaceted nature of fungal resistance to the available antifungal drugs on the market and further provides insights into the prevalence of fungal infections and the use of antifungal agents in sub-Saharan Africa.
Collapse
Affiliation(s)
- Emily Boakye-Yiadom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
- Department of Microbiology and Immunology, University of Health and Allied Sciences, Ho, Ghana
| | - Alex Odoom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Onyansaniba K. Ntim
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Fleischer C. N. Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Bright K. Ocansey
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, P.O. Box KB 4236, Ghana
| |
Collapse
|
15
|
Ostroumova OS, Efimova SS. Lipid-Centric Approaches in Combating Infectious Diseases: Antibacterials, Antifungals and Antivirals with Lipid-Associated Mechanisms of Action. Antibiotics (Basel) 2023; 12:1716. [PMID: 38136750 PMCID: PMC10741038 DOI: 10.3390/antibiotics12121716] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
One of the global challenges of the 21st century is the increase in mortality from infectious diseases against the backdrop of the spread of antibiotic-resistant pathogenic microorganisms. In this regard, it is worth targeting antibacterials towards the membranes of pathogens that are quite conservative and not amenable to elimination. This review is an attempt to critically analyze the possibilities of targeting antimicrobial agents towards enzymes involved in pathogen lipid biosynthesis or towards bacterial, fungal, and viral lipid membranes, to increase the permeability via pore formation and to modulate the membranes' properties in a manner that makes them incompatible with the pathogen's life cycle. This review discusses the advantages and disadvantages of each approach in the search for highly effective but nontoxic antimicrobial agents. Examples of compounds with a proven molecular mechanism of action are presented, and the types of the most promising pharmacophores for further research and the improvement of the characteristics of antibiotics are discussed. The strategies that pathogens use for survival in terms of modulating the lipid composition and physical properties of the membrane, achieving a balance between resistance to antibiotics and the ability to facilitate all necessary transport and signaling processes, are also considered.
Collapse
Affiliation(s)
- Olga S. Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia;
| | | |
Collapse
|
16
|
Cheng Z, Wu W, Liu Y, Chen S, Li H, Yang X, Zhu X, Chen X, Yan L, Chu Z, Sun P. Natural Enfumafungin Analogues from Hormonema carpetanum and Their Antifungal Activities. JOURNAL OF NATURAL PRODUCTS 2023; 86:2407-2413. [PMID: 37853717 DOI: 10.1021/acs.jnatprod.3c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Ibrexafungerp, an inhibitor of fungal β-(1,3)-d-glucan synthase, represents the first new class of antifungals to be approved in the last 20 years. Ibrexafungerp is a semisynthetic derivative of the naturally occurring triterpene glycoside enfumafungin. In order to search for new analogues of enfumafungin and to probe its biosynthesis, we undertook a reinvestigation of Hormonema carpetanum, which led to the isolation of two new analogues, enfumafungins B and C, together with enfumafungin. Due to the presence of a hemiacetal moiety in the structure, the enfumafungins appear as a mixture of two interconverting epimers during both the purification process and NMR data acquisition. The structure elucidation, including the differentiation of 25S* and 25R* epimers, was completed by combined analyses of NMR and MS spectroscopic data. The discovery of enfumafungins B and C may have implications for enfumafungin biosynthesis. The antifungal activity of enfumafungins B and C was significantly lower than that of enfumafungin, suggesting that the C-2 substituents and the C-19 carboxy acid are important for activity. Molecular docking simulations revealed significant hydrogen bond interactions between enfumafungins and β-(1,3)-d-glucan synthase, which may be useful for developing new antifungal agents.
Collapse
Affiliation(s)
- Zhi Cheng
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Wei Wu
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Yu Liu
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Shuo Chen
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Hongji Li
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Xingchi Yang
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Xiaofan Zhu
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Xuxiang Chen
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Lan Yan
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Zhiyong Chu
- Navy Medical Research Institute, Naval Medical University, 880 Xiangyin Road, Shanghai 200433, China
| | - Peng Sun
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, China
| |
Collapse
|
17
|
Khattak AA, Qian J, Xu L, Tomah AA, Ibrahim E, Khan MZI, Ahmed T, Hatamleh AA, Al-Dosary MA, Ali HM, Li B. Precision drug design against Acidovorax oryzae: leveraging bioinformatics to combat rice brown stripe disease. Front Cell Infect Microbiol 2023; 13:1225285. [PMID: 37886665 PMCID: PMC10598866 DOI: 10.3389/fcimb.2023.1225285] [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: 05/25/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
Bacterial brown stripe disease caused by Acidovorax oryzae is a major threat to crop yields, and the current reliance on pesticides for control is unsustainable due to environmental pollution and resistance. To address this, bacterial-based ligands have been explored as a potential treatment solution. In this study, we developed a protein-protein interaction (PPI) network for A. oryzae by utilizing shared differentially expressed genes (DEGs) and the STRING database. Using a maximal clique centrality (MCC) approach through CytoHubba and Network Analyzer, we identified hub genes within the PPI network. We then analyzed the genomic data of the top 10 proteins, and further narrowed them down to 2 proteins by utilizing betweenness, closeness, degree, and eigenvector studies. Finally, we used molecular docking to screen 100 compounds against the final two proteins (guaA and metG), and Enfumafungin was selected as a potential treatment for bacterial resistance caused by A. oryzae based on their binding affinity and interaction energy. Our approach demonstrates the potential of utilizing bioinformatics and molecular docking to identify novel drug candidates for precision treatment of bacterial brown stripe disease caused by A. oryzae, paving the way for more targeted and sustainable control strategies. The efficacy of Enfumafungin in inhibiting the growth of A. oryzae strain RS-1 was investigated through both computational and wet lab methods. The models of the protein were built using the Swiss model, and their accuracy was confirmed via a Ramachandran plot. Additionally, Enfumafungin demonstrated potent inhibitory action against the bacterial strain, with an MIC of 100 µg/mL, reducing OD600 values by up to 91%. The effectiveness of Enfumafungin was further evidenced through agar well diffusion assays, which exhibited the highest zone of inhibition at 1.42 cm when the concentration of Enfumafungin was at 100 µg/mL. Moreover, Enfumafungin was also able to effectively reduce the biofilm of A. oryzae RS-1 in a concentration-dependent manner. The swarming motility of A. oryzae RS-1 was also found to be significantly inhibited by Enfumafungin. Further validation through TEM observation revealed that bacterial cells exposed to Enfumafungin displayed mostly red fluorescence, indicating destruction of the bacterial cell membrane.
Collapse
Affiliation(s)
- Arif Ali Khattak
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Jiahui Qian
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Lihui Xu
- Institute of Eco-Environmental Protection, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Ali Athafah Tomah
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Plant Protection, College of Agriculture, University of Misan, AL-Amarah, Iraq
| | - Ezzeldin Ibrahim
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Department of Vegetable Diseases Research, Plant Pathology Research Institute, Agriculture Research Centre, Giza, Egypt
| | | | - Temoor Ahmed
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Xianghu Laboratory, Hangzhou, China
| | - Ashraf Atef Hatamleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Hayssam M. Ali
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Bin Li
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Dhawale S, Pandit M, Thete K, Ighe D, Gawale S, Bhosle P, Lokwani DK. In silico approach towards polyphenols as targeting glucosamine-6-phosphate synthase for Candida albicans. J Biomol Struct Dyn 2023; 41:12038-12054. [PMID: 36629053 DOI: 10.1080/07391102.2022.2164797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023]
Abstract
Candida albicans is one of the most common species of fungus with life-threatening systemic infections and a high mortality rate. The outer cell wall layer of C. albicans is packed with mannoproteins and glycosylated polysaccharide moieties that play an essential role in the interaction with host cells and tissues. The glucosamine-6-phosphate synthase enzyme produces N-acetylglucosamine, which is a crucial chemical component of the cell wall of Candida albicans. Collectively, these components are essential to maintain the cell shape and for infection. So, its disruption can have serious effects on cell growth and morphology, resulting in cell death. Hence, it is considered a good antifungal target. In this study, we have performed an in silico approach to analyze the inhibitory potential of some polyphenols obtained from plants. Those can be considered important in targeting against the enzyme glucosamine-6-phosphate synthase (PDB-2VF5). The results of the study revealed that the binding affinity of complexes theaflavin and 3-o-malonylglucoside have significant docking scores and binding free energy followed by significant ADMET parameters that predict the drug-likeness property and toxicity of polyphenols as potential ligands. A molecular dynamic simulation was used to test the validity of the docking scores, and it showed that the complex remained stable during the period of the simulation, which ranged from 0 to 100 ns. Theaflavins and 3-o-malonylglucoside may be effective against Candida albicans using a computer-aided drug design methodology that will further enable researchers for future in vitro and in vivo studies, according to our in silico study.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sachin Dhawale
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical education and research, Aurangabad, Maharashtra, India
| | - Madhuri Pandit
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical education and research, Aurangabad, Maharashtra, India
| | - Kanchan Thete
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical education and research, Aurangabad, Maharashtra, India
| | - Dnyaneshwari Ighe
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical education and research, Aurangabad, Maharashtra, India
| | - Sachin Gawale
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical education and research, Aurangabad, Maharashtra, India
| | - Pallavi Bhosle
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical education and research, Aurangabad, Maharashtra, India
| | | |
Collapse
|
19
|
Ghasemi R, Lotfali E, Rezaei K, Madinehzad SA, Tafti MF, Aliabadi N, Kouhsari E, Fattahi M. Meyerozyma guilliermondii species complex: review of current epidemiology, antifungal resistance, and mechanisms. Braz J Microbiol 2022; 53:1761-1779. [PMID: 36306113 PMCID: PMC9679122 DOI: 10.1007/s42770-022-00813-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 06/30/2022] [Indexed: 01/13/2023] Open
Abstract
Meyerozyma guilliermondii has been accepted as a complex composed of Meyerozyma guilliermondii, Meyerozyma carpophila, and Meyerozyma caribbica. M. guilliermondii is a saprophyte detected on human mucosa and skin. It can lead to serious infections in patients with risk factors like chemotherapy, immunodeficiency, gastrointestinal or cardiovascular surgery, and oncology disorders. Most deaths related to M. guilliermondii infections occur in individuals with malignancy. In recent decades, incidence of M. guilliermondii infections is increased. Sensitivity of this microorganism to conventional antifungals (e.g., amphotericin B, fluconazole, micafungin and anidulafungin) was reduced. Prophylactic and empirical uses of these drugs are linked to elevated minimal inhibitory concentrations (MICs) of M. guilliermondii. Drug resistance has concerned many researchers across the world. They are attempting to discover appropriate solution to combat this challenge. This study reviews the most important mechanisms of resistance to antifungals developed by in M. guilliermondii species complex.
Collapse
Affiliation(s)
- Reza Ghasemi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Lotfali
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rezaei
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ataollah Madinehzad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Falah Tafti
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikta Aliabadi
- Microbiology Department Islamic, Azad University Tehran Branch, Tehran, Iran
| | - Ebrahim Kouhsari
- Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahsa Fattahi
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
21
|
Mapook A, Hyde KD, Hassan K, Kemkuignou BM, Čmoková A, Surup F, Kuhnert E, Paomephan P, Cheng T, de Hoog S, Song Y, Jayawardena RS, Al-Hatmi AMS, Mahmoudi T, Ponts N, Studt-Reinhold L, Richard-Forget F, Chethana KWT, Harishchandra DL, Mortimer PE, Li H, Lumyong S, Aiduang W, Kumla J, Suwannarach N, Bhunjun CS, Yu FM, Zhao Q, Schaefer D, Stadler M. Ten decadal advances in fungal biology leading towards human well-being. FUNGAL DIVERS 2022; 116:547-614. [PMID: 36123995 PMCID: PMC9476466 DOI: 10.1007/s13225-022-00510-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/28/2022] [Indexed: 11/04/2022]
Abstract
Fungi are an understudied resource possessing huge potential for developing products that can greatly improve human well-being. In the current paper, we highlight some important discoveries and developments in applied mycology and interdisciplinary Life Science research. These examples concern recently introduced drugs for the treatment of infections and neurological diseases; application of -OMICS techniques and genetic tools in medical mycology and the regulation of mycotoxin production; as well as some highlights of mushroom cultivaton in Asia. Examples for new diagnostic tools in medical mycology and the exploitation of new candidates for therapeutic drugs, are also given. In addition, two entries illustrating the latest developments in the use of fungi for biodegradation and fungal biomaterial production are provided. Some other areas where there have been and/or will be significant developments are also included. It is our hope that this paper will help realise the importance of fungi as a potential industrial resource and see the next two decades bring forward many new fungal and fungus-derived products.
Collapse
Affiliation(s)
- Ausana Mapook
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
| | - Kevin D. Hyde
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- School of Science, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan China
- Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, 50200 Thailand
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200 Thailand
- Innovative Institute of Plant Health, Zhongkai University of Agriculture and Engineering, Haizhu District, Guangzhou, 510225 China
| | - Khadija Hassan
- Department Microbial Drugs, Helmholtz Centre for Infection Research (HZI), and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstrasse 7, 38124 Brunswick, Germany
| | - Blondelle Matio Kemkuignou
- Department Microbial Drugs, Helmholtz Centre for Infection Research (HZI), and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstrasse 7, 38124 Brunswick, Germany
| | - Adéla Čmoková
- Laboratory of Fungal Genetics and Metabolism, Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Frank Surup
- Department Microbial Drugs, Helmholtz Centre for Infection Research (HZI), and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstrasse 7, 38124 Brunswick, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Brunswick, Germany
| | - Eric Kuhnert
- Centre of Biomolecular Drug Research (BMWZ), Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 38, 30167 Hannover, Germany
| | - Pathompong Paomephan
- Department Microbial Drugs, Helmholtz Centre for Infection Research (HZI), and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstrasse 7, 38124 Brunswick, Germany
- Department of Biotechnology, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi, Bangkok, 10400 Thailand
| | - Tian Cheng
- Department Microbial Drugs, Helmholtz Centre for Infection Research (HZI), and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstrasse 7, 38124 Brunswick, Germany
- Laboratory of Fungal Genetics and Metabolism, Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Sybren de Hoog
- Center of Expertise in Mycology, Radboud University Medical Center / Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
- Microbiology, Parasitology and Pathology Graduate Program, Federal University of Paraná, Curitiba, Brazil
| | - Yinggai Song
- Department of Dermatology, Peking University First Hospital, Peking University, Beijing, China
| | - Ruvishika S. Jayawardena
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- School of Science, Mae Fah Luang University, Chiang Rai, 57100 Thailand
| | - Abdullah M. S. Al-Hatmi
- Center of Expertise in Mycology, Radboud University Medical Center / Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nadia Ponts
- INRAE, UR1264 Mycology and Food Safety (MycSA), 33882 Villenave d’Ornon, France
| | - Lena Studt-Reinhold
- Department of Applied Genetics and Cell Biology, Institute of Microbial Genetics, University of Natural Resources and Life Sciences, Vienna (BOKU), Tulln an der Donau, Austria
| | | | - K. W. Thilini Chethana
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- School of Science, Mae Fah Luang University, Chiang Rai, 57100 Thailand
| | - Dulanjalee L. Harishchandra
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- School of Science, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- Beijing Key Laboratory of Environment Friendly Management on Fruit Diseases and Pests in North China, Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097 China
| | - Peter E. Mortimer
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan China
- Centre for Mountain Futures (CMF), Kunming Institute of Botany, Chinese Academy of Science, Kunming, 650201 Yunnan China
| | - Huili Li
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan China
- Centre for Mountain Futures (CMF), Kunming Institute of Botany, Chinese Academy of Science, Kunming, 650201 Yunnan China
| | - Saisamorm Lumyong
- Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, 50200 Thailand
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200 Thailand
- Academy of Science, The Royal Society of Thailand, Bangkok, 10300 Thailand
| | - Worawoot Aiduang
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Jaturong Kumla
- Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, 50200 Thailand
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Nakarin Suwannarach
- Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, 50200 Thailand
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Chitrabhanu S. Bhunjun
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- School of Science, Mae Fah Luang University, Chiang Rai, 57100 Thailand
| | - Feng-Ming Yu
- Center of Excellence in Fungal Research, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- School of Science, Mae Fah Luang University, Chiang Rai, 57100 Thailand
- Yunnan Key Laboratory of Fungal Diversity and Green Development, Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan China
| | - Qi Zhao
- Yunnan Key Laboratory of Fungal Diversity and Green Development, Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan China
| | - Doug Schaefer
- Centre for Mountain Futures (CMF), Kunming Institute of Botany, Chinese Academy of Science, Kunming, 650201 Yunnan China
| | - Marc Stadler
- Department Microbial Drugs, Helmholtz Centre for Infection Research (HZI), and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstrasse 7, 38124 Brunswick, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Brunswick, Germany
| |
Collapse
|
22
|
Gao A, Kouznetsova VL, Tsigelny IF. Machine-Learning-Based Virtual Screening to Repurpose Drugs for Treatment of Candida albicans Infection. Mycoses 2022; 65:794-805. [PMID: 35639510 DOI: 10.1111/myc.13475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Approximately 30% of Candida genus isolates are resistant to all currently available antifungal drugs and it is highly important to develop new treatments. Additionally, many current drugs are toxic and cause unwanted side effects. 1,3-beta-glucan synthase is an essential enzyme that builds the cell walls of Candida. OBJECTIVES Targeting CaFKS1, a subunit of the synthase, could be used to fight Candida. METHODS In the present study, a machine-learning model based on chemical descriptors was trained to recognize drugs that inhibit CaFKS1. The model attained 96.72% accuracy for classifying between active and inactive drug compounds. Descriptors for FDA-approved and other drugs were calculated and the model was used to predict the potential activity of these drugs against CaFKS1. RESULTS Several drugs, including goserelin and icatibant, were detected as active with high confidence. Many of the drugs, interestingly, were gonadotrophin-releasing hormone (GnRH) antagonists or agonists. A literature search found that five of the predicted drugs inhibit Candida experimentally. CONCLUSIONS This study yields promising drugs to be repurposed to combat Candida albicans infection. Future steps include testing the drugs on fungal cells in vitro.
Collapse
Affiliation(s)
- Andrew Gao
- REHS Program, San Diego Supercomputer Center, University of California at San Diego, La Jolla, Calif, USA.,MAP Program, University of California at San Diego, La Jolla, Calif, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California at San Diego, La Jolla, Calif, USA.,BiAna, La Jolla, Calif, USA
| | - Igor F Tsigelny
- San Diego Supercomputer Center, University of California at San Diego, La Jolla, Calif, USA.,BiAna, La Jolla, Calif, USA.,Department of Neurosciences, University of California at San Diego, La Jolla, Calif, USA
| |
Collapse
|
23
|
Vicente I, Baroncelli R, Hermosa R, Monte E, Vannacci G, Sarrocco S. Role and genetic basis of specialised secondary metabolites in Trichoderma ecophysiology. FUNGAL BIOL REV 2022. [DOI: 10.1016/j.fbr.2021.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
24
|
Valero C, Colabardini AC, de Castro PA, Silva LP, Ries LNA, Pardeshi L, Wang F, Rocha MC, Malavazi I, Silva RN, Martins C, Domingos P, Pereira-Silva C, Bromley MJ, Wong KH, Goldman GH. Aspergillus Fumigatus ZnfA, a Novel Zinc Finger Transcription Factor Involved in Calcium Metabolism and Caspofungin Tolerance. FRONTIERS IN FUNGAL BIOLOGY 2021; 2:689900. [PMID: 37744107 PMCID: PMC10512341 DOI: 10.3389/ffunb.2021.689900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/16/2021] [Indexed: 09/26/2023]
Abstract
Invasive pulmonary aspergillosis is a life-threatening fungal infection especially in the immunocompromised patients. The low diversity of available antifungal drugs coupled with the emergence of antifungal resistance has become a worldwide clinical concern. The echinocandin Caspofungin (CSP) is recommended as a second-line therapy but resistance and tolerance mechanisms have been reported. However, how the fungal cell articulates the response to CSP is not completely understood. This work provides a detailed characterization of ZnfA, a transcription factor (TF) identified in previous screening studies that is involved in the A. fumigatus responses to calcium and CSP. This TF plays an important role in the regulation of iron homeostasis and cell wall organization in response to high CSP concentrations as revealed by Chromatin Immunoprecipitation coupled to DNA sequencing (ChIP-seq) analysis. Furthermore, ZnfA acts collaboratively with the key TF CrzA in modulating the response to calcium as well as cell wall and osmotic stresses. This study therefore describes the existence of an additional, previously unknown TF that bridges calcium signaling and the CSP cellular response and further exposes the complex connections that exist among different pathways which govern stress sensing and signaling in A. fumigatus.
Collapse
Affiliation(s)
- Clara Valero
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Ana Cristina Colabardini
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Patrícia Alves de Castro
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Lilian Pereira Silva
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Lakhansing Pardeshi
- Faculty of Health Sciences, University of Macau, Macau, China
- Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Fang Wang
- Faculty of Health Sciences, University of Macau, Macau, China
- Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Marina Campos Rocha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Paulo, Brazil
| | - Iran Malavazi
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Paulo, Brazil
| | | | - Celso Martins
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Patrícia Domingos
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Cristina Pereira-Silva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Michael J. Bromley
- Manchester Fungal Infection Group, Core Technology Facility, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Koon Ho Wong
- Faculty of Health Sciences, University of Macau, Macau, China
- Faculty of Health Sciences, Institute of Translational Medicine, University of Macau, Macau, China
| | - Gustavo H. Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
25
|
Curto MÁ, Butassi E, Ribas JC, Svetaz LA, Cortés JCG. Natural products targeting the synthesis of β(1,3)-D-glucan and chitin of the fungal cell wall. Existing drugs and recent findings. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 88:153556. [PMID: 33958276 DOI: 10.1016/j.phymed.2021.153556] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/12/2021] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND During the last three decades systemic fungal infections associated to immunosuppressive therapies have become a serious healthcare problem. Clinical development of new antifungals is an urgent requirement. Since fungal but not mammalian cells are encased in a carbohydrate-containing cell wall, which is required for the growth and viability of fungi, the inhibition of cell wall synthesizing machinery, such as β(1,3)-D-glucan synthases (GS) and chitin synthases (CS) that catalyze the synthesis of β(1-3)-D-glucan and chitin, respectively, represent an ideal mode of action of antifungal agents. Although the echinocandins anidulafungin, caspofungin and micafungin are clinically well-established GS inhibitors for the treatment of invasive fungal infections, much effort must still be made to identify inhibitors of other enzymes and processes involved in the synthesis of the fungal cell wall. PURPOSE Since natural products (NPs) have been the source of several antifungals in clinical use and also have provided important scaffolds for the development of semisynthetic analogues, this review was devoted to investigate the advances made to date in the discovery of NPs from plants that showed capacity of inhibiting cell wall synthesis targets. The chemical characterization, specific target, discovery process, along with the stage of development are provided here. METHODS An extensive systematic search for NPs against the cell wall was performed considering all the articles published until the end of 2020 through the following scientific databases: NCBI PubMed, Scopus and Google Scholar and using the combination of the terms "natural antifungals" and "plant extracts" with "fungal cell wall". RESULTS The first part of this review introduces the state of the art of the structure and biosynthesis of the fungal cell wall and considers exclusively those naturally produced GS antifungals that have given rise to both existing semisynthetic approved drugs and those derivatives currently in clinical trials. According to their chemical structure, natural GS inhibitors can be classified as 1) cyclic lipopeptides, 2) glycolipids and 3) acidic terpenoids. We also included nikkomycins and polyoxins, NPs that inhibit the CS, which have traditionally been considered good candidates for antifungal drug development but have finally been discarded after enduring unsuccessful clinical trials. Finally, the review focuses in the most recent findings about the growing field of plant-derived molecules and extracts that exhibit activity against the fungal cell wall. Thus, this search yielded sixteen articles, nine of which deal with pure compounds and seven with plant extracts or fractions with proven activity against the fungal cell wall. Regarding the mechanism of action, seven (44%) produced GS inhibition while five (31%) inhibited CS. Some of them (56%) interfered with other components of the cell wall. Most of the analyzed articles refer to tests carried out in vitro and therefore are in early stages of development. CONCLUSION This report delivers an overview about both existing natural antifungals targeting GS and CS activities and their mechanisms of action. It also presents recent discoveries on natural products that may be used as starting points for the development of potential selective and non-toxic antifungal drugs.
Collapse
Affiliation(s)
- M Ángeles Curto
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
| | - Estefanía Butassi
- Área Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Juan C Ribas
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
| | - Laura A Svetaz
- Área Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| | - Juan C G Cortés
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
26
|
Carvalho VSD, Gómez-Delgado L, Curto MÁ, Moreno MB, Pérez P, Ribas JC, Cortés JCG. Analysis and application of a suite of recombinant endo-β(1,3)-D-glucanases for studying fungal cell walls. Microb Cell Fact 2021; 20:126. [PMID: 34217291 PMCID: PMC8254974 DOI: 10.1186/s12934-021-01616-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/19/2021] [Indexed: 12/31/2022] Open
Abstract
Background The fungal cell wall is an essential and robust external structure that protects the cell from the environment. It is mainly composed of polysaccharides with different functions, some of which are necessary for cell integrity. Thus, the process of fractionation and analysis of cell wall polysaccharides is useful for studying the function and relevance of each polysaccharide, as well as for developing a variety of practical and commercial applications. This method can be used to study the mechanisms that regulate cell morphogenesis and integrity, giving rise to information that could be applied in the design of new antifungal drugs. Nonetheless, for this method to be reliable, the availability of trustworthy commercial recombinant cell wall degrading enzymes with non-contaminating activities is vital. Results Here we examined the efficiency and reproducibility of 12 recombinant endo-β(1,3)-d-glucanases for specifically degrading the cell wall β(1,3)-d-glucan by using a fast and reliable protocol of fractionation and analysis of the fission yeast cell wall. This protocol combines enzymatic and chemical degradation to fractionate the cell wall into the four main polymers: galactomannoproteins, α-glucan, β(1,3)-d-glucan and β(1,6)-d-glucan. We found that the GH16 endo-β(1,3)-d-glucanase PfLam16A from Pyrococcus furiosus was able to completely and reproducibly degrade β(1,3)-d-glucan without causing the release of other polymers. The cell wall degradation caused by PfLam16A was similar to that of Quantazyme, a recombinant endo-β(1,3)-d-glucanase no longer commercially available. Moreover, other recombinant β(1,3)-d-glucanases caused either incomplete or excessive degradation, suggesting deficient access to the substrate or release of other polysaccharides. Conclusions The discovery of a reliable and efficient recombinant endo-β(1,3)-d-glucanase, capable of replacing the previously mentioned enzyme, will be useful for carrying out studies requiring the digestion of the fungal cell wall β(1,3)-d-glucan. This new commercial endo-β(1,3)-d-glucanase will allow the study of the cell wall composition under different conditions, along the cell cycle, in response to environmental changes or in cell wall mutants. Furthermore, this enzyme will also be greatly valuable for other practical and commercial applications such as genome research, chromosomes extraction, cell transformation, protoplast formation, cell fusion, cell disruption, industrial processes and studies of new antifungals that specifically target cell wall synthesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-021-01616-0.
Collapse
Affiliation(s)
- Vanessa S D Carvalho
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
| | - Laura Gómez-Delgado
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
| | - M Ángeles Curto
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
| | - M Belén Moreno
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
| | - Pilar Pérez
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
| | - Juan Carlos Ribas
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain.
| | - Juan Carlos G Cortés
- Instituto de Biología Funcional y Genómica Zacarías González, 2. CSIC and Universidad de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
27
|
Kotey FCN, Dayie NTKD, Tetteh-Uarcoo PB, Donkor ES. Candida Bloodstream Infections: Changes in Epidemiology and Increase in Drug Resistance. Infect Dis (Lond) 2021; 14:11786337211026927. [PMID: 34248358 PMCID: PMC8236779 DOI: 10.1177/11786337211026927] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/30/2021] [Indexed: 11/17/2022] Open
Abstract
The literature on bloodstream infections (BSIs) have predominantly been biased towards bacteria, given their superior clinical significance in comparison with the other types of microorganisms. Fungal pathogens have epidemiologically received relatively less attention, although they constitute an important proportion of BSI aetiologies. In this review, the authors discuss the clinical relevance of fungal BSIs in the context of Candida species, as well as treatment options for the infections, emphasizing the compelling need to develop newer antifungals and strengthen antimicrobial stewardship programmes in the wake of the rapid spread of antifungal resistance.
Collapse
Affiliation(s)
- Fleischer CN Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Korle Bu, Accra, Ghana
- FleRhoLife Research Consult, Teshie, Accra, Ghana
| | - Nicholas TKD Dayie
- Department of Medical Microbiology, University of Ghana Medical School, Korle Bu, Accra, Ghana
| | | | - Eric S Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Korle Bu, Accra, Ghana
| |
Collapse
|
28
|
Emerging Prospects for Combating Fungal Infections by Targeting Phosphatidylinositol Transfer Proteins. Int J Mol Sci 2021; 22:ijms22136754. [PMID: 34201733 PMCID: PMC8269425 DOI: 10.3390/ijms22136754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/27/2022] Open
Abstract
The emergence of fungal “superbugs” resistant to the limited cohort of anti-fungal agents available to clinicians is eroding our ability to effectively treat infections by these virulent pathogens. As the threat of fungal infection is escalating worldwide, this dwindling response capacity is fueling concerns of impending global health emergencies. These developments underscore the urgent need for new classes of anti-fungal drugs and, therefore, the identification of new targets. Phosphoinositide signaling does not immediately appear to offer attractive targets due to its evolutionary conservation across the Eukaryota. However, recent evidence argues otherwise. Herein, we discuss the evidence identifying Sec14-like phosphatidylinositol transfer proteins (PITPs) as unexplored portals through which phosphoinositide signaling in virulent fungi can be chemically disrupted with exquisite selectivity. Recent identification of lead compounds that target fungal Sec14 proteins, derived from several distinct chemical scaffolds, reveals exciting inroads into the rational design of next generation Sec14 inhibitors. Development of appropriately refined next generation Sec14-directed inhibitors promises to expand the chemical weaponry available for deployment in the shifting field of engagement between fungal pathogens and their human hosts.
Collapse
|
29
|
Mela A, Momany M. Septins coordinate cell wall integrity and lipid metabolism in a sphingolipid-dependent process. J Cell Sci 2021; 135:256543. [PMID: 33912961 DOI: 10.1242/jcs.258336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 12/31/2020] [Indexed: 01/09/2023] Open
Abstract
Septins colocalize with membrane sterol-rich regions and facilitate recruitment of cell wall synthases during wall remodeling. We show that null mutants missing an Aspergillus nidulans core septin present in hexamers and octamers (ΔaspAcdc11, ΔaspBcdc3 or ΔaspCcdc12) are sensitive to multiple cell wall-disturbing agents that activate the cell wall integrity MAPK pathway. The null mutant missing the octamer-exclusive core septin (ΔaspDcdc10) showed similar sensitivity, but only to a single cell wall-disturbing agent and the null mutant missing the noncore septin (ΔaspE) showed only very mild sensitivity to a different single agent. Core septin mutants showed changes in wall polysaccharide composition and chitin synthase localization. Mutants missing any of the five septins resisted ergosterol-disrupting agents. Hexamer mutants showed increased sensitivity to sphingolipid-disrupting agents. Core septins mislocalized after treatment with sphingolipid-disrupting agents, but not after ergosterol-disrupting agents. Our data suggest that the core septins are involved in cell wall integrity signaling, that all five septins are involved in monitoring ergosterol metabolism, that the hexamer septins are required for sphingolipid metabolism and that septins require sphingolipids to coordinate the cell wall integrity response.
Collapse
Affiliation(s)
- Alexander Mela
- Fungal Biology Group and Plant Biology Department, University of Georgia, 2502 Miller Plant Science Building, Athens, GA 30602, USA
| | - Michelle Momany
- Fungal Biology Group and Plant Biology Department, University of Georgia, 2502 Miller Plant Science Building, Athens, GA 30602, USA
| |
Collapse
|
30
|
Hussain H, Mamadalieva NZ, Ali I, Elizbit, Green IR, Wang D, Zou L, Simal-Gandara J, Cao H, Xiao J. Fungal glycosides: Structure and biological function. Trends Food Sci Technol 2021; 110:611-651. [DOI: 10.1016/j.tifs.2021.02.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
31
|
Heard SC, Wu G, Winter JM. Antifungal natural products. Curr Opin Biotechnol 2021; 69:232-241. [PMID: 33640596 DOI: 10.1016/j.copbio.2021.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 12/19/2022]
Abstract
Natural products are specialized small molecules produced in Nature and play pivotal roles in many cellular processes. These compounds possess exquisite chemical diversity and represent some of the most important pharmaceutical agents in human health care. With the rampant rise of fungal pathogens that are becoming resistant to nearly all clinically available antibiotics, there is an increased urgency to find new antifungal therapies with novel modes of action. To meet this need, we must be able to quickly identify new bioactive chemical scaffolds within complex natural extracts, determine their mechanisms of action, and generate appreciable yields for preclinical studies. In this review, we will highlight naturally derived antifungal agents of clinical importance as well as those with strong potential as leads in drug development.
Collapse
Affiliation(s)
- Stephanie C Heard
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| | - Guangwei Wu
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, and Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China.
| | - Jaclyn M Winter
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
32
|
Jallow S, Govender NP. Ibrexafungerp: A First-in-Class Oral Triterpenoid Glucan Synthase Inhibitor. J Fungi (Basel) 2021; 7:jof7030163. [PMID: 33668824 PMCID: PMC7996284 DOI: 10.3390/jof7030163] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Ibrexafungerp (formerly SCY-078 or MK-3118) is a first-in-class triterpenoid antifungal or “fungerp” that inhibits biosynthesis of β-(1,3)-D-glucan in the fungal cell wall, a mechanism of action similar to that of echinocandins. Distinguishing characteristics of ibrexafungerp include oral bioavailability, a favourable safety profile, few drug–drug interactions, good tissue penetration, increased activity at low pH and activity against multi-drug resistant isolates including C. auris and C. glabrata. In vitro data has demonstrated broad and potent activity against Candida and Aspergillus species. Importantly, ibrexafungerp also has potent activity against azole-resistant isolates, including biofilm-forming Candida spp., and echinocandin-resistant isolates. It also has activity against the asci form of Pneumocystis spp., and other pathogenic fungi including some non-Candida yeasts and non-Aspergillus moulds. In vivo data have shown IBX to be effective for treatment of candidiasis and aspergillosis. Ibrexafungerp is effective for the treatment of acute vulvovaginal candidiasis in completed phase 3 clinical trials.
Collapse
Affiliation(s)
- Sabelle Jallow
- Centre for Healthcare-Associated Infections, Antimicrobial Resistance and Mycoses (CHARM), National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg 2131, South Africa;
- Correspondence: ; Tel.: +27-11-386-6395
| | - Nelesh P. Govender
- Centre for Healthcare-Associated Infections, Antimicrobial Resistance and Mycoses (CHARM), National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg 2131, South Africa;
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
33
|
Wong-Deyrup SW, Song X, Ng TW, Liu XB, Zeng JG, Qing ZX, Deyrup ST, He ZD, Zhang HJ. Plant-derived isoquinoline alkaloids that target ergosterol biosynthesis discovered by using a novel antifungal screening tool. Biomed Pharmacother 2021; 137:111348. [PMID: 33578237 DOI: 10.1016/j.biopha.2021.111348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 10/22/2022] Open
Abstract
The ergosterol pathway is a prime antifungal target as it is required for fungal survival, yet is not involved in human homeostasis. Methods to study the ergosterol pathway, however, are often time-consuming. The minimum inhibitory concentration (MIC) assay is a simple research tool that determines the lowest concentration at which a novel antimicrobial is active in vitro with limited scope to determine the mechanism of action for a drug. In this study, we show that by adding hydrogen peroxide, an oxidative stressor, or glutathione (GSH), an antioxidant, to modify a commonly performed MIC assay allowed us to screen selectively for new antifungal drugs that target ergosterol biosynthesis in fungi. A human pathogen and dermatophyte, Microsporum gypseum, was used as a test organism. When exposed to ergosterol targeting drugs, the hydrogen peroxide treatment significantly decreased fungal survival by reducing ergosterol in the cell wall, whereas GSH increased survival of M. gypseum. Further, by performing a series of experiments with M. gypseum and Trichophyton rubrum, it was determined that the oxidative stress from hydrogen peroxide causes cell death at different developmental stages based on fungal species. These findings allow us to describe a simple, high-throughput method for simultaneously screening new antifungal drugs for activity and effects on the ergosterol pathway. By using this tool, two isoquinoline alkaloids were discovered to be potent inhibitors of ergosterol biosynthesis in vitro by reducing the amount of ergosterol without affecting the expression of 1,3-β-glucan. Both compounds also significantly reduced the severity of acanthosis, hyperkeratosis, spongiosis and dermal edema in vivo.
Collapse
Affiliation(s)
- Siu Wah Wong-Deyrup
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China
| | - Xun Song
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China; School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen, PR China
| | - Tsz-Wai Ng
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China
| | - Xiu-Bin Liu
- Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization and National Chinese Medicinal Herbs Hunan Technology Center, Hunan Agricultural University, Changsha 410128, PR China; Hunan Co-Innovation Center for Utilization of Botanicals Functional Ingredients, Hunan University of Chinese Medicine, Changsha 410208, PR China
| | - Jian-Guo Zeng
- Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization and National Chinese Medicinal Herbs Hunan Technology Center, Hunan Agricultural University, Changsha 410128, PR China; Hunan Co-Innovation Center for Utilization of Botanicals Functional Ingredients, Hunan University of Chinese Medicine, Changsha 410208, PR China
| | - Zhi-Xing Qing
- Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization and National Chinese Medicinal Herbs Hunan Technology Center, Hunan Agricultural University, Changsha 410128, PR China; Hunan Co-Innovation Center for Utilization of Botanicals Functional Ingredients, Hunan University of Chinese Medicine, Changsha 410208, PR China
| | - Stephen T Deyrup
- Department of Chemistry and Biochemistry, Siena College, Loudonville, NY 12211, USA
| | - Zhen-Dan He
- School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen, PR China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Hong-Jie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China.
| |
Collapse
|
34
|
Gil-Rodríguez AM, Garcia-Gutierrez E. Antimicrobial mechanisms and applications of yeasts. ADVANCES IN APPLIED MICROBIOLOGY 2020; 114:37-72. [PMID: 33934852 DOI: 10.1016/bs.aambs.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Yeasts and humans have had a close relationship for millenia. Yeast have been used for food production since the first human societies. Since then, alternative uses have been discovered. Nowadays, antibiotic resistance constitutes a pressing need worldwide. In order to overcome this threat, one of the most important strategies is the search for new antimicrobials in natural sources. Moreover, biopreservation based on natural sources has emerged as an alternative to more common chemical preservatives. Yeasts constitute an underexploited source of antagonistic activity against other microorganisms. Here, we compile a summary of the antagonistic activity of yeast origin against other yeast and other microorganisms, such as bacteria or parasites. We present the mechanisms of action used by yeasts to display these activities. We also provide applications of these antagonistic activities in food industry and agriculture, medicine and veterinary, where yeast promise to play a pivotal role in the near future.
Collapse
|
35
|
Ibrexafungerp: An orally active β-1,3-glucan synthesis inhibitor. Bioorg Med Chem Lett 2020; 32:127661. [PMID: 33160023 DOI: 10.1016/j.bmcl.2020.127661] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 11/21/2022]
Abstract
We previously reported medicinal chemistry efforts that identified MK-5204, an orally efficacious β-1,3-glucan synthesis inhibitor derived from the natural product enfumafungin. Further extensive optimization of the C2 triazole substituent identified 4-pyridyl as the preferred replacement for the carboxamide of MK-5204, leading to improvements in antifungal activity in the presence of serum, and increased oral exposure. Reoptimizing the aminoether at C3 in the presence of this newly discovered C2 substituent, confirmed that the (R) t-butyl, methyl aminoether of MK-5204 provided the best balance of these two key parameters, culminating in the discovery of ibrexafungerp, which is currently in phase III clinical trials. Ibrexafungerp displayed significantly improved oral efficacy in murine infection models, making it a superior candidate for clinical development as an oral treatment for Candida and Aspergillus infections.
Collapse
|
36
|
Garcia-Effron G. Rezafungin-Mechanisms of Action, Susceptibility and Resistance: Similarities and Differences with the Other Echinocandins. J Fungi (Basel) 2020; 6:E262. [PMID: 33139650 PMCID: PMC7711656 DOI: 10.3390/jof6040262] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
Rezafungin (formerly CD101) is a new β-glucan synthase inhibitor that is chemically related with anidulafungin. It is considered the first molecule of the new generation of long-acting echinocandins. It has several advantages over the already approved by the Food and Drug Administration (FDA) echinocandins as it has better tissue penetration, better pharmacokinetic/phamacodynamic (PK/PD) pharmacometrics, and a good safety profile. It is much more stable in solution than the older echinocandins, making it more flexible in terms of dosing, storage, and manufacturing. These properties would allow rezafungin to be administered once-weekly (intravenous) and to be potentially administered topically and subcutaneously. In addition, higher dose regimens were tested with no evidence of toxic effect. This will eventually prevent (or reduce) the selection of resistant strains. Rezafungin also has several similarities with older echinocandins as they share the same in vitro behavior (very similar Minimum Inhibitory Concentration required to inhibit the growth of 50% of the isolates (MIC50) and half enzyme maximal inhibitory concentration 50% (IC50)) and spectrum, the same target, and the same mechanisms of resistance. The selection of FKS mutants occurred at similar frequency for rezafungin than for anidulafungin and caspofungin. In this review, rezafungin mechanism of action, target, mechanism of resistance, and in vitro data are described in a comparative manner with the already approved echinocandins.
Collapse
Affiliation(s)
- Guillermo Garcia-Effron
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, C.P. 3000 Santa Fe, Argentina; or ; Tel.: +54-9342-4575209 (ext. 135)
- Consejo Nacional de Investigaciones Científicas y Tecnológicas, C.P. 3000 Santa Fe, Argentina
| |
Collapse
|
37
|
Stanford FA, Voigt K. Iron Assimilation during Emerging Infections Caused by Opportunistic Fungi with emphasis on Mucorales and the Development of Antifungal Resistance. Genes (Basel) 2020; 11:genes11111296. [PMID: 33143139 PMCID: PMC7693903 DOI: 10.3390/genes11111296] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Iron is a key transition metal required by most microorganisms and is prominently utilised in the transfer of electrons during metabolic reactions. The acquisition of iron is essential and becomes a crucial pathogenic event for opportunistic fungi. Iron is not readily available in the natural environment as it exists in its insoluble ferric form, i.e., in oxides and hydroxides. During infection, the host iron is bound to proteins such as transferrin, ferritin, and haemoglobin. As such, access to iron is one of the major hurdles that fungal pathogens must overcome in an immunocompromised host. Thus, these opportunistic fungi utilise three major iron acquisition systems to overcome this limiting factor for growth and proliferation. To date, numerous iron acquisition pathways have been fully characterised, with key components of these systems having major roles in virulence. Most recently, proteins involved in these pathways have been linked to the development of antifungal resistance. Here, we provide a detailed review of our current knowledge of iron acquisition in opportunistic fungi, and the role iron may have on the development of resistance to antifungals with emphasis on species of the fungal basal lineage order Mucorales, the causative agents of mucormycosis.
Collapse
Affiliation(s)
- Felicia Adelina Stanford
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology–Hans Knöll Institute, Jena, Adolf-Reichwein-Straße 23, 07745 Jena, Germany;
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller University Jena, Neugasse 25, 07743 Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology–Hans Knöll Institute, Jena, Adolf-Reichwein-Straße 23, 07745 Jena, Germany;
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller University Jena, Neugasse 25, 07743 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena Microbial Resource Collection Adolf-Reichwein-Straße 23, 07745 Jena, Germany
- Correspondence: ; Tel.: +49-3641-532-1395; Fax: +49-3641-532-2395
| |
Collapse
|
38
|
Sumiyoshi M, Miyazaki T, Makau JN, Mizuta S, Tanaka Y, Ishikawa T, Makimura K, Hirayama T, Takazono T, Saijo T, Yamaguchi H, Shimamura S, Yamamoto K, Imamura Y, Sakamoto N, Obase Y, Izumikawa K, Yanagihara K, Kohno S, Mukae H. Novel and potent antimicrobial effects of caspofungin on drug-resistant Candida and bacteria. Sci Rep 2020; 10:17745. [PMID: 33082485 PMCID: PMC7576149 DOI: 10.1038/s41598-020-74749-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Echinocandins, including caspofungin, micafungin, and anidulafungin, are first-line antifungal agents for the treatment of invasive candidiasis. They exhibit fungicidal activity by inhibiting the synthesis of β-1,3-D-glucan, an essential component of the fungal cell wall. However, they are active only against proliferating fungal cells and unable to completely eradicate fungal cells even after a 24 h drug exposure in standard time-kill assays. Surprisingly, we found that caspofungin, when dissolved in low ionic solutions, had rapid and potent antimicrobial activities against multidrug-resistant (MDR) Candida and bacteria cells even in non-growth conditions. This effect was not observed in 0.9% NaCl or other ion-containing solutions and was not exerted by other echinocandins. Furthermore, caspofungin dissolved in low ionic solutions drastically reduced mature biofilm cells of MDR Candida auris in only 5 min, as well as Candida-bacterial polymicrobial biofilms in a catheter-lock therapy model. Caspofungin displayed ion concentration-dependent conformational changes and intracellular accumulation with increased reactive oxygen species production, indicating a novel mechanism of action in low ionic conditions. Importantly, caspofungin dissolved in 5% glucose water did not exhibit increased toxicity to human cells. This study facilitates the development of new therapeutic strategies in the management of catheter-related biofilm infections.
Collapse
Affiliation(s)
- Makoto Sumiyoshi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Taiga Miyazaki
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Juliann Nzembi Makau
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima, 890-0065, Japan
| | - Koichi Makimura
- Medical Mycology, Graduate School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Tatsuro Hirayama
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Tomomi Saijo
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Hiroyuki Yamaguchi
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shintaro Shimamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kazuko Yamamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yoshifumi Imamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Noriho Sakamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yasushi Obase
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shigeru Kohno
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
39
|
Kastelberg B, Tubau-Juni N, Ayubi T, Leung A, Leber A, Hontecillas R, Bassaganya-Riera J, Kale SD. NLRX1 is a key regulator of immune signaling during invasive pulmonary aspergillosis. PLoS Pathog 2020; 16:e1008854. [PMID: 32956405 PMCID: PMC7529209 DOI: 10.1371/journal.ppat.1008854] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/01/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022] Open
Abstract
Aspergillus fumigatus is an opportunistic fungal pathogen of immunocompromised patient populations. Mortality is thought to be context-specific and occurs via both enhanced fungal growth and immunopathogenesis. NLRX1 is a negative regulator of immune signaling and metabolic pathways implicated in host responses to microbes, cancers, and autoimmune diseases. Our study indicates loss of Nlrx1 results in enhanced fungal burden, pulmonary inflammation, immune cell recruitment, and mortality across immuno-suppressed and immuno-competent models of IPA using two clinically derived isolates (AF293, CEA10). We observed that the heightened mortality is due to enhanced recruitment of CD103+ dendritic cells (DCs) that produce elevated amounts of IL-4 resulting in a detrimental Th2-mediated immune response. Adoptive transfer of Nlrx1-/- CD103+ DCs in neutropenic NRG mice results in enhanced mortality that can be ablated using IL-4 neutralizing antibodies. In vitro analysis of CD103+ DCs indicates loss of Nlrx1 results in enhanced IL-4 production via elevated activation of the JNK/JunB pathways. Interestingly, loss of Nlrx1 also results in enhanced recruitment of monocytes and neutrophils. Chimeras of irradiated Nlrx1-/- mice reconstituted with wild type bone marrow have enhanced neutrophil recruitment and survival during models of IPA. This enhanced immune cell recruitment in the absence of Nlrx1 is mediated by excessive production of CXCL8/IL-8 family of chemokines and IL-6 via early and enhanced activation of P38 in response to A. fumigatus conidia as shown in BEAS-2B airway epithelial cells. In summary, our results point strongly towards the cell-specific and contextual function of Nlrx1 during invasive pulmonary aspergillosis and may lead to novel therapeutics to reduce Th2 responses by CD103+ DCs or heightened recruitment of neutrophils. Fungal infections are mitigated and controlled in part by a robust immune response and generation of reactive oxygen species. In certain instances, the immune response may become harmful to the host. Nlrx1 is a known negative regulator of inflammatory aspects of the immune system in response to viruses, bacteria, and cancers. In this study we describe the novel importance of Nlrx1 in controlling and fighting fungal infections in two different host cell populations through two distinct mechanisms. Nlrx1 may function as a future target to mitigate inflammation and immunopathogenesis during fungal pulmonary infection as well as enhance beneficial neutrophil recruitment.
Collapse
Affiliation(s)
- Bridget Kastelberg
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Nuria Tubau-Juni
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Tariq Ayubi
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Austin Leung
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Andrew Leber
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
| | - Shiv D. Kale
- Nutritional Immunology and Molecular Medicine Institute, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
40
|
MK-5204: An orally active β-1,3-glucan synthesis inhibitor. Bioorg Med Chem Lett 2020; 30:127357. [DOI: 10.1016/j.bmcl.2020.127357] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/11/2020] [Accepted: 06/14/2020] [Indexed: 11/21/2022]
|
41
|
McEvoy K, Normile TG, Poeta MD. Antifungal Drug Development: Targeting the Fungal Sphingolipid Pathway. J Fungi (Basel) 2020; 6:jof6030142. [PMID: 32825250 PMCID: PMC7559796 DOI: 10.3390/jof6030142] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Fungal infections are becoming more prevalent and problematic due to the continual rise of immune deficient patients as well as the progressive development of drug resistance towards currently available antifungal drugs. There has been a significant increase in the development of antifungal compounds with a similar mechanism of action of current drugs. In contrast, there has been very little progress in developing compounds inhibiting totally new fungal targets or/and fungal pathways. This review focuses on novel compounds recently discovered to target the fungal sphingolipids and their metabolizing enzymes.
Collapse
Affiliation(s)
- Kyle McEvoy
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA; (K.M.); (T.G.N.)
| | - Tyler G. Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA; (K.M.); (T.G.N.)
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA; (K.M.); (T.G.N.)
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Veterans Administration Medical Center, Northport, NY 11768, USA
- Correspondence: ; Tel.: +1-631-632-4024
| |
Collapse
|
42
|
Ma H, Zhao X, Yang L, Su P, Fu P, Peng J, Yang N, Guo G. Antimicrobial Peptide AMP-17 Affects Candida albicans by Disrupting Its Cell Wall and Cell Membrane Integrity. Infect Drug Resist 2020; 13:2509-2520. [PMID: 32801789 PMCID: PMC7398874 DOI: 10.2147/idr.s250278] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/13/2020] [Indexed: 01/25/2023] Open
Abstract
Background Candida albicans is associated with high mortality among immunocompromised patients. Resistance to and toxic side effects of antifungal drugs require the development of alternative antifungal agents. AMP-17 is a novel antimicrobial peptide derived from Musca domestica that exerts excellent antifungal effects against the Candida species. In this article, we discuss the potential mechanism of AMP-17 against C. albicans from the perspective of affecting the latter's cell external structure. Methods Recombinant AMP-17 was prepared by prokaryotic expression system, and its anti-C. albicans activity was detected by microdilution method. Microscopy and scanning electron microscopy were used to examine morphological changes in C. albicans. Cell wall-specific staining method was used to detect the change of cell wall integrity of C. albicans after AMP-17 treatment. AMP-17-induced damage to the C. albicans cell membrane was analyzed by fluorescent probes and glycerol assay kit. The expression of genes related to fungal cell wall and cell-membrane synthesis was detected by qRT-PCR. Results Morphological observations showed that the growth of C. albicans was significantly inhibited in AMP-17-treated cells; the cells appeared aggregated and dissolved, with severe irregularities in shape. Furthermore, AMP-17 damaged the integrity of C. albicans cell walls. The cell wall integrity rate of AMP-17-treated cells was only 21.7% compared to untreated cells. Moreover, the change of membrane dynamics and permeability suggested that the cell membrane was disrupted by AMP-17 treatment. Genetic analysis showed that after AMP-17 treatment, the cell wall synthesis-related gene FKS2 of C. albicans was up-regulated 3.46-fold, while the cell membrane ergosterol synthesis-related genes ERG1, ERG5, ERG6, and MET6 were down-regulated 5.88-, 17.54-, 13.33-, and 7.14-fold, respectively. Conclusion AMP-17 treatment disrupted the cell wall integrity and membrane structure of C. albicans and is likely a novel therapeutic option for prevention and control of C. albicans infections.
Collapse
Affiliation(s)
- Huiling Ma
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China.,Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Xinyu Zhao
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China
| | - Longbing Yang
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China
| | - Peipei Su
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China
| | - Ping Fu
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, People's Republic of China
| | - Jian Peng
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, People's Republic of China
| | - Na Yang
- School of Food Science, Guizhou Medical University, Guiyang 550025, People's Republic of China
| | - Guo Guo
- Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, People's Republic of China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, People's Republic of China.,School of Food Science, Guizhou Medical University, Guiyang 550025, People's Republic of China
| |
Collapse
|
43
|
Liu W, Yuan L, Wang S. Recent Progress in the Discovery of Antifungal Agents Targeting the Cell Wall. J Med Chem 2020; 63:12429-12459. [PMID: 32692166 DOI: 10.1021/acs.jmedchem.0c00748] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Due to the limit of available treatments and the emergence of drug resistance in the clinic, invasive fungal infections are an intractable problem with high morbidity and mortality. The cell wall, as a fungi-specific structure, is an appealing target for the discovery and development of novel and low-toxic antifungal agents. In an attempt to accelerate the discovery of novel cell wall targeted drugs, this Perspective will provide a comprehensive review of the progress made to date on the development of fungal cell wall inhibitors. Specifically, this review will focus on the targets, discovery process, chemical structures, antifungal activities, and structure-activity relationships. Although two types of cell wall antifungal agents are clinically available or in clinical trials, it is still a long way for the other cell wall targeted inhibitors to be translated into clinical applications. Future efforts should be focused on the identification of inhibitors against novel conserved cell wall targets.
Collapse
Affiliation(s)
- Wei Liu
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xuefu Middle Road, Xi'an 710021, People's Republic of China
| | - Lin Yuan
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xuefu Middle Road, Xi'an 710021, People's Republic of China
| | - Shengzheng Wang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, People's Republic of China
| |
Collapse
|
44
|
Abstract
Aspergillus fumigatus, one of the most important human-pathogenic fungal species, is able to cause aspergillosis, a heterogeneous group of diseases that presents a wide range of clinical manifestations. Invasive pulmonary aspergillosis is the most serious pathology in terms of patient outcome and treatment, with a high mortality rate ranging from 50% to 95% primarily affecting immunocompromised patients. Azoles have been used for many years as the main antifungal agents to treat and prevent invasive aspergillosis. However, there were several reports of evolution of clinical azole resistance in the last decade. Caspofungin, a noncompetitive β-1,3-glucan synthase inhibitor, has been used against A. fumigatus, but it is fungistatic and is recommended as second-line therapy for invasive aspergillosis. More information about caspofungin tolerance and resistance is necessary in order to refine antifungal strategies that target the fungal cell wall. Here, we screened a transcription factor (TF) deletion library for TFs that can mediate caspofungin tolerance and resistance. We have identified 11 TFs that are important for caspofungin sensitivity and/or for the caspofungin paradoxical effect (CPE). These TFs encode proteins involved in the basal modulation of the RNA polymerase II initiation sites, calcium metabolism or cell wall remodeling, and mitochondrial respiratory function. The study of those genes regulated by TFs identified in this work will provide a better understanding of the signaling pathways that are important for caspofungin tolerance and resistance. Aspergillus fumigatus is the leading cause of pulmonary fungal diseases. Azoles have been used for many years as the main antifungal agents to treat and prevent invasive aspergillosis. However, in the last 10 years there have been several reports of azole resistance in A. fumigatus and new strategies are needed to combat invasive aspergillosis. Caspofungin is effective against other human-pathogenic fungal species, but it is fungistatic only against A. fumigatus. Resistance to caspofungin in A. fumigatus has been linked to mutations in the fksA gene that encodes the target enzyme of the drug β-1,3-glucan synthase. However, tolerance of high caspofungin concentrations, a phenomenon known as the caspofungin paradoxical effect (CPE), is also important for subsequent adaptation and drug resistance evolution. Here, we identified and characterized the transcription factors involved in the response to CPE by screening an A. fumigatus library of 484 null transcription factors (TFs) in CPE drug concentrations. We identified 11 TFs that had reduced CPE and that encoded proteins involved in the basal modulation of the RNA polymerase II initiation sites, calcium metabolism, and cell wall remodeling. One of these TFs, FhdA, was important for mitochondrial respiratory function and iron metabolism. The ΔfhdA mutant showed decreased growth when exposed to Congo red or to high temperature. Transcriptome sequencing (RNA-seq) analysis and further experimental validation indicated that the ΔfhdA mutant showed diminished respiratory capacity, probably affecting several pathways related to the caspofungin tolerance and resistance. Our results provide the foundation to understand signaling pathways that are important for caspofungin tolerance and resistance.
Collapse
|
45
|
Jakubczyk D, Dussart F. Selected Fungal Natural Products with Antimicrobial Properties. Molecules 2020; 25:E911. [PMID: 32085562 PMCID: PMC7070998 DOI: 10.3390/molecules25040911] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 01/14/2023] Open
Abstract
Fungal natural products and their effects have been known to humankind for hundreds of years. For example, toxic ergot alkaloids produced by filamentous fungi growing on rye poisoned thousands of people and livestock throughout the Middle Ages. However, their later medicinal applications, followed by the discovery of the first class of antibiotics, penicillins and other drugs of fungal origin, such as peptidic natural products, terpenoids or polyketides, have altered the historically negative reputation of fungal "toxins". The development of new antimicrobial drugs is currently a major global challenge, mainly due to antimicrobial resistance phenomena. Therefore, the structures, biosynthesis and antimicrobial activity of selected fungal natural products are described here.
Collapse
Affiliation(s)
- Dorota Jakubczyk
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznań, Poland
| | - Francois Dussart
- Department of Agriculture, Horticulture and Engineering Science, Scotland’s Rural College (SRUC), Edinburgh EH9 3JG, UK;
| |
Collapse
|
46
|
Chhetri A, Loksztejn A, Nguyen H, Pianalto KM, Kim MJ, Hong J, Alspaugh JA, Yokoyama K. Length Specificity and Polymerization Mechanism of (1,3)-β-d-Glucan Synthase in Fungal Cell Wall Biosynthesis. Biochemistry 2020; 59:682-693. [PMID: 31899625 DOI: 10.1021/acs.biochem.9b00896] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
(1,3)-β-d-Glucan synthase (GS) catalyzes formation of the linear (1,3)-β-d-glucan in the fungal cell wall and is a target of clinically approved antifungal antibiotics. The catalytic subunit of GS, FKS protein, does not exhibit significant sequence homology to other glycosyltransferases, and thus, significant ambiguity about its catalytic mechanism remains. One of the major technical barriers in studying GS is the absence of activity assay methods that allow characterization of the lengths and amounts of (1,3)-β-d-glucan due to its poor solubility in water and organic solvents. Here, we report a successful development of a novel GS activity assay based on size-exclusion chromatography coupled with pulsed amperometric detection and radiation counting (SEC-PAD-RC), which allows for the simultaneous characterization of the amount and length of the polymer product. The assay revealed that the purified yeast GS produces glucan with a length of 6550 ± 760 mer, consistent with the reported degree of polymerization of (1,3)-β-d-glucan isolated from intact cells. Pre-steady state kinetic analysis revealed a highly efficient but rate-determining chain elongation rate of 51.5 ± 9.8 s-1, which represents the first observation of chain elongation by a nucleotide-sugar-dependent polysaccharide synthase. Coupling the SEC-PAD-RC method with substrate analogue mechanistic probes provided the first unambiguous evidence that GS catalyzes non-reducing end polymerization. On the basis of these observations, we propose a detailed model for the catalytic mechanism of GS. The approaches described here can be used to determine the mechanism of catalysis of other polysaccharide synthases.
Collapse
Affiliation(s)
- Abhishek Chhetri
- Department of Biochemistry , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Anna Loksztejn
- Department of Biochemistry , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Hai Nguyen
- Department of Biochemistry , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Kaila M Pianalto
- Department of Medicine , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Mi Jung Kim
- Department of Chemistry , Duke University , Durham , North Carolina 27708-0354 , United States
| | - Jiyong Hong
- Department of Chemistry , Duke University , Durham , North Carolina 27708-0354 , United States
| | - J Andrew Alspaugh
- Department of Medicine , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Kenichi Yokoyama
- Department of Biochemistry , Duke University School of Medicine , Durham , North Carolina 27710 , United States.,Department of Chemistry , Duke University , Durham , North Carolina 27708-0354 , United States
| |
Collapse
|
47
|
Wagener J, Striegler K, Wagener N. α- and β-1,3-Glucan Synthesis and Remodeling. Curr Top Microbiol Immunol 2020; 425:53-82. [PMID: 32193600 DOI: 10.1007/82_2020_200] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucans are characteristic and major constituents of fungal cell walls. Depending on the species, different glucan polysaccharides can be found. These differ in the linkage of the D-glucose monomers which can be either in α- or β-conformation and form 1,3, 1,4 or 1,6 O-glycosidic bonds. The linkages and polymer lengths define the physical properties of the glucan macromolecules, which may form a scaffold for other cell wall structures and influence the rigidity and elasticity of the wall. β-1,3-glucan is essential for the viability of many fungal pathogens. Therefore, the β-1,3-glucan synthase complex represents an excellent and primary target structure for antifungal drugs. Fungal cell wall β-glucan is also an important pathogen-associated molecular pattern (PAMP). To hide from innate immunity, many fungal pathogens depend on the synthesis of cell wall α-glucan, which functions as a stealth molecule to mask the β-glucans itself or links other masking structures to the cell wall. Here, we review the current knowledge about the biosynthetic machineries that synthesize β-1,3-glucan, β-1,6-glucan, and α-1,3-glucan. We summarize the discovery of the synthases, major regulatory traits, and the impact of glucan synthesis deficiencies on the fungal organisms. Despite all efforts, many aspects of glucan synthesis remain yet unresolved, keeping research directed toward cell wall biogenesis an exciting and continuously challenging topic.
Collapse
Affiliation(s)
- Johannes Wagener
- Institut Für Hygiene Und Mikrobiologie, University of Würzburg, Würzburg, Germany. .,National Reference Center for Invasive Fungal Infections (NRZMyk), Jena, Germany.
| | - Kristina Striegler
- Institut Für Hygiene Und Mikrobiologie, University of Würzburg, Würzburg, Germany
| | - Nikola Wagener
- Department of Cell Biology, Medical Faculty, University of Munich, Martinsried, Germany
| |
Collapse
|
48
|
Abstract
Antifungal therapy is a critical component of patient management for invasive fungal diseases. Yet, therapeutic choices are limited as only a few drug classes are available to treat systemic disease, and some infecting strains are resistant to one or more drug classes. The ideal antifungal inhibits a fungal-specific essential target not present in human cells to avoid off-target toxicities. The fungal cell wall is an ideal drug target because its integrity is critical to cell survival and a majority of biosynthetic enzymes and wall components is unique to fungi. Among currently approved antifungal agents and those in clinical development, drugs targeting biosynthetic enzymes of the cell wall show safe and efficacious antifungal properties, which validates the cell wall as a target. The echinocandins, which inhibit β-1,3-glucan synthase, are recommended as first-line therapy for Candida infections. Newer cell wall-active drugs in clinical development encompass next-generation glucan synthase inhibitors including a novel echinocandin and an enfumafungin, an inhibitor of Gwt1, a key component of GPI anchor protein biosynthesis, and a classic inhibitor of chitin biosynthesis. As the cell wall is rich in potential drug discovery targets, it is primed to help deliver the next generation of antifungal drugs.
Collapse
Affiliation(s)
- David S Perlin
- Center for Discovery and Innovation, 340 Kingsland Street, Nutley, 07110, USA.
| |
Collapse
|
49
|
Abhari K, Mousavi Khaneghah A. Alternative extraction techniques to obtain, isolate and purify proteins and bioactive from aquaculture and by-products. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 92:35-52. [PMID: 32402446 DOI: 10.1016/bs.afnr.2019.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Oceans cover more than 70% of the earth's surface and provide a great ecosystem for habitat of a large divers of marine species. The marine species are rich sources of bioactive compound that can be applied in medicine, pharmacology and food industry. Besides the marine species, fish processing industry also produces substantial volumes of by-products that can be used for a variety of purposes. Thus, it is important to find approaches to access to these valuable compounds. Nowadays, more factors have been considered in selecting an appropriate method for extraction of bioactive compounds such as consume less time and solvent, to be fast and ecofriendly. Concerns regarding entering the pollutions to the environment resulted to invest on the methods practicable with less chemical solvents and even green ones, however, implementation of stricter regulations and policies is required to encourage researchers to set up the procedures with reduced toxic agents to guarantee the environmental safety. In the current chapter the most common marine derived compounds and innovative methods for their extraction will be discussed.
Collapse
Affiliation(s)
- Khadijeh Abhari
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Mousavi Khaneghah
- Department of Food Science, Faculty of Food Engineering, State University of Campinas (UNICAMP), São Paulo, Brazil.
| |
Collapse
|
50
|
Thombare VJ, Holden JA, Reynolds EC, O'Brien-Simpson NM, Hutton CA. Celogentin mimetics as inhibitors of tubulin polymerization. J Pept Sci 2019; 26:e3239. [PMID: 31847053 DOI: 10.1002/psc.3239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/18/2022]
Abstract
Bicyclic analogues of celogentin C have been synthesized in which the side chain-side chain cross-links are replaced by thioether bonds. Several of the simplified bicyclic peptides displayed potent inhibition of tubulin polymerization.
Collapse
Affiliation(s)
- Varsha J Thombare
- School of Chemistry, The University of Melbourne, Victoria, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - James A Holden
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia.,Melbourne Dental School and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Eric C Reynolds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia.,Melbourne Dental School and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Neil M O'Brien-Simpson
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia.,Melbourne Dental School and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Craig A Hutton
- School of Chemistry, The University of Melbourne, Victoria, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|