1
|
Amann V, Kissmann AK, Firacative C, Rosenau F. Biofilm-Associated Candidiasis: Pathogenesis, Prevalence, Challenges and Therapeutic Options. Pharmaceuticals (Basel) 2025; 18:460. [PMID: 40283897 PMCID: PMC12030374 DOI: 10.3390/ph18040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The rising prevalence of fungal infections, especially those caused by Candida species, presents a major risk to global health. With approximately 1.5 million deaths annually, the urgency for effective treatment options has never been greater. Candida spp. are the leading cause of invasive infections, significantly impacting immunocompromised patients and those in healthcare settings. C. albicans, C. parapsilosis and the emerging species C. auris are categorized as highly dangerous species because of their pathogenic potential and increasing drug resistance. This review comparatively describes the formation of microbial biofilms of both bacterial and fungal origin, including major pathogens, thereby creating a novel focus. Biofilms can further complicate treatment, as these structures provide enhanced resistance to antifungal therapies. Traditional antifungal agents, including polyenes, azoles and echinocandins, have shown effectiveness, yet resistance development continues to rise, necessitating the exploration of novel therapeutic approaches. Antimicrobial peptides (AMPs) such as the anti-biofilm peptides Pom-1 and Cm-p5 originally isolated from snails represent promising candidates due to their unique mechanisms of action and neglectable cytotoxicity. This review article discusses the challenges posed by Candida infections, the characteristics of important species, the role of biofilms in virulence and the potential of new therapeutic options like AMPs.
Collapse
Affiliation(s)
- Valerie Amann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Ann-Kathrin Kissmann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Carolina Firacative
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogota 111221, Colombia;
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| |
Collapse
|
2
|
Busch RJ, Doty C, Mills CA, Latifi F, Herring LE, Konjufca V, Vargas-Muñiz JM. Deletion of core septin gene aspB in Aspergillus fumigatus results in fungicidal activity of caspofungin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640155. [PMID: 40060473 PMCID: PMC11888321 DOI: 10.1101/2025.02.25.640155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Septins are a family of GTP-binding proteins found in many eukaryotic lineages. Although highly conserved throughout many eukaryotes, their functions vary across species. In Aspergillus fumigatus, the etiological agent of invasive aspergillosis, septins participate in a variety of processes such as cell wall organization of conidia, septation, and response to cell wall stress. Previous studies determined that the ΔaspB strain had a greater sensitivity to anti-cell wall drugs, especially the echinocandin caspofungin, yet mechanisms behind this augmented sensitivity are unknown. We performed cell viability staining of the deletion strains post-caspofungin exposure and found that the ΔaspA, ΔaspB, and ΔaspC strains have significantly lower cell viability. Concomitant with the reduced viability, deletion strains are more susceptible to caspofungin on solid media. These results indicate that the septin cytoskeleton is important for A. fumigatus survival in the presence of caspofungin. Due to the potential of improved therapeutic outcome, we followed up using a neutropenic murine model of invasive aspergillosis. Animals infected with the ΔaspB strain and treated with caspofungin showed improved survival compared to the animals infected with akuB KU80 wild-type or complemented strains. Additionally, histological analysis showed reduced fungal burden and inflammation in the ΔaspB infected, caspofungin-treated group. Affinity purification coupled with quantitative proteomics identified proteins involved in the septin-dependent response to caspofungin, includng four candidate interactors involved in cell wall stress response. Deletion of these candidate genes resulted in increased susceptibility to caspofungin and moderately reduced viability post-drug exposure. Taken together, these data suggest that septin AspB contributes to the fungistatic response to caspofungin.
Collapse
Affiliation(s)
- Rebecca Jean Busch
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States
| | - Carson Doty
- School of Biological Sciences, Southern Illinois University-Carbondale, Carbondale, Illinois, United States
| | - C. Allie Mills
- Michael Hooker Metabolomics and Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Flutur Latifi
- Microbiology Program, Southern Illinois University-Carbondale, Carbondale, Illinois, United States
| | - Laura E. Herring
- Michael Hooker Metabolomics and Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Vjollca Konjufca
- Microbiology Program, Southern Illinois University-Carbondale, Carbondale, Illinois, United States
| | - José M Vargas-Muñiz
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States
- Fralin Life Science Institute, Virginia Tech, Blacksburg, VA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA
| |
Collapse
|
3
|
Gautam I, Yarava JR, Xu Y, Li R, Scott FJ, Mentink-Vigier F, Momany M, Latgé JP, Wang T. Comparative analysis of polysaccharide and cell wall structure in Aspergillus nidulans and Aspergillus fumigatus by solid-state NMR. Carbohydr Polym 2025; 348:122907. [PMID: 39562136 PMCID: PMC11576540 DOI: 10.1016/j.carbpol.2024.122907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
Invasive aspergillosis poses a significant threat to immunocompromised patients, leading to high mortality rates associated with these infections. Targeting the biosynthesis of cell wall carbohydrates is a promising strategy for antifungal drug development and will be advanced by a molecular-level understanding of the native structures of polysaccharides within their cellular context. Solid-state NMR spectroscopy has recently provided detailed insights into the cell wall organization of Aspergillus fumigatus, but genetic and biochemical evidence highlights species-specific differences among Aspergillus species. In this study, we employed a combination of 13C, 15N, and 1H-detection solid-state NMR, supplemented by Dynamic Nuclear Polarization (DNP), to compare the structural organization of cell wall polymers and their assembly in the cell walls of A. fumigatus and A. nidulans, both of which are key model organisms and human pathogens. The two species exhibited a similar rigid core architecture, consisting of chitin, α-glucan, and β-glucan, which contributed to comparable cell wall properties, including polymer dynamics, water retention, and supramolecular organization. However, differences were observed in the chitin, galactosaminogalactan, protein, and lipid content, as well as in the dynamics of galactomannan and the structure of the glucan matrix.
Collapse
Affiliation(s)
- Isha Gautam
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | | | - Yifan Xu
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Reina Li
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Faith J Scott
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | | | - Michelle Momany
- Fungal Biology Group & Department of Plant Biology, University of Georgia, Athens, GA, USA
| | - Jean-Paul Latgé
- Institute of Molecular Biology and Biotechnology, University of Crete, Heraklion, Greece; Fungal Respiratory Infections Research Unit and SFR ICAT, University of Angers, France
| | - Tuo Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
4
|
Andes D, Brüggemann RJ, Flanagan S, Lepak AJ, Lewis RE, Ong V, Rubino CM, Sandison T. The distinctive pharmacokinetic profile of rezafungin, a long-acting echinocandin developed in the era of modern pharmacometrics. J Antimicrob Chemother 2025; 80:18-28. [PMID: 39540899 PMCID: PMC11695911 DOI: 10.1093/jac/dkae415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Echinocandin drugs are the current first-line therapy for fungal infections caused by Candida spp. Most patients require once-daily intravenous (IV) administration in a hospital or outpatient setting for treatment, which may negatively impact their quality of life and stress healthcare resources. Similar to other echinocandins, the novel FDA-, EMA-, and Medical and Healthcare Products Regulatory Agency-approved echinocandin, rezafungin (CD101), exhibited strong antifungal activity against several fungal pathogens and a low drug-drug interaction liability, which are important for medically complex patients. A pharmacometric-based approach has been adopted throughout the development of rezafungin, which contrasts with older echinocandins where dosing regimens were largely derived empirically, and only recently based on pharmacometric guidance. This state-of-the-art approach used model-based simulations incorporating pre-clinical and clinical data as it became available to optimize the dosing regimen for rezafungin. The enhanced stability of the molecular structure and the safety profile of rezafungin allow for the administration of once-weekly IV doses, compared to the daily dosing requirement for other echinocandin drugs, with this distinctive pharmacokinetic profile of rezafungin resulting in a front-loaded dosing regimen with high exposures early in therapy for enhanced fungal killing. The long shelf-life of rezafungin makes this echinocandin more flexible in terms of storage and manufacturing. Demonstrated across clinical development, rezafungin may provide patients with next-generation first-line antifungal treatment for the treatment of candidaemia and invasive candidiasis.
Collapse
Affiliation(s)
- David Andes
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Roger J Brüggemann
- Department of Pharmacy and Radboudumc Institute for Medical Innovation, Radboud University Medical Center, and Radboudumc-CWZ Nijmegen Center of Expertise in Mycology, Nijmegen, The Netherlands
| | | | - Alexander J Lepak
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Russell E Lewis
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Voon Ong
- Cidara Therapeutics, San Diego, CA, USA
| | | | | |
Collapse
|
5
|
Sharma A, Singh G, Bhatti JS, Gill SK, Arya SK. Antifungal peptides: Therapeutic potential and challenges before their commercial success. Int J Biol Macromol 2025; 284:137957. [PMID: 39603306 DOI: 10.1016/j.ijbiomac.2024.137957] [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: 07/31/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Antifungal peptides (AFPs) are small cationic peptides that are found in a diverse range of taxa including bacteria, plants, mammals and insects. AFPs exhibit the strong antifungal activities against several pathogenic fungi, making them potential candidates for developing novel antifungal agents. AFP cause fungal cell death by rupturing the membranes of the fungal cell wall and inhibits the vital enzymes. Since AFPs are isolated from a range of natural sources, efforts are being made to create synthetic versions of these peptides with improved pharmacological properties. One of their key advantages is that they are less likely to develop resistance as compared to conventional antifungal medications. Although AFPs display immense potential as antifungal agents, challenges still exist in their stability, solubility, absorption, and time-consuming extraction process. Still, the possibility for AFPs to evolve into a novel class of antifungal medicine gives hope for improved treatments for fungal infections. This article offers the comprehensive information on AFPs origin, mode of action, prospective use in antifungal treatments. It also discusses about the application of antifungal peptides beyond the therapeutic field, such as in agriculture for crop protection, in food industry and in aquaculture field. It further elaborates on the challenges and potential paths associated with the progression of AFPs as advanced antifungal agents.
Collapse
Affiliation(s)
- Anindita Sharma
- Department of Biotechnology, Lovely Professional University, Phagwara, India
| | - Gursharan Singh
- Department of Medical Laboratory Sciences, Lovely Professional University, Phagwara, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine School of Health Sciences, Central University of Punjab, India
| | | | | |
Collapse
|
6
|
Smolobochkin A, Gazizov A, Appazov N, Sinyashin O, Burilov A. Progress in the Stereoselective Synthesis Methods of Pyrrolidine-Containing Drugs and Their Precursors. Int J Mol Sci 2024; 25:11158. [PMID: 39456938 PMCID: PMC11508981 DOI: 10.3390/ijms252011158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
The presented review systematizes and summarizes the data on the synthesis of pyrrolidine derivatives, which are precursors for obtaining drugs. Based on the analysis of published data, the most promising directions in the synthesis of biologically active compounds containing a pyrrolidine ring are identified. Stereoselective synthesis methods are classified based on the source of the pyrrolidine ring. The first group includes methods that use a pyrrolidine ring as the starting compound. The second group combines stereoselective methods of cyclization of acyclic starting compounds, which lead to optically pure pyrrolidine derivatives.
Collapse
Affiliation(s)
- Andrey Smolobochkin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, Kazan 420088, Russia; (A.G.); (O.S.); (A.B.)
| | - Almir Gazizov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, Kazan 420088, Russia; (A.G.); (O.S.); (A.B.)
| | - Nurbol Appazov
- Laboratory of Engineering Profile, Department of Engineering Technology, Korkyt Ata Kyzylorda University, Aiteke bi Str., 29A, Kyzylorda 120014, Kazakhstan
| | - Oleg Sinyashin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, Kazan 420088, Russia; (A.G.); (O.S.); (A.B.)
| | - Alexander Burilov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, Kazan 420088, Russia; (A.G.); (O.S.); (A.B.)
| |
Collapse
|
7
|
Mangrum MM, Vogel AK, Wagner AS, King AE, Miao J, Zhou Y, Phillips EK, Peters BM, Reynolds TB. Disruption to de novo uridine biosynthesis alters β-1,3-glucan masking in Candida albicans. mSphere 2024; 9:e0028724. [PMID: 39115319 PMCID: PMC11423711 DOI: 10.1128/msphere.00287-24] [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: 04/08/2024] [Accepted: 05/21/2024] [Indexed: 08/18/2024] Open
Abstract
The uridine derivatives UDP-glucose and UDP-N-acetylglucosamine are important for cell wall construction as they are the precursors for the synthesis of β-1,3-glucan and chitin, respectively. Previous studies have demonstrated attenuated virulence of uridine auxotrophs in mice, which has been attributed to insufficient uridine levels for growth in the host. We have discovered that uridine deprivation in the uridine auxotroph ura3ΔΔ disrupts cell wall architecture by increasing surface mannans, exposing β-1,3-glucan and chitin, and decreasing UDP-sugar levels. Cell wall architecture and UDP-sugars can be rescued with uridine supplementation. The cell wall architectural disruptions in the ura3ΔΔ mutant also impact immune activation since the mutant elicited greater TNFα secretion from RAW264.7 macrophages than wild type. To determine if cell wall defects contributed to decreased virulence in the ura3ΔΔ mutant, we used a murine model of systemic infection. Mice infected with the ura3ΔΔ mutant exhibited increased survival and reduced kidney fungal burden compared with mice infected with wild type. However, suppression of the immune response with cyclophosphamide did not rescue virulence in mice infected with the ura3ΔΔ mutant, indicating the attenuation in virulence of uridine auxotrophs can be attributed to decreased growth in the host but not increased exposure of β-1,3-glucan. Moreover, the ura3ΔΔ mutant is unable to grow on ex vivo kidney agar, which demonstrates its inability to colonize the kidneys due to poor growth. Thus, although uridine auxotrophy elicits changes to cell wall architecture that increase the exposure of immunogenic polymers, metabolic fitness costs more strongly drive the observed virulence attenuation.IMPORTANCECandida albicans is a common cause of bloodstream infections (candidemia). Treatment of these bloodstream infections is made difficult because of increasing antifungal resistance and drug toxicity. Thus, new tactics are needed for antifungal drug development, with immunotherapy being of particular interest. The cell wall of C. albicans is composed of highly immunogenic polymers, particularly β-1,3-glucan. However, β-1,3-glucan is naturally masked by an outer layer of mannoproteins, which hampers the detection of the fungus by the host immune system. Alteration in cell wall components has been shown to increase β-1,3-glucan exposure; however, it is unknown how the inability to synthesize precursors to cell wall components affects unmasking. Here, we demonstrate how cell wall architecture is altered in response to a deficit in precursors for cell wall synthesis and how uridine is a crucial component of these precursors.
Collapse
Affiliation(s)
- Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Amanda K. Vogel
- Integrated Program in Biomedical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Ainsley E. King
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Jian Miao
- Pharmaceutical Sciences Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yue Zhou
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Elise K. Phillips
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Brian M. Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
8
|
Costa PCQGD, Nogueira PL, Nascimento YMD, Sobral MV, Silvestre GFG, Castro RDD. Bioactive potential of Eugenia luschnathiana essential oil and extract: antifungal activity against Candida species isolated from oncological patients. BRAZ J BIOL 2024; 84:e286419. [PMID: 39292142 DOI: 10.1590/1519-6984.286419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 09/19/2024] Open
Abstract
Immunosuppressed individuals, including those undergoing cancer treatment, are more vulnerable to fungal infections, such as oral candidiasis, impacting their quality of life. Given the limitations of current therapies, the discovery of new antifungal agents, including those of natural origin, is crucial for the proper managing of these infections. We investigated the phytochemical profile and antifungal activity of both the essential oil and crude ethanolic extract (CEE) obtained from Eugenia luschnathiana against reference strains and clinical isolates of Candida from oncology patients. Toxicological characterization was also conducted. Gas chromatography coupled to mass spectrometry (GC-MS) and 1H Nuclear Magnetic Resonance (NMR) were used for phytochemical analysis. Antifungal evaluation was conducted to determine the Minimum Inhibitory Concentration (MIC) and Minimum Fungicidal Concentration (MFC); evaluation of potential mechanisms of action; activity on a fungal biofilm; evaluation of the cytotoxic effect on human keratinocytes of the HaCat lineage by the MTT method; determination of lethality for Artemia salina larvae. GC-MS identified a predominance of sesquiterpenes in the essential oil, notably (E)-Caryophyllene. The 1H NMR spectrum identified aliphatic, osidic, and aromatic compounds in the crude ethanolic extract. The essential oil showed no antifungal activity. However, the CEE exhibited fungicidal activity, with MIC and MFC ranging from 1.95 µg/mL to 3.90 µg/mL. The antifungal effect was affected by sorbitol, indicating a possible mechanism targeting fungal cell wall structures. At low concentration (19.5 µg/mL), the CEE inhibited 62,78% of C. albicans biofilm. The CEE demonstrated a promising toxicity profile, with an LC50 of 142.4 µg/mL against Artemia salina. In conclusion, the CEE from Eugenia luschnathiana exhibited potent antifungal activity, likely through cell wall disruption, biofilm inhibition, and a favorable toxicity profile for further exploration.
Collapse
Affiliation(s)
- P C Q G da Costa
- Universidade Federal da Paraíba - UFPB, Centro de Ciências da Saúde, Departamento de Clínica e Odontologia Social, Programa de Pós-graduação em Odontologia, João Pessoa, PB, Brasil
| | - P L Nogueira
- Universidade Federal da Paraíba - UFPB, Centro de Ciências da Saúde, Departamento de Clínica e Odontologia Social, Programa de Pós-graduação em Odontologia, João Pessoa, PB, Brasil
| | - Y M do Nascimento
- Universidade Federal da Paraíba - UFPB, Centro de Ciências Exatas e da Natureza, Departamento de Ciências Farmacêuticas, Programa de Pós-graduação em produtos naturais e sintéticos bioativos, João Pessoa, PB, Brasil
| | - M V Sobral
- Universidade Federal da Paraíba - UFPB, Centro de Ciências Exatas e da Natureza, Departamento de Ciências Farmacêuticas, Programa de Pós-graduação em produtos naturais e sintéticos bioativos, João Pessoa, PB, Brasil
| | - G F G Silvestre
- Universidade Federal da Paraíba - UFPB, Centro de Ciências Exatas e da Natureza, Departamento de Ciências Farmacêuticas, Programa de Pós-graduação em produtos naturais e sintéticos bioativos, João Pessoa, PB, Brasil
| | - R D de Castro
- Universidade Federal da Paraíba - UFPB, Centro de Ciências da Saúde, Departamento de Clínica e Odontologia Social, Programa de Pós-graduação em Odontologia, João Pessoa, PB, Brasil
| |
Collapse
|
9
|
Holzknecht J, Marx F. Navigating the fungal battlefield: cysteine-rich antifungal proteins and peptides from Eurotiales. FRONTIERS IN FUNGAL BIOLOGY 2024; 5:1451455. [PMID: 39323611 PMCID: PMC11423270 DOI: 10.3389/ffunb.2024.1451455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024]
Abstract
Fungi are ubiquitous in the environment and play a key role in the decomposition and recycling of nutrients. On the one hand, their special properties are a great asset for the agricultural and industrial sector, as they are used as source of nutrients, producers of enzymes, pigments, flavorings, and biocontrol agents, and in food processing, bio-remediation and plant growth promotion. On the other hand, they pose a serious challenge to our lives and the environment, as they are responsible for fungal infections in plants, animals and humans. Although host immunity opposes invading pathogens, certain factors favor the manifestation of fungal diseases. The prevalence of fungal infections is on the rise, and there is an alarming increase in the resistance of fungal pathogens to approved drugs. The limited number of antimycotics, the obstacles encountered in the development of new drugs due to the poor tolerability of antifungal agents in patients, the limited number of unique antifungal targets, and the low species specificity contribute to the gradual depletion of the antifungal pipeline and newly discovered antifungal drugs are rare. Promising candidates as next-generation therapeutics are antimicrobial proteins and peptides (AMPs) produced by numerous prokaryotic and eukaryotic organisms belonging to all kingdom classes. Importantly, filamentous fungi from the order Eurotiales have been shown to be a rich source of AMPs with specific antifungal activity. A growing number of published studies reflects the efforts made in the search for new antifungal proteins and peptides (AFPs), their efficacy, species specificity and applicability. In this review, we discuss important aspects related to fungi, their impact on our life and issues involved in treating fungal infections in plants, animals and humans. We specifically highlight the potential of AFPs from Eurotiales as promising alternative antifungal therapeutics. This article provides insight into the structural features, mode of action, and progress made toward their potential application in a clinical and agricultural setting. It also identifies the challenges that must be overcome in order to develop AFPs into therapeutics.
Collapse
Affiliation(s)
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Innsbruck Medical University,
Innsbruck, Austria
| |
Collapse
|
10
|
Padmavathi AR, Reddy GKK, Murthy PS, Nancharaiah YV. New arsenals for old armour: Biogenic nanoparticles in the battle against drug-resistant Candidaalbicans. Microb Pathog 2024; 194:106800. [PMID: 39025380 DOI: 10.1016/j.micpath.2024.106800] [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: 04/29/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Candida albicans is a common commensal fungus and fourth most frequent causative agent of nosocomial infections including life-threatening invasive candidiasis in humans. The effectiveness of present antifungal therapies using azoles, polyenes, flucytosine and echinocandins has plateaued in managing fungal infections. The limitations of these antifungal drugs are related to polymorphic morphology, biofilm formation, emergence of drug-resistant strains and production of several virulence factors. Development of new antifungal agents, which can particularly afflict multiple cellular targets and limiting evolving resistant strains are needed. Recently, metal nanoparticles have emerged as a source of new antifungal agents for antifungal formulations. Furthermore, green nanotechnology deals with the use of biosynthetic routes that offer new avenue for synthesizing antifungal nanoparticles coupled with less toxic chemical inventory and environmental sustainability. This article reviews the recent developments on C. albicans pathogenesis, biofilm formation, drug resistance, mode of action of antifungal drugs and antifungal activities of metal nanoparticles. The antifungal efficacy and mode of action of metal nanoparticles are described in the context of prospective therapeutic applications.
Collapse
Affiliation(s)
- Alwar Ramanujam Padmavathi
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India.
| | - G Kiran Kumar Reddy
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India
| | - P Sriyutha Murthy
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India
| | - Y V Nancharaiah
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India
| |
Collapse
|
11
|
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
|
12
|
Gautam I, Yarava JR, Xu Y, Li R, Scott FJ, Mentink-Vigier F, Momany M, Latgé JP, Wang T. Comparative Analysis of Polysaccharide and Cell Wall Structure in Aspergillus nidulans and Aspergillus fumigatus by Solid-State NMR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607833. [PMID: 39185159 PMCID: PMC11343165 DOI: 10.1101/2024.08.13.607833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Invasive aspergillosis poses a significant threat to immunocompromised patients, leading to high mortality rates associated with these infections. Targeting the biosynthesis of cell wall carbohydrates is a promising strategy for antifungal drug development and will be advanced by a molecular-level understanding of the native structures of polysaccharides within their cellular context. Solid-state NMR spectroscopy has recently provided detailed insights into the cell wall organization of Aspergillus fumigatus, but genetic and biochemical evidence highlights species-specific differences among Aspergillus species. In this study, we employed a combination of 13C, 15N, and 1H-detection solid-state NMR, supplemented by Dynamic Nuclear Polarization (DNP), to compare the structural organization of cell wall polymers and their assembly in the cell walls of A. fumigatus and A. nidulans, both of which are key model organisms and human pathogens. The two species exhibited a similar rigid core architecture, consisting of chitin, α-glucan, and β-glucan, which contributed to comparable cell wall properties, including polymer dynamics, water retention, and supramolecular organization. However, differences were observed in the chitin, galactosaminogalactan, protein, and lipid content, as well as in the dynamics of galactomannan and the structure of the glucan matrix.
Collapse
Affiliation(s)
- Isha Gautam
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | | | - Yifan Xu
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Reina Li
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Faith J. Scott
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | | | - Michelle Momany
- Department of Plant Biology, University of Georgia, Athens, GA, USA
| | - Jean-Paul Latgé
- Institute of Molecular Biology and Biotechnology, University of Crete, Heraklion, Greece
- Fungal Respiratory Infections Research Unit and SFR ICAT, University of Angers, France
| | - Tuo Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
13
|
Dickwella Widanage MC, Gautam I, Sarkar D, Mentink-Vigier F, Vermaas JV, Ding SY, Lipton AS, Fontaine T, Latgé JP, Wang P, Wang T. Adaptative survival of Aspergillus fumigatus to echinocandins arises from cell wall remodeling beyond β-1,3-glucan synthesis inhibition. Nat Commun 2024; 15:6382. [PMID: 39085213 PMCID: PMC11291495 DOI: 10.1038/s41467-024-50799-8] [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: 05/13/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Antifungal echinocandins inhibit the biosynthesis of β-1,3-glucan, a major and essential polysaccharide component of the fungal cell wall. However, the efficacy of echinocandins against the pathogen Aspergillus fumigatus is limited. Here, we use solid-state nuclear magnetic resonance (ssNMR) and other techniques to show that echinocandins induce dynamic changes in the assembly of mobile and rigid polymers within the A. fumigatus cell wall. The reduction of β-1,3-glucan induced by echinocandins is accompanied by a concurrent increase in levels of chitin, chitosan, and highly polymorphic α-1,3-glucans, whose physical association with chitin maintains cell wall integrity and modulates water permeability. The rearrangement of the macromolecular network is dynamic and controls the permeability and circulation of the drug throughout the cell wall. Thus, our results indicate that echinocandin treatment triggers compensatory rearrangements in the cell wall that may help A. fumigatus to tolerate the drugs' antifungal effects.
Collapse
Affiliation(s)
- Malitha C Dickwella Widanage
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
- National High Magnetic Field Laboratory, Tallahassee, FL, USA
| | - Isha Gautam
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | | | | | - Josh V Vermaas
- MSU-DOE Plant Research Laboratory, East Lansing, MI, USA
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Shi-You Ding
- Department of Plant Biology, Michigan State University, East Lansing, MI, USA
| | - Andrew S Lipton
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Thierry Fontaine
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie et Pathogénicité Fongiques, F-, 75015, Paris, France
| | - Jean-Paul Latgé
- Institute of Molecular Biology and Biotechnology, University of Crete, Heraklion, Greece
| | - Ping Wang
- Departments of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Tuo Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
14
|
Jiang J, Keniya MV, Puri A, Zhan X, Cheng J, Wang H, Lin G, Lee YK, Jaber N, Hassoun Y, Shor E, Shi Z, Lee SH, Xu M, Perlin DS, Dai W. Structural and Biophysical Dynamics of Fungal Plasma Membrane Proteins and Implications for Echinocandin Action in Candida glabrata. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596243. [PMID: 38854035 PMCID: PMC11160696 DOI: 10.1101/2024.05.29.596243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Fungal plasma membrane proteins represent key therapeutic targets for antifungal agents, yet their structure and spatial distribution in the native context remain poorly characterized. Herein, we employ an integrative multimodal approach to elucidate the structural and functional organization of plasma membrane protein complexes in Candida glabrata , focusing on prominent and essential membrane proteins, the polysaccharide synthase β-(1,3)-glucan synthase (GS) and the proton pump Pma1. Cryo-electron tomography (cryo-ET) and live cell imaging reveal that GS and Pma1 are heterogeneously distributed into distinct plasma membrane microdomains. Treatment with caspofungin, an echinocandin antifungal that targets GS, alters the plasma membrane and disrupts the native distribution of GS and Pma1. Based on these findings, we propose a model for echinocandin action that considers how drug interactions with the plasma membrane environment lead to inhibition of GS. Our work underscores the importance of interrogating the structural and dynamic characteristics of fungal plasma membrane proteins in situ to understand function and facilitate precisely targeted development of novel antifungal therapies.
Collapse
|
15
|
Xu D, Wang M, Zhang X, Mao H, Xu H, Zhang B, Zeng X, Li F. The Putative Cytochrome b5 Domain-Containing Protein CaDap1 Homologue Is Involved in Antifungal Drug Tolerance, Cell Wall Chitin Maintenance, and Virulence in Candida albicans. J Fungi (Basel) 2024; 10:316. [PMID: 38786671 PMCID: PMC11122062 DOI: 10.3390/jof10050316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Candida albicans (Ca), a prominent opportunistic fungal pathogen in humans, has garnered considerable attention due to its infectious properties. Herein, we have identified and characterized CaCDAP1 (Ca orf19.1034), a homolog of ScDAP1 found in Saccharomyces cerevisiae. CaCDAP1 encodes a 183-amino acid protein with a conserved cytochrome b5-like heme-binding domain. The deletion of CaDAP1 renders Ca cells susceptible to caspofungin and terbinafine. CaDAP1 deletion confers resistance to Congo Red and Calcofluor White, and sensitivity to sodium dodecyl sulfate. The deletion of CaDAP1 results in a 50% reduction in chitin content within the cell wall, the downregulation of phosphorylation levels in CaMkc1, and the upregulation of phosphorylation levels in CaCek1. Notably, CaDAP1 deletion results in the abnormal hyphal development of Ca cells and diminishes virulence in a mouse systemic infection model. Thus, CaDAP1 emerges as a critical regulator governing cellular responses to antifungal drugs, the synthesis of cell wall chitin, and virulence in Ca.
Collapse
Affiliation(s)
- Dayong Xu
- College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (M.W.); (X.Z.); (H.M.); (H.X.); (B.Z.); (X.Z.)
| | | | | | | | | | | | | | - Feng Li
- College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (M.W.); (X.Z.); (H.M.); (H.X.); (B.Z.); (X.Z.)
| |
Collapse
|
16
|
Mehravar S, Leite GS, Pimentel M, Rezaie A. Antifungal effects of echinocandins diminish when exposed to intestinal lumen contents: a finding with potentially significant clinical implications. Front Pharmacol 2024; 15:1376656. [PMID: 38601473 PMCID: PMC11004442 DOI: 10.3389/fphar.2024.1376656] [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: 01/25/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Echinocandins, a prominent class of antifungals, are known for their broad-spectrum activity and favorable safety profiles. However, their bioavailability and efficacy via oral route are suboptimal. In this study, caspofungin and micafungin, the two most commonly used echinocandins, were evaluated in various in vitro environments simulating intestinal lumen. The results revealed that while both antifungals are effective in standard medium, their efficacy significantly diminishes in the presence of human small bowel aspirates and bovine bile. The study suggests that bowel contents and specifically bile acids may be a suppressive component, hindering the antifungal effects of echinocandins. This novel exploration sheds light on the poor oral bioavailability of echinocandins. The findings imply that echinocandins alone, regardless of administration route, may not be optimal for gastrointestinal (GI) fungal infections or invasive fungal infections originating from intestinal translocation. Further clinical investigations are warranted to validate and expand upon these observations.
Collapse
Affiliation(s)
- Sepideh Mehravar
- Medically Associated Science and Technology (MAST) Program, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Gabriela S. Leite
- Medically Associated Science and Technology (MAST) Program, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars Sinai Medical Center, Los Angeles, CA, United States
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Ali Rezaie
- Medically Associated Science and Technology (MAST) Program, Cedars Sinai Medical Center, Los Angeles, CA, United States
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
17
|
Duarte ABS, Perez-Castillo Y, da Nóbrega Alves D, de Castro RD, de Souza RL, de Sousa DP, Oliveira EE. Antifungal activity against Candida albicans of methyl 3,5-dinitrobenzoate loaded nanoemulsion. Braz J Microbiol 2024; 55:25-39. [PMID: 38135805 PMCID: PMC10920570 DOI: 10.1007/s42770-023-01214-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The objective of this study was to evaluate the antifungal activity of free methyl 3,5 dinitrobenzoate (MDNB) and its nanoemulsion (MDNB-NE) against strains of Candida albicans. Additionally, a molecular modeling study was also carried out to propose the mechanism of action and toxicity of MDNB. These results demonstrated the MDNB-NE presented a droplet size of 181.16 ± 3.20 nm and polydispersity index of 0.30 ± 0.03. MDNB and MDNB-NE inhibited the growth of all strains with minimum inhibitory concentrations of 0.27-1.10 mM. The biological results corroborated the molecular model, which pointed to a multi-target antifungal mechanism of action for MDNB in C. albicans. The study could serve as a basis for further research involving compounds with nitro groups with antifungal.
Collapse
Affiliation(s)
- Allana Brunna Sucupira Duarte
- Post Graduation Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Yunierkis Perez-Castillo
- Bio-Cheminformatics Research Group and Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Quito, Ecuador
| | - Danielle da Nóbrega Alves
- Laboratory of Experimental Pharmacology and Cell Culture, Department of Clinical and Social Dentistry, Federal University of Paraíba, João Pessoa, Brazil
| | - Ricardo Dias de Castro
- Laboratory of Experimental Pharmacology and Cell Culture, Department of Clinical and Social Dentistry, Federal University of Paraíba, João Pessoa, Brazil
| | | | | | - Elquio Eleamen Oliveira
- Laboratory of Synthesis and Drug Delivery, State University of Paraíba, João Pessoa, Brazil.
| |
Collapse
|
18
|
Jospe-Kaufman M, Ben-Zeev E, Mottola A, Dukhovny A, Berman J, Carmeli S, Fridman M. Reshaping Echinocandin Antifungal Drugs To Circumvent Glucan Synthase Point-Mutation-Mediated Resistance. Angew Chem Int Ed Engl 2024; 63:e202314728. [PMID: 38161189 DOI: 10.1002/anie.202314728] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Echinocandins are a class of antifungal drugs that inhibit the activity of the β-(1,3)-glucan synthase complex, which synthesizes fungal cell wall β-(1,3)-glucan. Echinocandin resistance is linked to mutations in the FKS gene, which encodes the catalytic subunit of the glucan synthase complex. We present a molecular-docking-based model that provides insight into how echinocandins interact with the target Fks protein: echinocandins form a ternary complex with both Fks and membrane lipids. We used reductive dehydration of alcohols to generate dehydroxylated echinocandin derivatives and evaluated their potency against a panel of Candida pathogens constructed by introducing resistance-conferring mutations in the FKS gene. We found that removing the hemiaminal alcohol, which drives significant conformational alterations in the modified echinocandins, reduced their efficacy. Conversely, eliminating the benzylic alcohol of echinocandins enhanced potency by up to two orders of magnitude, in a manner dependent upon the resistance-conferring mutation. Strains that have developed resistance to either rezafungin, the most recently clinically approved echinocandin, or its dehydroxylated derivative RZF-1, exhibit high resistance to rezafungin while demonstrating moderate resistance to RZF-1. These findings provide valuable insight for combating echinocandin resistance through chemical modifications.
Collapse
Affiliation(s)
- Moriah Jospe-Kaufman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Efrat Ben-Zeev
- The Whol Drug Discovery institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Austin Mottola
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Anna Dukhovny
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Shmuel Carmeli
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| |
Collapse
|
19
|
Feng Z, Lu H, Jiang Y. Promising immunotherapeutic targets for treating candidiasis. Front Cell Infect Microbiol 2024; 14:1339501. [PMID: 38404288 PMCID: PMC10884116 DOI: 10.3389/fcimb.2024.1339501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
In the last twenty years, there has been a significant increase in invasive fungal infections, which has corresponded with the expanding population of individuals with compromised immune systems. As a result, the mortality rate linked to these infections remains unacceptably high. The currently available antifungal drugs, such as azoles, polyenes, and echinocandins, face limitations in terms of their diversity, the escalating resistance of fungi and the occurrence of significant adverse effects. Consequently, there is an urgent need to develop new antifungal medications. Vaccines and antibodies present a promising avenue for addressing fungal infections due to their targeted antifungal properties and ability to modulate the immune response. This review investigates the structure and function of cell wall proteins, secreted proteins, and functional proteins within C. albicans. Furthermore, it seeks to analyze the current advancements and challenges in macromolecular drugs to identify new targets for the effective management of candidiasis.
Collapse
Affiliation(s)
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Lefranc M, Accoceberry I, Fitton-Ouhabi V, Biteau N, Noël T. Rapamycin and caspofungin show synergistic antifungal effects in caspofungin-susceptible and caspofungin-resistant Candida strains in vitro. J Antimicrob Chemother 2024; 79:151-156. [PMID: 37991226 DOI: 10.1093/jac/dkad359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVES Caspofungin is an echinocandin antifungal agent that inhibits synthesis of glucan required for the fungal cell wall. Resistance is mediated by mutation of Fks1 glucan synthase, among which S645P is the most common resistance-associated polymorphism. Rapamycin is a macrolide that inhibits the mechanistic target of rapamycin (mTOR) protein kinase activity. This study investigated the interaction between rapamycin and caspofungin in inhibiting the growth of WT Candida albicans and Fks1 S645P mutant clinical isolate, and WT Candida lusitaniae and genetically engineered isogenic strain with Fks1 S645P mutation at equivalent position. METHODS Interactions between caspofungin and rapamycin were evaluated using the microdilution chequerboard method in liquid medium. The results were analysed using the Loewe additivity model (FIC index, FICI) and the Bliss independence model (response surface, RS, analysis). RESULTS Synergy between rapamycin and caspofungin was shown for C. albicans and C. lusitaniae strains by RS analysis of the chequerboard tests. Synergy was observed in strains susceptible and resistant to caspofungin. Weak subinhibitory concentrations of rapamycin were sufficient to restore caspofungin susceptibility. CONCLUSIONS We report here, for the first time, synergy between caspofungin and rapamycin in Candida species. Synergy was shown for strains susceptible and resistant to caspofungin. This study highlights the possible implication of the TOR pathway in sensing antifungal-mediated cell wall stress and in modulating the cellular response to echinocandins in Candida yeasts.
Collapse
Affiliation(s)
- Maxime Lefranc
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, CHU Bordeaux, 33000 Bordeaux, France
| | - Isabelle Accoceberry
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, CHU Bordeaux, 33000 Bordeaux, France
| | - Valérie Fitton-Ouhabi
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
| | - Nicolas Biteau
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
| | - Thierry Noël
- University Bordeaux, CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, 33000 Bordeaux, France
| |
Collapse
|
21
|
Li L, Wu H, Wang J, Ji Z, Fang T, Lu H, Yan L, Shen F, Zhang D, Jiang Y, Ni T. Discovery of Novel 8-Hydroxyquinoline Derivatives with Potent In Vitro and In Vivo Antifungal Activity. J Med Chem 2023; 66:16364-16376. [PMID: 37975824 DOI: 10.1021/acs.jmedchem.3c01771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Fungal pathogens can cause life-threatening infections, yet current antifungals are inadequate at treating many of these, highlighting the importance of novel drug discovery. Here, we report hit compound L14, a novel 8-hydroxyquinoline derivative with potent and broad-spectrum antifungal activity. In vitro experiments exhibited that L14 had better activity and lower cytotoxicity than that of clioquinol and showed synergy in combination with fluconazole (FLC). In a Candida albicans-infected murine model, L14 at 2 mg/kg showed better in vivo efficacy than clioquinol at reducing fungal burden and extending the survival of C. albicans-infected mice. In addition, L14 alone or in combination with FLC had significant inhibitory activity against hypha and biofilm formation. Overall, our data indicated that 8-hydroxyquinoline derivative L14 has favorable pharmacokinetics and acceptable safety profiles and could be further investigated as a promising antifungal hit compound.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Hao Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Jiayin Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou 350112, China
| | - Zhe Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Ting Fang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Lan Yan
- School of Pharmacy, Naval Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Fuming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Dazhi Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
- School of Pharmacy, Naval Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
22
|
Ning Y, Xiao M, Perlin DS, Zhao Y, Lu M, Li Y, Luo Z, Dai R, Li S, Xu J, Liu L, He H, Liu Y, Li F, Guo Y, Chen Z, Xu Y, Sun T, Zhang L. Decreased echinocandin susceptibility in Candida parapsilosis causing candidemia and emergence of a pan-echinocandin resistant case in China. Emerg Microbes Infect 2023; 12:2153086. [PMID: 36440795 PMCID: PMC9793909 DOI: 10.1080/22221751.2022.2153086] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022]
Abstract
Candida parapsilosis is becoming a predominant non-albicans cause of invasive candidiasis (IC). Echinocandins are the preferred choice for IC treatment and prophylaxis. Resistance to echinocandins in C. parapsilosis has emerged in several countries, but little is known about the susceptibility profile in China or about mechanisms of resistance. Here, we investigated the echinocandin susceptibilities of 2523 C. parapsilosis isolates collected from China and further explored the resistance mechanism among echinocandin-resistant isolates. Anidulafungin exhibited the highest MICs (MIC50/90, 1 and 2 µg/mL; GM, 0.948 µg/mL), while caspofungin showed better activity (0.5 and 1 µg/mL; 0.498 µg/mL). Significantly higher echinocandin MICs were observed among blood-derived isolates compared to others, especially for caspofungin (GM, 1.348 µg/mL vs 0.478 µg/mL). Isolates from ICU and surgical wards also showed higher MICs. Twenty isolates showed intermediate phenotypes for at least one echinocandin. One was resistant to all three echinocandins, fluconazole and voriconazole, which caused breakthrough IC during long-term exposure to micafungin. WGS revealed this isolate carried a mutation S656P in hotspot1 region of Fks1. Bioinformatics analyses suggested that this mutation might lead to an altered protein conformation. CRISPR Cas9-mediated introduction of this mutation into a susceptible reference C. parapsilosis strain increased MICs of all echinocandins 64-fold, with similar results found in the subspecies, C. orthopsilosis and C. metapsilosis. This is the first report of a multi-azole resistant and pan-echinocandin resistant C. parapsilosis isolate, and the identification of a FKS1S656P conferring pan-echinocandin resistance. Our study underscores the necessity of rigorous management of antifungal use and of monitoring for antifungal susceptibility.
Collapse
Affiliation(s)
- Yating Ning
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| | - Meng Xiao
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Yanan Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Minya Lu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| | - Yi Li
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| | - Zhengyu Luo
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| | - Rongchen Dai
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Shengjie Li
- Medical Research Centre, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People’s Republic of China
| | - Jiajun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Lingli Liu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Hong He
- Department of Clinical Laboratory, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yun Liu
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Fushun Li
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yuguang Guo
- Department of Laboratory Medicine, Liaoning Provincial People’s Hospital, Shenyang, People’s Republic of China
| | - Zhongju Chen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| | - Tianshu Sun
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
- Medical Research Centre, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People’s Republic of China
| | - Li Zhang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
| |
Collapse
|
23
|
Feng Y, Lu H, Whiteway M, Jiang Y. Understanding fluconazole tolerance in Candida albicans: implications for effective treatment of candidiasis and combating invasive fungal infections. J Glob Antimicrob Resist 2023; 35:314-321. [PMID: 37918789 DOI: 10.1016/j.jgar.2023.10.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/07/2023] [Accepted: 10/22/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVES Fluconazole (FLC) tolerant phenotypes in Candida species contribute to persistent candidemia and the emergence of FLC resistance. Therefore, making FLC fungicidal and eliminating FLC tolerance are important for treating invasive fungal diseases (IFDs) caused by Candida species. However, the mechanisms of FLC tolerance in Candida species remain to be fully explored. METHODS This review discusses the high incidence of FLC tolerance in Candida species and the importance of successfully clearing FLC tolerance in treating candidiasis. We further define and characterize FLC tolerance in C. albicans. RESULTS This review identifies global factors affecting FLC tolerance and suggest that FLC tolerance is a strategy of C. albicans response to FLC damage whose mechanism differs from FLC resistance. CONCLUSIONS This review highlights the significance of the cell membrane and cell wall integrity in FLC tolerance, guiding approaches to combat IFDs caused by Candida species..
Collapse
Affiliation(s)
- Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | | | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
24
|
Abstract
Candida auris is a multidrug-resistant fungal pathogen that presents a serious threat to global human health. Since the first reported case in 2009 in Japan, C. auris infections have been reported in more than 40 countries, with mortality rates between 30% and 60%. In addition, C. auris has the potential to cause outbreaks in health care settings, especially in nursing homes for elderly patients, owing to its efficient transmission via skin-to-skin contact. Most importantly, C. auris is the first fungal pathogen to show pronounced and sometimes untreatable clinical drug resistance to all known antifungal classes, including azoles, amphotericin B, and echinocandins. In this review, we explore the causes of the rapid spread of C. auris. We also highlight its genome organization and drug resistance mechanisms and propose future research directions that should be undertaken to curb the spread of this multidrug-resistant pathogen.
Collapse
Affiliation(s)
- Anuradha Chowdhary
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India;
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Kusum Jain
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India;
| | - Neeraj Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
25
|
Wagner AS, Lumsdaine SW, Mangrum MM, Reynolds TB. Caspofungin-induced β(1,3)-glucan exposure in Candida albicans is driven by increased chitin levels. mBio 2023; 14:e0007423. [PMID: 37377417 PMCID: PMC10470516 DOI: 10.1128/mbio.00074-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/04/2023] [Indexed: 06/29/2023] Open
Abstract
To successfully induce disease, Candida albicans must effectively evade the host immune system. One mechanism used by C. albicans to achieve this is to mask immunogenic β(1,3)-glucan epitopes within its cell wall under an outer layer of mannosylated glycoproteins. Consequently, induction of β(1,3)-glucan exposure (unmasking) via genetic or chemical manipulation increases fungal recognition by host immune cells in vitro and attenuates disease during systemic infection in mice. Treatment with the echinocandin caspofungin is one of the most potent drivers of β(1,3)-glucan exposure. Several reports using murine infection models suggest a role for the immune system, and specifically host β(1,3)-glucan receptors, in mediating the efficacy of echinocandin treatment in vivo. However, the mechanism by which caspofungin-induced unmasking occurs is not well understood. In this report, we show that foci of unmasking co-localize with areas of increased chitin within the yeast cell wall in response to caspofungin, and that inhibition of chitin synthesis via nikkomycin Z attenuates caspofungin-induced β(1,3)-glucan exposure. Furthermore, we find that both the calcineurin and Mkc1 mitogen-activated protein kinase pathways work synergistically to regulate β(1,3)-glucan exposure and chitin synthesis in response to drug treatment. When either of these pathways are interrupted, it results in a bimodal population of cells containing either high or low chitin content. Importantly, increased unmasking correlates with increased chitin content within these cells. Microscopy further indicates that caspofungin-induced unmasking correlates with actively growing cells. Collectively, our work presents a model in which chitin synthesis induces unmasking within the cell wall in response to caspofungin in growing cells. IMPORTANCE Systemic candidiasis has reported mortality rates ranging from 20% to 40%. The echinocandins, including caspofungin, are first-line antifungals used to treat systemic candidiasis. However, studies in mice have shown that echinocandin efficacy relies on both its cidal impacts on Candida albicans, as well as a functional immune system to successfully clear invading fungi. In addition to direct C. albicans killing, caspofungin increases exposure (unmasking) of immunogenic β(1,3)-glucan moieties. To evade immune detection, β(1,3)-glucan is normally masked within the C. albicans cell wall. Consequently, unmasked β(1,3)-glucan renders these cells more visible to the host immune system and attenuates disease progression. Therefore, discovery of how caspofungin-induced unmasking occurs is needed to elucidate how the drug facilitates host immune system-mediated clearance in vivo. We report a strong and consistent correlation between chitin deposition and unmasking in response to caspofungin and propose a model in which altered chitin synthesis drives increased unmasking during drug exposure.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | | | - Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
26
|
Lu H, Hong T, Jiang Y, Whiteway M, Zhang S. Candidiasis: From cutaneous to systemic, new perspectives of potential targets and therapeutic strategies. Adv Drug Deliv Rev 2023; 199:114960. [PMID: 37307922 DOI: 10.1016/j.addr.2023.114960] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Candidiasis is an infection caused by fungi from a Candida species, most commonly Candida albicans. C. albicans is an opportunistic fungal pathogen typically residing on human skin and mucous membranes of the mouth, intestines or vagina. It can cause a wide variety of mucocutaneous barrier and systemic infections; and becomes a severe health problem in HIV/AIDS patients and in individuals who are immunocompromised following chemotherapy, treatment with immunosuppressive agents or after antibiotic-induced dysbiosis. However, the immune mechanism of host resistance to C. albicans infection is not fully understood, there are a limited number of therapeutic antifungal drugs for candidiasis, and these have disadvantages that limit their clinical application. Therefore, it is urgent to uncover the immune mechanisms of the host protecting against candidiasis and to develop new antifungal strategies. This review synthesizes current knowledge of host immune defense mechanisms from cutaneous candidiasis to invasive C. albicans infection and documents promising insights for treating candidiasis through inhibitors of potential antifungal target proteins.
Collapse
Affiliation(s)
- Hui Lu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Ting Hong
- Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada.
| | - Shiqun Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Osset-Trénor P, Pascual-Ahuir A, Proft M. Fungal Drug Response and Antimicrobial Resistance. J Fungi (Basel) 2023; 9:jof9050565. [PMID: 37233275 DOI: 10.3390/jof9050565] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
Antifungal resistance is a growing concern as it poses a significant threat to public health. Fungal infections are a significant cause of morbidity and mortality, especially in immunocompromised individuals. The limited number of antifungal agents and the emergence of resistance have led to a critical need to understand the mechanisms of antifungal drug resistance. This review provides an overview of the importance of antifungal resistance, the classes of antifungal agents, and their mode of action. It highlights the molecular mechanisms of antifungal drug resistance, including alterations in drug modification, activation, and availability. In addition, the review discusses the response to drugs via the regulation of multidrug efflux systems and antifungal drug-target interactions. We emphasize the importance of understanding the molecular mechanisms of antifungal drug resistance to develop effective strategies to combat the emergence of resistance and highlight the need for continued research to identify new targets for antifungal drug development and explore alternative therapeutic options to overcome resistance. Overall, an understanding of antifungal drug resistance and its mechanisms will be indispensable for the field of antifungal drug development and clinical management of fungal infections.
Collapse
Affiliation(s)
- Paloma Osset-Trénor
- Department of Biotechnology, Instituto de Biología Molecular y Celular de Plantas IBMCP, Universidad Politécnica de Valencia, 46022 Valencia, Spain
| | - Amparo Pascual-Ahuir
- Department of Biotechnology, Instituto de Biología Molecular y Celular de Plantas IBMCP, Universidad Politécnica de Valencia, 46022 Valencia, Spain
| | - Markus Proft
- Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia IBV-CSIC, Consejo Superior de Investigaciones Científicas CSIC, 46010 Valencia, Spain
| |
Collapse
|
28
|
Dos Reis TF, de Castro PA, Bastos RW, Pinzan CF, Souza PFN, Ackloo S, Hossain MA, Drewry DH, Alkhazraji S, Ibrahim AS, Jo H, Lightfoot JD, Adams EM, Fuller KK, deGrado WF, Goldman GH. A host defense peptide mimetic, brilacidin, potentiates caspofungin antifungal activity against human pathogenic fungi. Nat Commun 2023; 14:2052. [PMID: 37045836 PMCID: PMC10090755 DOI: 10.1038/s41467-023-37573-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
Fungal infections cause more than 1.5 million deaths a year. Due to emerging antifungal drug resistance, novel strategies are urgently needed to combat life-threatening fungal diseases. Here, we identify the host defense peptide mimetic, brilacidin (BRI) as a synergizer with caspofungin (CAS) against CAS-sensitive and CAS-resistant isolates of Aspergillus fumigatus, Candida albicans, C. auris, and CAS-intrinsically resistant Cryptococcus neoformans. BRI also potentiates azoles against A. fumigatus and several Mucorales fungi. BRI acts in A. fumigatus by affecting cell wall integrity pathway and cell membrane potential. BRI combined with CAS significantly clears A. fumigatus lung infection in an immunosuppressed murine model of invasive pulmonary aspergillosis. BRI alone also decreases A. fumigatus fungal burden and ablates disease development in a murine model of fungal keratitis. Our results indicate that combinations of BRI and antifungal drugs in clinical use are likely to improve the treatment outcome of aspergillosis and other fungal infections.
Collapse
Affiliation(s)
- Thaila Fernanda Dos Reis
- 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
| | - Rafael Wesley Bastos
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Camila Figueiredo Pinzan
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Pedro F N Souza
- Visiting professor at Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Suzanne Ackloo
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, Suite 700, Toronto, ON, M5G 1L7, Canada
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - David Harold Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sondus Alkhazraji
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, 90502, USA
| | - Ashraf S Ibrahim
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, 90502, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Jorge D Lightfoot
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Emily M Adams
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Kevin K Fuller
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - William F deGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Gustavo H Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
29
|
Rabaan AA, Sulaiman T, Al-Ahmed SH, Buhaliqah ZA, Buhaliqah AA, AlYuosof B, Alfaresi M, Al Fares MA, Alwarthan S, Alkathlan MS, Almaghrabi RS, Abuzaid AA, Altowaileb JA, Al Ibrahim M, AlSalman EM, Alsalman F, Alghounaim M, Bueid AS, Al-Omari A, Mohapatra RK. Potential Strategies to Control the Risk of Antifungal Resistance in Humans: A Comprehensive Review. Antibiotics (Basel) 2023; 12:antibiotics12030608. [PMID: 36978475 PMCID: PMC10045400 DOI: 10.3390/antibiotics12030608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/30/2023] Open
Abstract
Fungal infections are becoming one of the main causes of morbidity and mortality in people with weakened immune systems. Mycoses are becoming more common, despite greater knowledge and better treatment methods, due to the regular emergence of resistance to the antifungal medications used in clinical settings. Antifungal therapy is the mainstay of patient management for acute and chronic mycoses. However, the limited availability of antifungal drug classes limits the range of available treatments. Additionally, several drawbacks to treating mycoses include unfavourable side effects, a limited activity spectrum, a paucity of targets, and fungal resistance, all of which continue to be significant issues in developing antifungal drugs. The emergence of antifungal drug resistance has eliminated accessible drug classes as treatment choices, which significantly compromises the clinical management of fungal illnesses. In some situations, the emergence of strains resistant to many antifungal medications is a major concern. Although new medications have been developed to address this issue, antifungal drug resistance has grown more pronounced, particularly in patients who need long-term care or are undergoing antifungal prophylaxis. Moreover, the mechanisms that cause resistance must be well understood, including modifications in drug target affinities and abundances, along with biofilms and efflux pumps that diminish intracellular drug levels, to find novel antifungal drugs and drug targets. In this review, different classes of antifungal agents, and their resistance mechanisms, have been discussed. The latter part of the review focuses on the strategies by which we can overcome this serious issue of antifungal resistance in humans.
Collapse
Affiliation(s)
- Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Tarek Sulaiman
- Infectious Diseases Section, Medical Specialties Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Shamsah H Al-Ahmed
- Specialty Paediatric Medicine, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Zainab A Buhaliqah
- Department of Family Medicine, Primary Healthcare Center, Dammam 32433, Saudi Arabia
| | - Ali A Buhaliqah
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Buthina AlYuosof
- Directorate of Public Health, Dammam Network, Eastern Health Cluster, Dammam 31444, Saudi Arabia
| | - Mubarak Alfaresi
- Department of Pathology and Laboratory Medicine, Zayed Military Hospital, Abu Dhabi 3740, United Arab Emirates
- Department of Pathology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Mona A Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mohammed S Alkathlan
- Infectious Diseases Department, King Fahad Specialist Hospital, Buraydah 52382, Saudi Arabia
| | - Reem S Almaghrabi
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Abdulmonem A Abuzaid
- Medical Microbiology Department, Security Forces Hospital Programme, Dammam 32314, Saudi Arabia
| | - Jaffar A Altowaileb
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Maha Al Ibrahim
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Eman M AlSalman
- Department of Family Medicine, Primary Health Care Centers, Qatif Health Network, Qatif 31911, Saudi Arabia
| | - Fatimah Alsalman
- Department of Emergency Medicine, Oyun City Hospital, Al-Ahsa 36312, Saudi Arabia
| | | | - Ahmed S Bueid
- Microbiology Laboratory, King Faisal General Hospital, Al-Ahsa 31982, Saudi Arabia
| | - Awad Al-Omari
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Research Center, Dr. Sulaiman Al Habib Medical Group, Riyadh 11372, Saudi Arabia
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar 758002, India
| |
Collapse
|
30
|
Rabaan AA, Alfaraj AH, Alshengeti A, Alawfi A, Alwarthan S, Alhajri M, Al-Najjar AH, Al Fares MA, Najim MA, Almuthree SA, AlShurbaji ST, Alofi FS, AlShehail BM, AlYuosof B, Alynbiawi A, Alzayer SA, Al Kaabi N, Abduljabbar WA, Bukhary ZA, Bueid AS. Antibodies to Combat Fungal Infections: Development Strategies and Progress. Microorganisms 2023; 11:microorganisms11030671. [PMID: 36985244 PMCID: PMC10051215 DOI: 10.3390/microorganisms11030671] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
The finding that some mAbs are antifungal suggests that antibody immunity may play a key role in the defense of the host against mycotic infections. The discovery of antibodies that guard against fungi is a significant advancement because it gives rise to the possibility of developing vaccinations that trigger protective antibody immunity. These vaccines might work by inducing antibody opsonins that improve the function of non-specific (such as neutrophils, macrophages, and NK cells) and specific (such as lymphocyte) cell-mediated immunity and stop or aid in eradicating fungus infections. The ability of antibodies to defend against fungi has been demonstrated by using monoclonal antibody technology to reconsider the function of antibody immunity. The next step is to develop vaccines that induce protective antibody immunity and to comprehend the mechanisms through which antibodies mediate protective effects against fungus.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
- Correspondence:
| | - Amal H. Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Amer Alshengeti
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah 41491, Saudi Arabia
- Department of Infection Prevention and Control, Prince Mohammad Bin Abdulaziz Hospital, National Guard Health Affairs, Al-Madinah 41491, Saudi Arabia
| | - Abdulsalam Alawfi
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah 41491, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mashael Alhajri
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Amal H. Al-Najjar
- Drug & Poison Information Center, Pharmacy Department, Security Forces Hospital Program, Riyadh 11481, Saudi Arabia
| | - Mona A. Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Mustafa A. Najim
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Madinah 41411, Saudi Arabia
| | - Souad A. Almuthree
- Department of Infectious Disease, King Abdullah Medical City, Makkah 43442, Saudi Arabia
| | - Sultan T. AlShurbaji
- Outpatient Pharmacy, Dr. Sulaiman Alhabib Medical Group, Diplomatic Quarter, Riyadh 91877, Saudi Arabia
| | - Fadwa S. Alofi
- Department of Infectious Diseases, King Fahad Hospital, Madinah 42351, Saudi Arabia
| | - Bashayer M. AlShehail
- Pharmacy Practice Department, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Buthina AlYuosof
- Directorate of Public Health, Dammam Network, Eastern Health Cluster, Dammam 31444, Saudi Arabia
| | - Ahlam Alynbiawi
- Infectious Diseases Section, Medical Specialties Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Suha A. Alzayer
- Parasitology Laboratory Department, Qatif Comprehensive Inspection Center, Qatif 31911, Saudi Arabia
| | - Nawal Al Kaabi
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates
| | - Wesam A. Abduljabbar
- Department of Medical Laboratory Sciences, Fakeeh College for Medical Science, Jeddah 21134, Saudi Arabia
| | - Zakiyah A. Bukhary
- Department of Internal Medicine, King Fahad General Hospital, Jeddah 23325, Saudi Arabia
| | - Ahmed S. Bueid
- Microbiology Laboratory, King Faisal General Hospital, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
31
|
Armstrong‐James D. Antifungal chemotherapies and immunotherapies for the future. Parasite Immunol 2023; 45:e12960. [PMID: 36403106 PMCID: PMC10078527 DOI: 10.1111/pim.12960] [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: 07/14/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/21/2022]
Abstract
Human fungal pathogens cause a broad plethora of infections, spanning cutaneous dermatophytoses to invasive infections in immunocompromised hosts. As eukaryotic pathogens are capable of morphotype switching, they present unique challenges both for drug development and the immunological response. Whilst current antifungal therapies are limited to the orally available triazoles, intravenous echonocandins and polyenes, and flucytosine and terbinafine, there has been recent significant progress in the antifungal armamentorium with ibrexafungerp, a novel orally available terpanoid that inhibits 1,3-beta-D-glucan-approved by Food and Drug Administration in 2021, and fosmanogepix, an orally available pro-drug of manogepix, which targets glycosylphosphatidylinositol-anchored protein maturation entering Phase 3 studies for candidaemia. A number of further candidates are in development. There has been significant use of existing immunotherapies such as recombinant interferon-γ and G-CSF for fungal disease in immunocompromised patients, and there are emerging opportunities for monoclonal antibodies targeting TH2 inflammation. Omalizumab, an anti-IgE monoclonal antibody in asthma, is now used routinely for the treatment of allergic bronchopulmonary aspergillosis, and further agents targeting IL-4 and IL-5 are being evaluated. In addition, T-cell CAR therapy is showing early promise for fungal disease. Thus, we are likely to see rapid advances to our approach to the management of fungal disease in the near future.
Collapse
Affiliation(s)
- Darius Armstrong‐James
- Department of Infectious DiseasesMedical Research Council Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUK
| |
Collapse
|
32
|
Wang Y, Wang K, Rivera A, Xue C. Development of a Heat-Killed fbp1 Mutant Strain as a Therapeutic Agent To Treat Invasive Cryptococcus Infection. Microbiol Spectr 2023; 11:e0495522. [PMID: 36719231 PMCID: PMC10101017 DOI: 10.1128/spectrum.04955-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/11/2023] [Indexed: 02/01/2023] Open
Abstract
In previous studies, we determined that the F-box protein Fbp1, a subunit of the SCF(Fbp1) E3 ligase in Cryptococcus neoformans, is essential for fungal pathogenesis. Heat-killed fbp1Δ cells (HK-fbp1) can confer vaccine-induced immunity against lethal challenge with clinically important invasive fungal pathogens, e.g., C. neoformans, C. gattii, and Aspergillus fumigatus. In this study, we found that either CD4+ T cells or CD8+ T cells were sufficient to confer protection against lethal challenge by C. neoformans in HK-fbp1-induced immunity. Given the potent effect of HK-fbp1 as a preventative vaccine, we further tested the potential efficacy of administering HK-fbp1 cells as a therapeutic agent for treating animals after infection. Remarkably, administration of HK-fbp1 provided robust host protection against preexisting C. neoformans infection. The mice infected with wild-type H99 cells and then treated with HK-fbp1 showed significant reduction of fungal burden in the infected lung and no dissemination of fungal cells to the brain and spleen. We find that early treatment is critical for the effective use of HK-fbp1 as a therapeutic agent. Immune analysis revealed that early treatment with HK-fbp1 cells elicited Th1-biased protective immune responses that help block fungal dissemination and promote better host protection. Our data thus suggest that HK-fbp1 is both an effective prophylactic vaccine candidate against C. neoformans infection in both immunocompetent and immunocompromised populations and a potential novel therapeutic strategy to treat early-stage cryptococcosis. IMPORTANCE Invasive fungal infections, e.g., cryptococcosis, are often life threatening and difficult to treat with very limited therapeutic options. There is no vaccine available in clinical use to prevent or treat fungal infections. Our previous studies demonstrated that heat-killed fbp1Δ cells (HK-fbp1) in Cryptococcus neoformans can be harnessed to confer protection against a challenge by the virulent parental strain, even in immunocompromised animals, such as ones lacking CD4+ T cells. In this study, we further determined that T cells are required for vaccine-induced protection against homologous challenge and that either CD4+ or CD8+ cells are sufficient. This finding is particularly important for the potential utility of this vaccine candidate in the context of HIV/AIDS-induced immune deficiency, the main risk factor for cryptococcosis in humans. Furthermore, in addition to the utility of HK-fbp1 as a prophylactic vaccine, we found that HK-fbp1 administration can inhibit disease dissemination when animals are treated at an early stage during Cryptococcus infection. Our findings could significantly expand the utility of HK-fbp1 not only as a prophylactic vaccine but also as a novel therapy against cryptococcosis. In all, our studies showed that the HK-fbp1 strain can be used both preventively and therapeutically to elicit robust host protection against cryptococcosis.
Collapse
Affiliation(s)
- Yina Wang
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Keyi Wang
- Graduate School of Biomedical Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Amariliz Rivera
- Department of Pediatrics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Chaoyang Xue
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
33
|
Branco J, Miranda IM, Rodrigues AG. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. J Fungi (Basel) 2023; 9:jof9010080. [PMID: 36675901 PMCID: PMC9862255 DOI: 10.3390/jof9010080] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Candida parapsilosis is the second most common Candida species isolated in Asia, Southern Europe, and Latin America and is often involved in invasive infections that seriously impact human health. This pathogen is part of the psilosis complex, which also includes Candida orthopsilosis and Candida metapsilosis. C. parapsilosis infections are particularly prevalent among neonates with low birth weights, individuals who are immunocompromised, and patients who require prolonged use of a central venous catheter or other indwelling devices, whose surfaces C. parapsilosis exhibits an enhanced capacity to adhere to and form biofilms. Despite this well-acknowledged prevalence, the biology of C. parapsilosis has not been as extensively explored as that of Candida albicans. In this paper, we describe the molecular mechanistic pathways of virulence in C. parapsilosis and show how they differ from those of C. albicans. We also describe the mode of action of antifungal drugs used for the treatment of Candida infections, namely, polyenes, echinocandins, and azoles, as well as the resistance mechanisms developed by C. parapsilosis to overcome them. Finally, we stress the importance of the ongoing search for species-specific features that may aid the development of effective control strategies and thus reduce the burden on patients and healthcare costs.
Collapse
Affiliation(s)
- Joana Branco
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Correspondence: ; Tel./Fax: +351-225513662
| | - Isabel M. Miranda
- Cardiovascular Research & Development Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Acácio G. Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
34
|
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
|
35
|
3,5-Dinitrobenzoate and 3,5-Dinitrobenzamide Derivatives: Mechanistic, Antifungal, and In Silico Studies. J CHEM-NY 2022. [DOI: 10.1155/2022/2336175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fungal infections, including those caused by Candida spp., are recognized in immunocompromised individuals for their high rates of morbidity and mortality. Microorganism resistance to conventional drugs compromises treatment effectiveness and yet also reveals the need to develop new drugs. In many compounds, nitro groups contribute to antimicrobial activity; thus, the inhibitory activity of a collection of twenty esters and amides (derived from 3,5-dinitrobenzoic acid) against Candida spp. was elucidated using microdilution methods to determine the Minimum Inhibitory Concentration (MIC) and Minimum Fungicide Concentration (MFC), as well as probable mechanisms of action. The structures of the synthesized compounds were characterized by FTIR spectroscopy, 1H-NMR, 13C NMR, and HRMS. Of the tested derivatives, ten presented fungicidal activity against at least one of the tested strains. Ethyl 3,5-dinitrobenzoate (2) exhibited the most potent antifungal activity against Candida albicans (MIC = 125 µg/mL; 0.52 mM), Candida krusei (MIC = 100 µg/mL; 4.16 mM), and Candida tropicalis (MIC = 500 µg/ml; 2.08 mM). The structure of the second most potent derivative (propyl 3,5-dinitrobenzoate (3) reveals that esters with short alkyl side chains exhibit better biological activity profiles. Compounds 2 and 3 presented a mechanism of action involving the fungal cell membrane. Though compound 2 modeling against C. albicans revealed a multitarget antifungal mechanism of action, involving various cellular processes, interference in the synthesis of ergosterol was observed. Our results demonstrate that certain ester derivatives containing aromatic ring nitro groups may be useful in the search for new antifungal drugs.
Collapse
|
36
|
Li L, Lu H, Zhang X, Whiteway M, Wu H, Tan S, Zang J, Tian S, Zhen C, Meng X, Li W, Zhang D, Zhang M, Jiang Y. Baicalein Acts against Candida albicans by Targeting Eno1 and Inhibiting Glycolysis. Microbiol Spectr 2022; 10:e0208522. [PMID: 35900099 PMCID: PMC9430770 DOI: 10.1128/spectrum.02085-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Baicalein (BE) is a promising antifungal small-molecule compound with an extended antifungal spectrum, good synergy with fluconazole, and low toxicity, but its target protein and antifungal mechanism remain elusive. In this study, we found that BE can function against Candida albicans by disrupting glycolysis through targeting Eno1 and inhibiting its function. Eno1 acts as a key therapeutic target of the drug, as BE had no antifungal activity against the eno1 null mutant in a Galleria mellonella model of C. albicans infection. To investigate the mechanism of action, we solved the crystal structure of C. albicans Eno1(CaEno1) and then compared the difference between this structure and that of Eno1 from humans. The predicted primary binding site of BE on CaEno1 is between amino acids D261 and W274, with D263, S269, and K273 playing critical roles in the interaction with BE. Both positions S269 and K273 have different residues in the human Eno1 (hEno1). This finding suggests that BE may bind selectively to CaEno1, which would limit the potential for side effects in humans. Our findings demonstrate that Eno1 is a target protein of BE and thus may serve as a novel target for the development of antifungal therapeutics acting through the inhibition of glycolysis. IMPORTANCE Baicalein (BE) is a promising antifungal agent which has been well characterized, but its target protein is still undiscovered. The protein Eno1 plays a crucial role in the survival of Candida albicans. However, there are few antifungal agents which inhibit the functions of Eno1. Here, we found that BE can function against Candida albicans by disrupting glycolysis through targeting Eno1 and inhibiting its function. We further solved the crystal structure of C. albicans Eno1(CaEno1) and predicted that the primary binding site of BE on CaEno1 is between amino acids D261 and W274, with D263, S269, and K273 playing critical roles in the interaction with BE. Our findings will be helpful to get specific small-molecule inhibitors of CaEno1 and open the way for the development of new antifungal therapeutics targeted at inhibiting glycolysis.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuan Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, CAS Center for Excellence in Biomacromolecules, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Hao Wu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shanlun Tan
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jianye Zang
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, CAS Center for Excellence in Biomacromolecules, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Shujuan Tian
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Cheng Zhen
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xianlei Meng
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, CAS Center for Excellence in Biomacromolecules, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Wanqian Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dazhi Zhang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Min Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, CAS Center for Excellence in Biomacromolecules, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
37
|
El-Waseif AA, Abd El-Ghani GS, Abo El maaty SA, G. Hassan M. Cytotoxicity and Promising Anti-Biofilm of Curcuma Silver Nanoparticles against Candida albicans. RESEARCH JOURNAL OF PHARMACY AND TECHNOLOGY 2022:3355-3359. [DOI: 10.52711/0974-360x.2022.00561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
The fungal pathogens considered the major human opportunistic, 50–60% of candidiasis cases patients Candida albicans. Green synthesis of AgNPs from NaNO3 using Curcuma extract was performed. UV–Vis spectrophotometry and TEM characterization were measured. AgNPs absorption peak showed between 420-440 nm and results of TEM observed, the AgNPs has a uniform regular coccus shape and size of AgNPs was ranging from 50 to 12nm with average 37nm. The activity of AgNPs as antifungal agent against Candida albicans was determined using agar well diffusion method. AgNPs showed greater antifungal activity with inhibition zone diameter 28mm at 30μg/ml. The antibiofilm activity of AgNPs was assayed in 96-well polystyrene plates and 3 ml polyethylene tube. Obviously, remarkable progressive inhibition of biofilm formation was noticed with increasing of AgNPs concentrations. Determination of AgNPs cytotoxicity on normal lung fibroblast (WI-38) cells using MTT protocol was performed. The viability observed at concentration 31.25 was 44% with IC50 value 30.5μg ml−1. When Curcuma AgNPs are used to local infected wound, may inhibit Candida cells without bad side effect on host cells surrounding the infected wounds.
Collapse
Affiliation(s)
- Amr A. El-Waseif
- Botany and Microbiology Dept., Faculty of Science (Boys), Al-Azhar University, Cairo, Egypt
| | | | | | - Mervat G. Hassan
- Botany and Microbiology Dept., Faculty of Science, Banha University, Egypt
| |
Collapse
|
38
|
Kane A, Carter DA. Augmenting Azoles with Drug Synergy to Expand the Antifungal Toolbox. Pharmaceuticals (Basel) 2022; 15:482. [PMID: 35455479 PMCID: PMC9027798 DOI: 10.3390/ph15040482] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/23/2022] Open
Abstract
Fungal infections impact the lives of at least 12 million people every year, killing over 1.5 million. Wide-spread use of fungicides and prophylactic antifungal therapy have driven resistance in many serious fungal pathogens, and there is an urgent need to expand the current antifungal arsenal. Recent research has focused on improving azoles, our most successful class of antifungals, by looking for synergistic interactions with secondary compounds. Synergists can co-operate with azoles by targeting steps in related pathways, or they may act on mechanisms related to resistance such as active efflux or on totally disparate pathways or processes. A variety of sources of potential synergists have been explored, including pre-existing antimicrobials, pharmaceuticals approved for other uses, bioactive natural compounds and phytochemicals, and novel synthetic compounds. Synergy can successfully widen the antifungal spectrum, decrease inhibitory dosages, reduce toxicity, and prevent the development of resistance. This review highlights the diversity of mechanisms that have been exploited for the purposes of azole synergy and demonstrates that synergy remains a promising approach for meeting the urgent need for novel antifungal strategies.
Collapse
Affiliation(s)
| | - Dee A. Carter
- School of Life and Environmental Sciences and Sydney ID, University of Sydney, Camperdown, NSW 2006, Australia;
| |
Collapse
|
39
|
Logviniuk D, Jaber QZ, Dobrovetsky R, Kozer N, Ksiezopolska E, Gabaldón T, Carmeli S, Fridman M. Benzylic Dehydroxylation of Echinocandin Antifungal Drugs Restores Efficacy against Resistance Conferred by Mutated Glucan Synthase. J Am Chem Soc 2022; 144:5965-5975. [PMID: 35347986 PMCID: PMC8991007 DOI: 10.1021/jacs.2c00269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Each year, infections caused by fungal pathogens claim the lives of about 1.6 million people and affect the health of over a billion people worldwide. Among the most recently developed antifungal drugs are the echinocandins, which noncompetitively inhibit β-glucan synthase, a membrane-bound protein complex that catalyzes the formation of the main polysaccharide component of the fungal cell wall. Resistance to echinocandins is conferred by mutations in FKS genes, which encode the catalytic subunit of the β-glucan synthase complex. Here, we report that selective removal of the benzylic alcohol of the nonproteinogenic amino acid 3S,4S-dihydroxy-l-homotyrosine of the echinocandins anidulafungin and rezafungin, restored their efficacy against a large panel of echinocandin-resistant Candida strains. The dehydroxylated compounds did not significantly affect the viability of human-derived cell culture lines. An analysis of the efficacy of the dehydroxylated echinocandins against resistant Candida strains, which contain mutations in the FKS1 and/or FKS2 genes of the parental strains, identified amino acids of the Fks proteins that are likely to reside in proximity to the l-homotyrosine residue of the bound drug. This study describes the first example of a chemical modification strategy to restore the efficacy of echinocandin drugs, which have a critical place in the arsenal of antifungal drugs, against resistant fungal pathogens.
Collapse
Affiliation(s)
- Dana Logviniuk
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Qais Z Jaber
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Roman Dobrovetsky
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Noga Kozer
- The Wohl Drug Discovery institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ewa Ksiezopolska
- Barcelona Supercomputing Centre (BSC-CNS), Jordi Girona, 29, Barcelona 08034, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, Barcelona 08028, Spain
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Jordi Girona, 29, Barcelona 08034, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, Barcelona 08028, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Passeig de Lluís Companys, 23, Barcelona 08010, Spain.,Centro Investigación Biomédica En Red de Enfermedades Infecciosas, Madrid 28029, Spain
| | - Shmuel Carmeli
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
40
|
The novel antifungal agent AB-22 displays in vitro activity against hyphal growth and biofilm formation in Candida albicans and potency for treating systemic candidiasis. J Microbiol 2022; 60:438-443. [DOI: 10.1007/s12275-022-2016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 10/18/2022]
|
41
|
Szymański M, Chmielewska S, Czyżewska U, Malinowska M, Tylicki A. Echinocandins - structure, mechanism of action and use in antifungal therapy. J Enzyme Inhib Med Chem 2022; 37:876-894. [PMID: 35296203 PMCID: PMC8933026 DOI: 10.1080/14756366.2022.2050224] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
With increasing number of immunocompromised patients as well as drug resistance in fungi, the risk of fatal fungal infections in humans increases as well. The action of echinocandins is based on the inhibition of β-(1,3)-d-glucan synthesis that builds the fungal cell wall. Caspofungin, micafungin, anidulafungin and rezafungin are semi-synthetic cyclic lipopeptides. Their specific chemical structure possess a potential to obtain novel derivatives with better pharmacological properties resulting in more effective treatment, especially in infections caused by Candida and Aspergillus species. In this review we summarise information about echinocandins with closer look on their chemical structure, mechanism of action, drug resistance and usage in clinical practice. We also introduce actual trends in modification of this antifungals as well as new methods of their administration, and additional use in viral and bacterial infections.
Collapse
Affiliation(s)
- Mateusz Szymański
- Department of Microbiology and Biotechnology, Laboratory of Cytobiochemistry, University of Bialystok, Bialystok, Poland
| | - Sandra Chmielewska
- Doctoral School of Exact and Natural Sciences, University of Bialystok, Bialystok, Poland
| | - Urszula Czyżewska
- Department of Microbiology and Biotechnology, Laboratory of Cytobiochemistry, University of Bialystok, Bialystok, Poland
| | - Marta Malinowska
- Department of Organic Chemistry, Laboratory of Natural Product Chemistry, University of Bialystok, Bialystok, Poland
| | - Adam Tylicki
- Department of Microbiology and Biotechnology, Laboratory of Cytobiochemistry, University of Bialystok, Bialystok, Poland
| |
Collapse
|
42
|
Abstract
![]()
When used in combination
with azole antifungal drugs, cyclooxygenase
(COX) inhibitors such as ibuprofen improve antifungal efficacy. We
report the conjugation of a chiral antifungal azole pharmacophore
to COX inhibitors and the evaluation of activity of 24 hybrids. Hybrids
derived from ibuprofen and flurbiprofen were considerably more potent
than fluconazole and comparable to voriconazole against a panel of Candida species. The potencies of hybrids composed
of an S-configured azole pharmacophore were higher
than those with an R-configured pharmacophore. Tolerance,
defined as the ability of a subpopulation of cells to grow in the
presence of the drug, to the hybrids was lower than to fluconazole
and voriconazole. The hybrids were active against a mutant lacking
CYP51, the target of azole drugs, indicating that these agents act
via a dual mode of action. This study established that azole-COX inhibitor
hybrids are a novel class of potent antifungals with clinical potential.
Collapse
Affiliation(s)
- Rebecca Elias
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Pallabita Basu
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
43
|
Sharma D, Paul RA, Rudramurthy SM, Kashyap N, Bhattacharya S, Soman R, Shankarnarayan SA, Chavan D, Singh S, Das P, Kaur H, Ghosh AK, Prasad R, Sanyal K, Chakrabarti A. Impact of FKS1 Genotype on Echinocandin In Vitro Susceptibility in Candida auris and In Vivo Response in a Murine Model of Infection. Antimicrob Agents Chemother 2022; 66:e0165221. [PMID: 34780273 PMCID: PMC8765266 DOI: 10.1128/aac.01652-21] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022] Open
Abstract
Echinocandins are frontline antifungal agents in the management of invasive infections due to multidrug resistant Candida auris. The study aimed to evaluate echinocandin resistance in C. auris isolates of multicentric origin, identify the resistance mechanism, and analyze the pharmacodynamic response to caspofungin in a neutropenic mouse model of infection. A total of 199 C. auris isolates originating from 30 centers across India were tested for susceptibility to echinocandins. Isolates with reduced susceptibility were evaluated for FKS1 mutations and in vivo response to caspofungin in a murine model of disseminated candidiasis. In addition, the response to echinocandins was assessed in light of in vitro growth kinetics, chitin content; and transcript levels of chitin synthase and FKS1 genes. We report 10 resistant C. auris isolates with four FKS1 mutations: F635Y (n = 2), F635L (n = 4), S639F (n = 3), and R1354S (n = 1). Of these, F635Y and R1354S exhibited the most profound resistance in mouse model of disseminated infection. S639F and F635L mutations conferred a moderate in vivo resistance, whereas wild-type isolates exhibiting borderline MIC were susceptible in vivo. FKS1 genotype was more accurate predictor of in vivo response than the MIC of the isolates. Isolates with high basal or inducible chitin content exhibited higher in vitro MIC in FKS1 mutant compared to wild type. FKS1 mutations play a major role in clinically relevant echinocandin resistance in C. auris with differential in vivo outcomes. This study could have implications for clinical practice and, therefore, warrants further studies.
Collapse
Affiliation(s)
- Dipti Sharma
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Raees A. Paul
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | | | - Nisha Kashyap
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | | | | | | | | | - Shreya Singh
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | | | - Harsimran Kaur
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Anup K. Ghosh
- Department of Medical Microbiology, PGIMER, Chandigarh, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology & Integrative Sciences and Health, Amity University Haryana, Gurgaon, India
| | - Kaustuv Sanyal
- Jawaharlal Nehru Center for Advanced Scientific Research, Bengaluru, India
| | | |
Collapse
|
44
|
Abdel-Haq N, Smith SM, Asmar BI. Micafungin injection for the treatment of invasive candidiasis in pediatric patients under 4 months of age. Expert Rev Anti Infect Ther 2021; 20:493-505. [PMID: 34882043 DOI: 10.1080/14787210.2022.2013807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Neonates and young infants with invasive candidiasis are particularly at increased risk of dissemination including hematogenous Candida meningoencephalitis. The echinocandins including micafungin have emerged as a preferred agent in most cases of candidemia and invasive candidiasis but data in pediatric patients under 4 months of age are limited. AREAS COVERED In this report, we review the micafungin use in infants younger than 4 months of age. Animal studies as well as clinical data that support its use in neonatal candidiasis are reviewed. In addition, the status of FDA approval and the rationale of micafungin dosing recommendations in infants <4 months are discussed. EXPERT OPINION A dose of 4 mg/kg was approved for treatment of candidemia, Candida peritonitis and abscesses excluding meningoencephalitis or ocular involvement in patients younger than 4 months of age. However, because of the risk of central nervous system dissemination as well as the difficulty in establishing this diagnosis, this dose is inadequate to treat ill infants with candidemia. More studies are needed to establish the safety and efficacy of micafungin daily dose of at least 10 mg/kg in infants younger than 4 months of age when hematogenous Candida meningoencephalitis or ocular involvement cannot be excluded.
Collapse
Affiliation(s)
- Nahed Abdel-Haq
- Division of Infection Diseases, Children's Hospital of Michigan, Detroit, MI, USA.,Children's Hospital of Michigan, Detroit, MI, USA.,Department of Pediatrics, Central Michigan University, Mount Pleasant, MI, USA.,Department of Pediatrics, Wayne State University, Detroit, MI, USA
| | | | - Basim I Asmar
- Division of Infection Diseases, Children's Hospital of Michigan, Detroit, MI, USA.,Children's Hospital of Michigan, Detroit, MI, USA.,Department of Pediatrics, Central Michigan University, Mount Pleasant, MI, USA.,Department of Pediatrics, Wayne State University, Detroit, MI, USA
| |
Collapse
|
45
|
Ghassemi N, Poulhazan A, Deligey F, Mentink-Vigier F, Marcotte I, Wang T. Solid-State NMR Investigations of Extracellular Matrixes and Cell Walls of Algae, Bacteria, Fungi, and Plants. Chem Rev 2021; 122:10036-10086. [PMID: 34878762 DOI: 10.1021/acs.chemrev.1c00669] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Extracellular matrixes (ECMs), such as the cell walls and biofilms, are important for supporting cell integrity and function and regulating intercellular communication. These biomaterials are also of significant interest to the production of biofuels and the development of antimicrobial treatment. Solid-state nuclear magnetic resonance (ssNMR) and magic-angle spinning-dynamic nuclear polarization (MAS-DNP) are uniquely powerful for understanding the conformational structure, dynamical characteristics, and supramolecular assemblies of carbohydrates and other biomolecules in ECMs. This review highlights the recent high-resolution investigations of intact ECMs and native cells in many organisms spanning across plants, bacteria, fungi, and algae. We spotlight the structural principles identified in ECMs, discuss the current technical limitation and underexplored biochemical topics, and point out the promising opportunities enabled by the recent advances of the rapidly evolving ssNMR technology.
Collapse
Affiliation(s)
- Nader Ghassemi
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Alexandre Poulhazan
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States.,Department of Chemistry, Université du Québec à Montréal, Montreal H2X 2J6, Canada
| | - Fabien Deligey
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | | | - Isabelle Marcotte
- Department of Chemistry, Université du Québec à Montréal, Montreal H2X 2J6, Canada
| | - Tuo Wang
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| |
Collapse
|
46
|
da Silva GO, Farias BCS, da Silva RB, Teixeira EH, Cordeiro RDA, Hissa DC, Melo VMM. Effects of lipopeptide biosurfactants on clinical strains of Malassezia furfur growth and biofilm formation. Med Mycol 2021; 59:1191-1201. [PMID: 34424316 DOI: 10.1093/mmy/myab051] [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: 05/27/2021] [Revised: 07/23/2021] [Accepted: 08/19/2021] [Indexed: 01/02/2023] Open
Abstract
Lipopeptide biosurfactants (LBs) are biological molecules with low toxicity that have aroused growing interest in the pharmaceutical industry. Their chemical structure confers antimicrobial and antibiofilm properties against different species. Despite their potential, few studies have demonstrated their capability against Malassezia spp., commensal yeasts which can cause dermatitis and serious infections. Thus, the aim of this study was to evaluate the antifungal activity of biosurfactants produced by new strains of Bacillus subtilis TIM10 and B. vallismortis TIM68 against M. furfur and their potential for removal and inhibition of yeast biofilms. Biosurfactants were classified as lipopeptides by FTIR, and their composition was characterized by ESI-Q-TOF/MS, showing ions for iturin, fengycin, and surfactin, with a greater abundance of surfactin. Through the broth microdilution method, both biosurfactants inhibited the growth of clinical M. furfur strains. Biosurfactant TIM10 showed greater capacity for growth inhibition, with no statistical difference compared to those obtained by the commercial antifungal fluconazole for M. furfur 153DR5 and 154DR8 strains. At minimal inhibitory concentrations (MIC-2), TIM10 and TIM68 were able to inhibit biofilm formation, especially TIM10, with an inhibition rate of approximately 90%. In addition, both biosurfactants were able to remove pre-formed biofilm. Both biosurfactants showed no toxicity against murine fibroblasts, even at concentrations above MIC-2. Our results show the effectiveness of LBs in controlling the growth and biofilm formation of M. furfur clinical strains and highlight the potential of these agents to compose new formulations for the treatment of these fungi.
Collapse
Affiliation(s)
- Gabrielly Oliveira da Silva
- Laboratório de Ecologia Microbiana e Biotecnologia (LEMBiotech). Departamento de Biologia, Federal University of Ceara, Avenida Humberto Monte 2977, Fortaleza - CE 60455-760, Brazil
| | - Bárbara Cibelle Soares Farias
- Laboratório de Ecologia Microbiana e Biotecnologia (LEMBiotech). Departamento de Biologia, Federal University of Ceara, Avenida Humberto Monte 2977, Fortaleza - CE 60455-760, Brazil
| | - Renally Barbosa da Silva
- Laboratório Integrado de Biomoléculas (LIBS). Departamento de Patologia e Medicina Legal, Federal University of Ceara, Rua Coronel Nunes de Melo, Fortaleza - CE 60430-275, Brazil
| | - Edson Holanda Teixeira
- Laboratório Integrado de Biomoléculas (LIBS). Departamento de Patologia e Medicina Legal, Federal University of Ceara, Rua Coronel Nunes de Melo, Fortaleza - CE 60430-275, Brazil
| | - Rossana de Aguiar Cordeiro
- Departamento de Patologia e Medicina Legal, Federal University of Ceara, Rua Coronel Nunes de Melo, Fortaleza - CE 60430-275, Brazil
| | - Denise Cavalcante Hissa
- Laboratório de Recursos Genéticos (LARGEN). Departamento de Biologia, Federal University of Ceara, Avenida Humberto Monte 2977, Fortaleza - CE 60455-760, Brazil
| | - Vânia Maria Maciel Melo
- Laboratório de Ecologia Microbiana e Biotecnologia (LEMBiotech). Departamento de Biologia, Federal University of Ceara, Avenida Humberto Monte 2977, Fortaleza - CE 60455-760, Brazil
| |
Collapse
|
47
|
Bajaj K, Buchanan RM, Grapperhaus CA. Antifungal activity of thiosemicarbazones, bis(thiosemicarbazones), and their metal complexes. J Inorg Biochem 2021; 225:111620. [PMID: 34619407 DOI: 10.1016/j.jinorgbio.2021.111620] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/06/2021] [Accepted: 09/18/2021] [Indexed: 12/25/2022]
Abstract
Fungi are ubiquitous in nature, and typically cause little or no environmental or pathogenic damage to their plant, animal, and human hosts. However, a small but growing number of pathogenic fungi are spreading world-wide at an alarming rate threatening global ecosystem health and proliferation. Many of these emerging pathogens have developed multi-drug resistance to front line therapeutics increasing the urgency for the development of new antifungal agents. This review examines the development of thiosemicarbazones, bis(thiosemicarbazones), and their metal complexes as potential antifungal agents against more than 65 different fungal strains. The fungistatic activity of the compounds are quantified based on the zone of inhibition, minimum inhibitory concentration, or growth inhibition percentage. In this review, reported activities were standardized based on molar concentrations to simplify comparisons between different compounds. Of all the fungal strains reported in the review, A. niger in particular was very resistant towards a majority of tested compounds. Our analysis of the data shows that metal complexes are typically more active than non-coordinated ligands with copper(II) and zinc(II) complexes generally displaying the highest activity.
Collapse
Affiliation(s)
- Kritika Bajaj
- Department of Chemistry, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, United States of America
| | - Robert M Buchanan
- Department of Chemistry, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, United States of America
| | - Craig A Grapperhaus
- Department of Chemistry, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, United States of America.
| |
Collapse
|
48
|
Wang Q, Li Y, Cai X, Li R, Zheng B, Yang E, Liang T, Yang X, Wan Z, Liu W. Two Sequential Clinical Isolates of Candida glabrata with Multidrug-Resistance to Posaconazole and Echinocandins. Antibiotics (Basel) 2021; 10:antibiotics10101217. [PMID: 34680798 PMCID: PMC8532709 DOI: 10.3390/antibiotics10101217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/28/2021] [Accepted: 10/02/2021] [Indexed: 12/17/2022] Open
Abstract
Candida glabrata is one of the most prevalent causative pathogens of invasive candidiasis, and multidrug-resistant strains are emerging. We identified two clinical isolates of C. glabrata, BMU10720 and BMU10722 sequentially isolated from one patient with multidrug-resistance to posaconazole (POS), caspofungin (CAS), micafungin (MCF), and anidulafungin (ANF). Overexpression of ERG11 in BMU10720 and CDR1 in BMU10722 were detected at basal level. When exposed to POS, CDR1 was significantly up-regulated in both isolates compared with susceptible reference strain, while ERG11 was up-regulated considerably only in BMU10720. PDR1 sequencing revealed that both isolates harbored P76S, P143T, and D243N substitutions, while ERG11 was intact. Cdr1 inhibitor FK520 reversed POS-resistance by down-regulating ERG11 expression. FKS sequencing revealed that both isolates harbored S663P substitution in FKS2, and four single nucleotide polymorphisms (SNPs) existed in FKS2 genes between BMU10720 and BMU10722, while FKS1 was intact. Both FKS1 and FKS2 were up-regulated by CAS in BMU10720 and BMU10722. FK520 down-regulated FKS2 expression induced by CAS through inhibiting calcineurin, resulting in synergic effect with echinocandins as well as Congo Red and Calcofluor White, two cell wall-perturbing agents. In conclusion, the multidrug-resistance of C. glabrata isolates in our study was conferred by different mechanisms. CDR1 and ERG11 overexpression in one isolate and only CDR1 overexpression in the other isolate may mediate POS-resistance. S663P mutation in FKS2 and up-regulation of FKS2 may contribute to echinocandin-resistance in both isolates.
Collapse
Affiliation(s)
- Qiqi Wang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China; (Q.W.); (R.L.); (T.L.); (X.Y.); (Z.W.)
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Research Center for Medical Mycology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
| | - Yun Li
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing 100034, China; (Y.L.); (B.Z.)
| | - Xuan Cai
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China; (Q.W.); (R.L.); (T.L.); (X.Y.); (Z.W.)
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Research Center for Medical Mycology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
| | - Bo Zheng
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing 100034, China; (Y.L.); (B.Z.)
| | - Ence Yang
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China;
| | - Tianyu Liang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China; (Q.W.); (R.L.); (T.L.); (X.Y.); (Z.W.)
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Research Center for Medical Mycology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
| | - Xinyu Yang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China; (Q.W.); (R.L.); (T.L.); (X.Y.); (Z.W.)
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Research Center for Medical Mycology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
| | - Zhe Wan
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China; (Q.W.); (R.L.); (T.L.); (X.Y.); (Z.W.)
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Research Center for Medical Mycology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
| | - Wei Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China; (Q.W.); (R.L.); (T.L.); (X.Y.); (Z.W.)
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Research Center for Medical Mycology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- Correspondence: ; Tel.: +86-10-8357-3075
| |
Collapse
|
49
|
Fayed B, Jayakumar MN, Soliman SSM. Caspofungin-resistance in Candida auris is cell wall-dependent phenotype and potential prevention by zinc oxide nanoparticles. Med Mycol 2021; 59:1243-1256. [PMID: 34612496 DOI: 10.1093/mmy/myab059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 01/07/2023] Open
Abstract
Candida auris is an emergent nosocomial multi-drug-resistant yeast that represents a global health threat. Recently, C. auris clinical isolates with caspofungin resistance were identified. Mutation in FKS1 gene was determined as a mechanism of resistance. However, the ability of C. auris to develop acquired and cross-resistance has never been investigated. Herein, this resistance ability due to caspofungin and associate mechanisms were investigated. C. auris clinical isolate was successively cultured for 10 generations in the presence of caspofungin compared to fluconazole-treatment and untreated controls. This was followed by the analysis of target gene expression and phenotypic changes. The obtained results showed that caspofungin-treated C. auris exhibited elevated MIC50(caspofungin), slower growth, elevated chitin content, overexpression of caspofungin target genes, and cross-resistance to fluconazole. Interestingly, caspofungin exposure induced cell-cell adhesion and biofilm formation. C. auris gradually lost caspofungin resistance after removal of antifungal pressure, while keeping the overexpression of fungal cell wall-related genes including ALS5. We propose that C. auris ageing in the presence of caspofungin caused the development of persistent phenotypic changes in the fungal cell wall, leading to acquired and physical cross-resistance mechanisms. Surprisingly, formulation of caspofungin in zinc oxide nanoparticles prevented the aforementioned behavioral changes regardless of the pathogen generations. LAY SUMMARY Candida auris developed resistance against caspofungin. Our data indicated that this resistance mechanism is unique because of changes in the genes related to cell wall adhesions. Formulation of caspofungin in ZnO nanoparticles was able to overcome these phenotypic changes.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.,Chemistry of Natural and Microbial Product Department, National Research Centre, Cairo, 12622, Egypt
| | - Manju Nidagodu Jayakumar
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Sameh S M Soliman
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.,College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| |
Collapse
|
50
|
In Vitro Selection of Thioether-Closed Macrocyclic Peptide Ligands by Means of the RaPID System. Methods Mol Biol 2021. [PMID: 34596852 DOI: 10.1007/978-1-0716-1689-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The Random nonstandard Peptides Integrated Discovery (RaPID) system enables efficient screening of macrocyclic peptides with high affinities against target molecules. Random peptide libraries are prepared by in vitro translation using the Flexible In vitro Translation (FIT) system, which allows for incorporation of diverse nonproteinogenic amino acids into peptides by genetic code reprogramming. By introducing an N-chloroacetyl amino acid at the N-terminus and a Cys at the downstream, macrocyclic peptide libraries can be readily generated via posttranslational thioether formation. Here, we describe how to prepare a thioether-closed macrocyclic peptide library, and its application to the RaPID screening.
Collapse
|