1
|
Karamchandani BM, Maurya PA, Pawar AA, Pable A, Awale M, Dalvi SG, Banat IM, Satpute SK. The role of statistical methods in optimizing and enhancing fungal chitosan commercial production. 3 Biotech 2025; 15:70. [PMID: 40041691 PMCID: PMC11872829 DOI: 10.1007/s13205-025-04236-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/09/2025] [Indexed: 03/09/2025] Open
Abstract
This review portrays the role of fermentation technology in the production of fungal chitosan (FCH) under optimized conditions using statistical methods. It is noteworthy to mention that FCH is superior than crustacean chitosan (CCH) due to its low molecular weight (LMW) (≈ 20-30 kDa), polymer homogeneity, high degree of deacetylation (DDA) (74-92%), along with thermal stability, solubility at wide physiological pH and greener extraction process. Employment of suitable submerged fermentation conditions improves the quality (high DDA and LMW) of FCH for varied applications. Literature survey depicted the crucial role of recent advancements of statistical tools and software in FCH fermentation technology. A close look at the literature over the past three decades showed ≈ 64% of FCH production from Absidia coerulea, Rhizopus oryzae, R. japonicus, Aspergillus niger, A. terreus, A. flavus, Cunninghamella elegans (≈ 16% each) followed by Mucor rouxii (≈ 11%). Other fungi namely, Benjaminiella poitrasii, Penicillium chrysogenum and Trametes versicolor, Gongronella butleri and Ganoderma lucidum (≈ 5% each) have been reported. The Design of Experiments (DOE), like response surface methodology (RSM) including Plackett-Burman Design (PBD), Central composite design (CCD), Box Behnken design (BBD) and Taguchi have improved biomass and FCH yield meaningfully. Among different approaches, One-factor-at-a-time (OFAT) approach was the foremost choice (≈ 29%) followed by CCD (≈ 12%) and OFAT combined with CCD (≈11%) were employed by researchers to optimize FCH production from potent strains. Around 6% of the reports suggest that BBD, Taguchi, FC-BBD, CCD, 2 > 2 factorials have been employed at an individual level to achieve a high yield of FCH. Those methods can be employed either individually or in combination. This article comprehensively presents the basic information, performances of the statistical methods/tools of DOE and software employed for successful scaling-up of FCH while highlighting their merits, limitations, and challenges. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04236-2.
Collapse
Affiliation(s)
- Bhoomika M. Karamchandani
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007 Maharashtra India
| | - Priya A. Maurya
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007 Maharashtra India
| | - Ameya A. Pawar
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007 Maharashtra India
| | - Anupama Pable
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007 Maharashtra India
| | - Manik Awale
- Department of Statistics, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007 Maharashtra India
| | - Sunil G. Dalvi
- Tissue Culture Section, Vasantdada Sugar Institute, Pune, India
| | - Ibrahim M. Banat
- School of Biomedical Sciences, Faculty of Life and Health Sciences, University of Ulster, Coleraine, NI BT52 1SA UK
| | - Surekha K. Satpute
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007 Maharashtra India
| |
Collapse
|
2
|
Lends A, Lamon G, Delcourte L, Sturny-Leclere A, Grélard A, Morvan E, Abdul-Shukkoor MB, Berbon M, Vallet A, Habenstein B, Dufourc EJ, Schanda P, Aimanianda V, Loquet A. Molecular Distinction of Cell Wall and Capsular Polysaccharides in Encapsulated Pathogens by In Situ Magic-Angle Spinning NMR Techniques. J Am Chem Soc 2025; 147:6813-6824. [PMID: 39955787 DOI: 10.1021/jacs.4c16975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
Pathogenic fungal and bacterial cells are enveloped within a cell wall, a molecular barrier at their cell surface, and a critical architecture that constantly evolves during pathogenesis. Understanding the molecular composition, structural organization, and mobility of polysaccharides constituting this cell envelope is crucial to correlate cell wall organization with its role in pathogenicity and to identify potential antifungal targets. For the fungal pathogen Cryptococcus neoformans, the characterization of the cell envelope has been complexified by the presence of an additional external polysaccharide capsular shell. Here, we investigate how magic-angle spinning (MAS) solid-state NMR techniques increase the analytical capabilities to characterize the structure and dynamics of this encapsulated pathogen. The versatility of proton detection experiments, dynamic-based filters, and relaxation measurements facilitate the discrimination of the highly mobile external capsular structure from the internal rigid cell wall of C. neoformans. In addition, we report the in situ detection of triglyceride molecules from lipid droplets based on NMR dynamic filters. Together, we demonstrate a nondestructive technique to study the cell wall architecture of encapsulated microbes using C. neoformans as a model, an airborne opportunistic fungal pathogen that infects mainly immunocompromised but also competent hosts.
Collapse
Affiliation(s)
- Alons Lends
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Gaelle Lamon
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Loic Delcourte
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Aude Sturny-Leclere
- Institut Pasteur, Unité Mycologie Moléculaire, Université Paris Cité, F-75015 Paris, France
| | - Axelle Grélard
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Estelle Morvan
- Inserm, IECB, UAR3033, US01, Université de Bordeaux, CNRS, F-33607 Pessac, France
| | | | - Mélanie Berbon
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Alicia Vallet
- Institut de Biologie Structurale, CEA, Université Grenoble Alpes, CNRS, 71 avenue des martyrs, F-38000 Grenoble, France
| | - Birgit Habenstein
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Erick J Dufourc
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
| | - Paul Schanda
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Vishukumar Aimanianda
- Institut Pasteur, Unité Mycologie Moléculaire, Université Paris Cité, F-75015 Paris, France
| | - Antoine Loquet
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33607 Pessac, France
- Inserm, IECB, UAR3033, US01, Université de Bordeaux, CNRS, F-33607 Pessac, France
| |
Collapse
|
3
|
Kırboğa KK, Karim A, Küçüksille EU, Rudrapal M, Khan J, Achar RR, Silina E, Manturova N, Stupin V. Exploring the antifungal potential of Cannabis sativa-derived stilbenoids and cannabinoids against novel targets through in silico protein interaction profiling. Front Chem 2025; 12:1515424. [PMID: 39834844 PMCID: PMC11743709 DOI: 10.3389/fchem.2024.1515424] [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: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Cannabinoid and stilbenoid compounds derived from Cannabis sativa were screened against eight specific fungal protein targets to identify potential antifungal agents. The proteins investigated included Glycosylphosphatidylinositol (GPI), Enolase, Mannitol-2-dehydrogenase, GMP synthase, Dihydroorotate dehydrogenase (DHODH), Heat shock protein 90 homolog (Hsp90), Chitin Synthase 2 (CaChs2), and Mannitol-1-phosphate 5-dehydrogenase (M1P5DH), all of which play crucial roles in fungal survival and pathogenicity. This research evaluates the binding affinities and interaction profiles of selected cannabinoids and stilbenoids with these eight proteins using molecular docking and molecular dynamics simulations. The ligands with the highest binding affinities were identified, and their pharmacokinetic profiles were analyzed using ADMET analysis. The results indicate that GMP synthase exhibited the highest binding affinity with Cannabistilbene I (-9.1 kcal/mol), suggesting hydrophobic solid interactions and multiple hydrogen bonds. Similarly, Chitin Synthase 2 demonstrated significant binding with Cannabistilbene I (-9.1 kcal/mol). In contrast, ligands such as Cannabinolic acid and 8-hydroxycannabinolic acid exhibited moderate binding affinities, underscoring the variability in interaction strengths among different proteins. Despite promising in silico results, experimental validation is necessary to confirm therapeutic potential. This research lays a crucial foundation for future studies, emphasizing the importance of evaluating binding affinities, pharmacokinetic properties, and multi-target interactions to identify promising antifungal agents.
Collapse
Affiliation(s)
- Kevser Kübra Kırboğa
- Faculty of Engineering, Department of Bioengineering, Bilecik Şeyh Edebali University, Bilecik, Türkiye
| | - Aman Karim
- Faculty of Multidisciplinary Studies, Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Ecir Uğur Küçüksille
- Faculty of Engineering, Department of Computer Engineering, Isparta Suleyman Demirel University, Isparta, Türkiye
| | - Mithun Rudrapal
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan’s Foundation for Science, Technology and Research, Guntur, India
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Raghu Ram Achar
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, India
| | - Ekaterina Silina
- Institute of Digital Biodesign and Modeling of Living Systems, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Natalia Manturova
- Department of Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Victor Stupin
- Department of Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
4
|
Avina SL, Pawar S, Rivera A, Xue C. Will the Real Immunogens Please Stand Up: Exploiting the Immunogenic Potential of Cryptococcal Cell Antigens in Fungal Vaccine Development. J Fungi (Basel) 2024; 10:840. [PMID: 39728336 PMCID: PMC11676676 DOI: 10.3390/jof10120840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/30/2024] [Indexed: 12/28/2024] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen that is a continuous global health concern, especially for immunocompromised populations. The World Health Organization recognized C. neoformans as one of four critical fungal pathogens, thus emphasizing the need for increased research efforts and clinical resource expansion. Currently, there are no fungal vaccines available for clinical use. Exciting new findings in cryptococcal vaccine development have identified whole cell-based and subunit-based vaccinations to help mitigate health risks and make commercialization attainable. Importantly, recent work has focused on how different cryptococcal cell-wall antigens modified in these vaccine candidates allow us to manipulate their immunogenicity to produce a desired long-term protective anti-fungal immune response. In this review, we discuss the different cryptococcal cell immunogens, namely the polysaccharide capsule, glucans, chitin/chitosan, mannoproteins, and extracellular vesicles, and their role in novel cryptococcal vaccination approaches. Additionally, we examine the immunological mechanisms responsible for protection in these vaccine candidates and the similar host response-stimulation pathways induced through different immunogen exposure.
Collapse
Affiliation(s)
- Samantha L. Avina
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ 07103, USA; (S.L.A.); (S.P.); (A.R.)
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Siddhi Pawar
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ 07103, USA; (S.L.A.); (S.P.); (A.R.)
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Amariliz Rivera
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ 07103, USA; (S.L.A.); (S.P.); (A.R.)
- Department of Pediatrics and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Chaoyang Xue
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ 07103, USA; (S.L.A.); (S.P.); (A.R.)
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
5
|
Student M, Hellmann MJ, Cord-Landwehr S, Moerschbacher BM. Chitins and chitosans-A tale of discovery and disguise, of attachment and attainment. CURRENT OPINION IN PLANT BIOLOGY 2024; 82:102661. [PMID: 39536646 DOI: 10.1016/j.pbi.2024.102661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Chitin polymers are an essential structural component of fungal cell walls, but host chitinases can weaken them, contributing to disease resistance in fungal pathogens. Chitin oligomers thus produced are immunogenic signal molecules eliciting additional disease resistance mechanisms. Fungi may counteract these, e.g. by partial deacetylation of chitin, converting it into chitosans, protecting the cell walls against chitinase attack, and inactivating elicitor active oligomers. This molecular stealth hypothesis for fungal pathogenicity has repeatedly been tested by mutating single or multiple chitin deacetylase genes, supporting the hypothesis but simultaneously suggesting additional roles for chitin deacetylation in virulence, such as surface attachment and sensing, host tissue penetration and colonization, as well as spore formation, stabilization, and germination. Interestingly, recent evidence suggests that host plants have evolved counter strategies by inhibiting fungal chitin deacetylases, lending further credibility to the suggested action of these enzymes as pathogenicity/virulence factors, and possibly offering leads toward novel functional fungicides.
Collapse
Affiliation(s)
- Mounashree Student
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany
| | - Margareta J Hellmann
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany
| | - Stefan Cord-Landwehr
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany
| | - Bruno M Moerschbacher
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany.
| |
Collapse
|
6
|
Antunes D, Domingues R, Cruz-Almeida M, Rodrigues L, Borges O, Carvalho A, Casadevall A, Fernandes C, Gonçalves T. Cell wall nanoparticles from hyphae of Alternaria infectoria grown with caspofungin, nikkomycin, or pyroquilon trigger different activation profiles in macrophages. Microbiol Spectr 2024; 12:e0064524. [PMID: 39329485 PMCID: PMC11537108 DOI: 10.1128/spectrum.00645-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/02/2024] [Indexed: 09/28/2024] Open
Abstract
Alternaria infectoria causes opportunistic human infections and is a source of allergens leading to respiratory allergies. In this work, we prepared cell wall nanoparticles (CWNPs) as a novel approach to study macrophage immunomodulation by fungal hyphal cell walls. A. infectoria was grown in the presence of caspofungin, an inhibitor of β(1,3)-glucan synthesis; nikkomycin Z, an inhibitor of chitin synthases; and pyroquilon, an inhibitor of dihydroxynaphthalene (DHN)-melanin synthesis. Distinct CWNPs were obtained from these cultures, referred to as casCWNPs, nkCWNPs, and pyrCWNPs, respectively. CWNPs are round-shaped particles with a diameter of 70-200 nm diameter particles that when added to macrophages are taken up by membrane ruffling. CWNPs with no DHN-melanin and more glucan (pyrCWNPs) caused early macrophage activation and lowest viability, with the cells exhibiting ultrastructural modifications such as higher vacuolization and formation of autophagy-like structures. CasCWNPs promoted the highest tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β) increase, also resulting in the release of partially degraded chitin, an aspect never observed in macrophage-like cells and fungi. After 6 h of interaction with CWNPs, only half were viable, except with control CWNPs. Overall, this work indicates that compounds that modify the fungal cell wall led to CWNPs with new properties that may have implications for the effects of drugs during antifungal therapy. CWNPs provide a new tool to study the interaction of hyphal fungal cell wall components with phagocytic cells and enable to show how the modification of cell wall components in A. infectoria can modulate the response by macrophages.IMPORTANCEAlternaria species are ubiquitous environmental fungi to which the human host can continuously be exposed, through the inhalation of fungal spores but also of fragments of hyphae, from desegregated mycelia. These fungi are involved in hypersensitization and severe respiratory allergies, such as asthma, and can cause opportunistic infections in immunodepressed human host leading to severe disease. The first fungal structures to interact with the host cells are the cell wall components, and their modulation leads to differential immune responses. Here, we show that fungal cells grown with cell wall inhibitors led to cell wall nanoparticles with new properties in their interaction with macrophages. With this strategy, we overcame the limitation of in vitro assays interacting with filamentous fungi and showed that the absence of DNH-melanin leads to higher virulence, while caspofungin leads to cells walls that trigger higher hydrolysis of chitin and higher production of cytokines.
Collapse
Affiliation(s)
- Daniela Antunes
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rita Domingues
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana Cruz-Almeida
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Olga Borges
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Univ Coimbra, FFUC—Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Chantal Fernandes
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Teresa Gonçalves
- Univ Coimbra, CNC-UC—Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Univ Coimbra, FMUC—Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
7
|
du Plooy LM, Telzrow CL, Nichols CB, Probst C, Castro-Lopez N, Wormley FL, Alspaugh JA. A fungal ubiquitin ligase and arrestin binding partner contribute to pathogenesis and survival during cellular stress. mBio 2024; 15:e0098124. [PMID: 39235249 PMCID: PMC11481503 DOI: 10.1128/mbio.00981-24] [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: 04/04/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Cellular responses to external stress allow microorganisms to adapt to a vast array of environmental conditions, including infection sites. The molecular mechanisms behind these responses are studied to gain insight into microbial pathogenesis, which could lead to new antimicrobial therapies. Here, we explore a role for arrestin protein-mediated ubiquitination in stress response and pathogenesis in the pathogenic fungus Cryptococcus neoformans. In a previous study, we identified four arrestin-like proteins in C. neoformans and found that one of these is required for efficient membrane synthesis, likely by directing interaction between fatty acid synthases and the Rsp5 E3 ubiquitin ligase. Here, we further explore Cn Rsp5 function and determine that this single Ub ligase is absolutely required for pathogenesis and survival in the presence of cellular stress. Additionally, we show that a second arrestin-like protein, Ali2, similarly facilitates interaction between Rsp5 and some of its protein targets. Of the four postulated C. neoformans arrestin-like proteins, Ali2 appears to contribute the most to C. neoformans pathogenesis, likely by directing Rsp5 to pathogenesis-related ubiquitination targets. A proteomics-based differential ubiquitination screen revealed that several known cell surface proteins are ubiquitinated by Rsp5 and a subset also requires Ali2 for their ubiquitination. Rsp5-mediated ubiquitination alters the stability and the localization of these proteins. A loss of Rsp5-mediated ubiquitination results in cell wall defects that increase susceptibility to external stresses. These findings support a model in which arrestin-like proteins guide Rsp5 to ubiquitinate specific target proteins, some of which are required for survival during stress. IMPORTANCE Microbial proteins involved in human infectious diseases often need to be modified by specific chemical additions to be fully functional. Here, we explore the role of a particular protein modification, ubiquitination, in infections due to the human fungal pathogen Cryptococcus neoformans. We identified a complex of proteins responsible for adding ubiquitin groups to fungal proteins, and this complex is required for virulence. These proteins are fungal specific and might be targets for novel anti-infection therapy.
Collapse
Affiliation(s)
- Lukas M. du Plooy
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Calla L. Telzrow
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Connie B. Nichols
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Corinna Probst
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Natalia Castro-Lopez
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Biology, Texas Christian University, Fort Worth, Texas, USA
| | - Floyd L. Wormley
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Biology, Texas Christian University, Fort Worth, Texas, USA
| | - J. Andrew Alspaugh
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
8
|
Peng X, Kong Q, Wei Q, Guo S, Chen Q, Peng M, An B, Wang X, Zhang C, Sang H. Verapamil enhances the activity of Caspofungin against Cryptococcus neoformans, coinciding with inhibited Ca 2+/CN pathway and damage to cell wall integrity. Int J Antimicrob Agents 2024; 64:107303. [PMID: 39151646 DOI: 10.1016/j.ijantimicag.2024.107303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVES Given the challenges posed by toxicity and drug resistance in the treatment of cryptococcal infections, we sought to explore the antifungal potential of verapamil (VER), a calcium channel blocker, against Cryptococcus neoformans (C. neoformans), and its potential synergy with antifungals, specifically caspofungin (CAS). MATERIALS AND METHODS In vitro and in vivo (Galleria mellonella) models were employed to assess VER's antifungal activity and its interaction with CAS. Mechanisms underlying the synergism were explored through analysis of cell wall integrity, membrane permeability, and gene expression related to the calcineurin pathway. Additionally, the influence of Ca2+ on chitin deacetylase activity was investigated. RESULTS VER exhibited a pronounced antifungal effect on C. neoformans and synergized with CAS, enhancing antifungal efficacy in Galleria mellonella. VER reduced chitosan content and disrupted cell wall integrity, evidenced by melanin leakage and fluorescence staining. VER+CAS modified membrane permeability, triggering intracellular ROS accumulation and mitochondrial membrane potential alterations. VER mitigated CAS-induced calcium fluctuations and downregulated calcineurin pathway genes. Furthermore, it was found that the enzyme activity of chitin deacetylase of C. neoformans is significantly influenced by the presence of Ca2+, suggesting that the use of VER may affect this activity. CONCLUSIONS The synergistic antifungal effect of VER and CAS represents a promising therapeutic strategy for cryptococcal infections. The multifaceted mechanisms, including disruption of cell wall integrity and modulation of membrane permeability, and regulation of intracellular calcium signaling pathways, offer new insights into antifungal drug development.
Collapse
Affiliation(s)
- Xinyuan Peng
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qingtao Kong
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qian Wei
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shilin Guo
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiying Chen
- Department of Dermatology, the First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Min Peng
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Binyi An
- Department of Dermatology, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Wang
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chen Zhang
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hong Sang
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
9
|
Gómez-Gaviria M, Mora-Montes HM. Exploring the potential of chitin and chitosan in nanobiocomposites for fungal immunological detection and antifungal action. Carbohydr Res 2024; 543:109220. [PMID: 39038396 DOI: 10.1016/j.carres.2024.109220] [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: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Chitin is a polymer of N-acetylglucosamine and an essential component of the fungal cell wall. Chitosan is the deacetylated form of chitin and is also important for maintaining the integrity of this structure. Both polysaccharides are widely distributed in nature and have been shown to have a variety of applications in biomedicine, including their potential in immune sensing and as potential antifungal agents. In addition, chitin has been reported to play an important role in the pathogen-host interaction, involving innate and adaptive immune responses. This paper will explore the role of chitin and chitosan when incorporated into nanobiocomposites to improve their efficacy in detecting fungi of medical interest and inhibiting their growth. Potential applications in diagnostic and therapeutic medicine will be discussed, highlighting their promise in the development of more sensitive and effective tools for the early diagnosis of fungal infections. This review aims to highlight the importance of the convergence of nanotechnology and biology in addressing public health challenges.
Collapse
Affiliation(s)
- Manuela Gómez-Gaviria
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto, Mexico
| | - Héctor M Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto, Mexico.
| |
Collapse
|
10
|
Redrado-Hernández S, Macías-León J, Castro-López J, Belén Sanz A, Dolader E, Arias M, González-Ramírez AM, Sánchez-Navarro D, Petryk Y, Farkaš V, Vincke C, Muyldermans S, García-Barbazán I, Del Agua C, Zaragoza O, Arroyo J, Pardo J, Gálvez EM, Hurtado-Guerrero R. Broad Protection against Invasive Fungal Disease from a Nanobody Targeting the Active Site of Fungal β-1,3-Glucanosyltransferases. Angew Chem Int Ed Engl 2024; 63:e202405823. [PMID: 38856634 DOI: 10.1002/anie.202405823] [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: 04/01/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/11/2024]
Abstract
Invasive fungal disease accounts for about 3.8 million deaths annually, an unacceptable rate that urgently prompts the discovery of new knowledge-driven treatments. We report the use of camelid single-domain nanobodies (Nbs) against fungal β-1,3-glucanosyltransferases (Gel) involved in β-1,3-glucan transglycosylation. Crystal structures of two Nbs with Gel4 from Aspergillus fumigatus revealed binding to a dissimilar CBM43 domain and a highly conserved catalytic domain across fungal species, respectively. Anti-Gel4 active site Nb3 showed significant antifungal efficacy in vitro and in vivo prophylactically and therapeutically against different A. fumigatus and Cryptococcus neoformans isolates, reducing the fungal burden and disease severity, thus significantly improving immunocompromised animal survival. Notably, C. deneoformans (serotype D) strains were more susceptible to Nb3 and genetic Gel deletion than C. neoformans (serotype A) strains, indicating a key role for β-1,3-glucan remodelling in C. deneoformans survival. These findings add new insight about the role of β-1,3-glucan in fungal biology and demonstrate the potential of nanobodies in targeting fungal enzymes to combat invasive fungal diseases.
Collapse
Grants
- PID2022-136362NB-I00 Ministerio de Asuntos Económicos y Transformación Digital, Gobierno de España
- BIO2016-79289-P Ministerio de Economía y Competitividad, Gobierno de España
- PID2019-105223GB-I00 Ministerio de Ciencia, Innovación y Universidades y Agencia Estatal de Investigación, Gobierno de España
- PID2022-136888NB-I00 Ministerio de Ciencia e Innovación y Agencia Estatal de Investigación, Gobierno de España
- PID2020-114546RB Ministerio de Ciencia e Innovación y Agencia Estatal de Investigación, Gobierno de España
- PID2020-113963RB-I00 Ministerio de Ciencia e Innovación y Agencia Estatal de Investigación, Gobierno de España
- S2017/BMD3691-InGEMICS-CM Comunidad de Madrid
- B29_17R, E34_R17, LMP58_18 and LMP139_21 Gobierno de Aragon
- Nanofungi Precipita (crowdfunding)
- BIOSTRUCTX_5186 FP7 (2007-2013), BioStruct-X
Collapse
Affiliation(s)
- Sergio Redrado-Hernández
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
| | - Javier Macías-León
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - Jorge Castro-López
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - Ana Belén Sanz
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Elena Dolader
- Department of Microbiology, Pediatry, Radiology and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Maykel Arias
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
- Department of Microbiology, Pediatry, Radiology and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Andrés Manuel González-Ramírez
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - David Sánchez-Navarro
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - Yuliya Petryk
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Vladimír Farkaš
- Department of Glycobiology, Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, 84538, Bratislava, Slovakia
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Irene García-Barbazán
- Mycology Reference Laboratory. National Centre for Microbiology., Health Institute Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Celia Del Agua
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Department of Pathology, Hospital Clínico Universitario Lozano Blesa, IIS-Aragón, 50009, Zaragoza, Spain
| | - Oscar Zaragoza
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
- Mycology Reference Laboratory. National Centre for Microbiology., Health Institute Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Javier Arroyo
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Julián Pardo
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
- Department of Microbiology, Pediatry, Radiology and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Eva M Gálvez
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
| | - Ramon Hurtado-Guerrero
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
- Fundación ARAID, 50018, Zaragoza, Spain
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200, Copenhagen, Denmark
| |
Collapse
|
11
|
Li Y, Chadwick B, Pham T, Xie X, Lin X. Aspartyl peptidase May1 induces host inflammatory response by altering cell wall composition in the fungal pathogen Cryptococcus neoformans. mBio 2024; 15:e0092024. [PMID: 38742885 PMCID: PMC11237595 DOI: 10.1128/mbio.00920-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Cryptococcus neoformans causes cryptococcal meningoencephalitis, a disease that kills more than 180,000 people annually. Contributing to its success as a fungal pathogen is its cell wall surrounded by a capsule. When the cryptococcal cell wall is compromised, exposed pathogen-associated molecular pattern molecules (PAMPs) could trigger host recognition and initiate attack against this fungus. Thus, cell wall composition and structure are tightly regulated. The cryptococcal cell wall is unusual in that chitosan, the acetylated form of chitin, is predominant over chitin and is essential for virulence. Recently, it was shown that acidic pH weakens the cell wall and increases exposure of PAMPs partly due to decreased chitosan levels. However, the molecular mechanism responsible for the cell wall remodeling in acidic pH is unknown. In this study, by screening for genes involved in cryptococcal tolerance to high levels of CO2, we serendipitously discovered that the aspartyl peptidase May1 contributes to cryptococcal sensitivity to high levels of CO2 due to acidification of unbuffered media. Overexpression of MAY1 increases the cryptococcal cell size and elevates PAMP exposure, causing a hyper-inflammatory response in the host while MAY1 deletion does the opposite. We discovered that May1 weakens the cell wall and reduces the chitosan level, partly due to its involvement in the degradation of Chs3, the sole chitin synthase that supplies chitin to be converted to chitosan. Consistently, overexpression of CHS3 largely rescues the phenotype of MAY1oe in acidic media. Collectively, we demonstrate that May1 remodels the cryptococcal cell wall in acidic pH by reducing chitosan levels through its influence on Chs3. IMPORTANCE The fungal cell wall is a dynamic structure, monitoring and responding to internal and external stimuli. It provides a formidable armor to the fungus. However, in a weakened state, the cell wall also triggers host immune attack when PAMPs, including glucan, chitin, and mannoproteins, are exposed. In this work, we found that the aspartyl peptidase May1 impairs the cell wall of Cryptococcus neoformans and increases the exposure of PAMPs in the acidic environment by reducing the chitosan level. Under acidic conditions, May1 is involved in the degradation of the chitin synthase Chs3, which supplies chitin to be deacetylated to chitosan. Consistently, the severe deficiency of chitosan in acidic pH can be rescued by overexpressing CHS3. These findings improve our understanding of cell wall remodeling and reveal a potential target to compromise the cell wall integrity in this important fungal pathogen.
Collapse
Affiliation(s)
- Yeqi Li
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Benjamin Chadwick
- Department of Plant Biology, University of Georgia, Athens, Georgia, USA
| | - Tuyetnhu Pham
- Department of Plant Biology, University of Georgia, Athens, Georgia, USA
| | - Xiaofeng Xie
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Department of Plant Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
12
|
Schiphof K, Kawauchi M, Tsuji K, Yoshimi A, Tanaka C, Nakazawa T, Honda Y. Functional analysis of basidiomycete specific chitin synthase genes in the agaricomycete fungus Pleurotus ostreatus. Fungal Genet Biol 2024; 172:103893. [PMID: 38657898 DOI: 10.1016/j.fgb.2024.103893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Chitin is an essential structural component of fungal cell walls composed of transmembrane proteins called chitin synthases (CHSs), which have a large range of reported effects in ascomycetes; however, are poorly understood in agaricomycetes. In this study, evolutionary and molecular genetic analyses of chs genes were conducted using genomic information from nine ascomycete and six basidiomycete species. The results support the existence of seven previously classified chs clades and the discovery of three novel basidiomycete-specific clades (BI-BIII). The agaricomycete fungus Pleurotus ostreatus was observed to have nine putative chs genes, four of which were basidiomycete-specific. Three of these basidiomycete specific genes were disrupted in the P. ostreatus 20b strain (ku80 disruptant) through homologous recombination and transformants were obtained (Δchsb2, Δchsb3, and Δchsb4). Despite numerous transformations Δchsb1 was unobtainable, suggesting disruption of this gene causes a crucial negative effect in P. ostreatus. Disruption of these chsb2-4 genes caused sparser mycelia with rougher surfaces and shorter aerial hyphae. They also caused increased sensitivity to cell wall and membrane stress, thinner cell walls, and overexpression of other chitin and glucan synthases. These genes have distinct roles in the structural formation of aerial hyphae and cell walls, which are important for understanding basidiomycete evolution in filamentous fungi.
Collapse
Affiliation(s)
- Kim Schiphof
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Moriyuki Kawauchi
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan.
| | - Kenya Tsuji
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Akira Yoshimi
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan; Graduate School of Global Environmental Studies, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Chihiro Tanaka
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan; Graduate School of Global Environmental Studies, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Takehito Nakazawa
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yoichi Honda
- Graduate School of Agriculture, Kyoto University, Kitashirakawaoiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
13
|
Loza L, Doering TL. A fungal protein organizes both glycogen and cell wall glucans. Proc Natl Acad Sci U S A 2024; 121:e2319707121. [PMID: 38743622 PMCID: PMC11126952 DOI: 10.1073/pnas.2319707121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Glycogen is a glucose storage molecule composed of branched α-1,4-glucan chains, best known as an energy reserve that can be broken down to fuel central metabolism. Because fungal cells have a specialized need for glucose in building cell wall glucans, we investigated whether glycogen is used for this process. For these studies, we focused on the pathogenic yeast Cryptococcus neoformans, which causes ~150,000 deaths per year worldwide. We identified two proteins that influence formation of both glycogen and the cell wall: glycogenin (Glg1), which initiates glycogen synthesis, and a protein that we call Glucan organizing enzyme 1 (Goe1). We found that cells missing Glg1 lack α-1,4-glucan in their walls, indicating that this material is derived from glycogen. Without Goe1, glycogen rosettes are mislocalized and β-1,3-glucan in the cell wall is reduced. Altogether, our results provide mechanisms for a close association between glycogen and cell wall.
Collapse
Affiliation(s)
- Liza Loza
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO63110
| | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO63110
| |
Collapse
|
14
|
Kwon S, Choi Y, Kim ES, Lee KT, Bahn YS, Jung KW. Pleiotropic roles of LAMMER kinase, Lkh1 in stress responses and virulence of Cryptococcus neoformans. Front Cell Infect Microbiol 2024; 14:1369301. [PMID: 38774630 PMCID: PMC11106425 DOI: 10.3389/fcimb.2024.1369301] [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: 01/12/2024] [Accepted: 03/27/2024] [Indexed: 05/24/2024] Open
Abstract
Dual-specificity LAMMER kinases are highly evolutionarily conserved in eukaryotes and play pivotal roles in diverse physiological processes, such as growth, differentiation, and stress responses. Although the functions of LAMMER kinase in fungal pathogens in pathogenicity and stress responses have been characterized, its role in Cryptococcus neoformans, a human fungal pathogen and a model yeast of basidiomycetes, remains elusive. In this study, we identified a LKH1 homologous gene and constructed a strain with a deleted LKH1 and a complemented strain. Similar to other fungi, the lkh1Δ mutant showed intrinsic growth defects. We observed that C. neoformans Lkh1 was involved in diverse stress responses, including oxidative stress and cell wall stress. Particularly, Lkh1 regulates DNA damage responses in Rad53-dependent and -independent manners. Furthermore, the absence of LKH1 reduced basidiospore formation. Our observations indicate that Lkh1 becomes hyperphosphorylated upon treatment with rapamycin, a TOR protein inhibitor. Notably, LKH1 deletion led to defects in melanin synthesis and capsule formation. Furthermore, we found that the deletion of LKH1 led to the avirulence of C. neoformans in a systemic cryptococcosis murine model. Taken together, Lkh1 is required for the stress response, sexual differentiation, and virulence of C. neoformans.
Collapse
Affiliation(s)
- Sunhak Kwon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Jeonbuk, Republic of Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yeseul Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Eui-Seong Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea
| | - Kyung-Tae Lee
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea
| | - Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Kwang-Woo Jung
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Jeonbuk, Republic of Korea
| |
Collapse
|
15
|
de Oliveira HC, Santos MDM, Camillo-Andrade AC, Castelli RF, Dos Reis FCG, Carvalho PC, Rodrigues ML. Proteomics reveals that the antifungal activity of fenbendazole against Cryptococcus neoformans requires protein kinases. Int J Antimicrob Agents 2024; 63:107157. [PMID: 38548248 DOI: 10.1016/j.ijantimicag.2024.107157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024]
Abstract
Cryptococcus neoformans is responsible for over 100 000 deaths annually, and the treatment of this fungal disease is expensive and not consistently effective. Unveiling new therapeutic avenues is crucial. Previous studies have suggested that the anthelmintic drug fenbendazole is an affordable and nontoxic candidate to combat cryptococcosis. However, its mechanism of anticryptococcal activity has been only superficially investigated. In this study, we examined the global cellular response of C. neoformans to fenbendazole using a proteomic approach (data are available via ProteomeXchange with identifier PXD047041). Fenbendazole treatment mostly impacted the abundance of proteins related to metabolic pathways, RNA processing, and intracellular traffic. Protein kinases, in particular, were significantly affected by fenbendazole treatment. Experimental validation of the proteomics data using a collection of C. neoformans mutants led to the identification of critical roles of five protein kinases in fenbendazole's antifungal activity. In fact, mutants lacking the expression of genes encoding Chk1, Tco2, Tco3, Bub1, and Sch9 kinases demonstrated greater resistance to fenbendazole compared to wild-type cells. In combination with the standard antifungal drug amphotericin B, fenbendazole reduced the cryptococcal burden in mice. These findings not only contribute to the elucidation of fenbendazole's mode of action but also support its use in combination therapy with amphotericin B. In conclusion, our data suggest that fenbendazole holds promise for further development as an anticryptococcal agent.
Collapse
Affiliation(s)
| | - Marlon D M Santos
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba, Brazil; Analytical Biochemistry and Proteomics Unit. IIBCE/Institut Pasteur de Montevideo, Uruguay
| | - Amanda C Camillo-Andrade
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba, Brazil; Analytical Biochemistry and Proteomics Unit. IIBCE/Institut Pasteur de Montevideo, Uruguay
| | - Rafael F Castelli
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba, Brazil
| | - Flavia C G Dos Reis
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba, Brazil; Centro de Desenvolvimento Tecnológico em Saúde (CDTS), Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Paulo C Carvalho
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba, Brazil
| | - Marcio L Rodrigues
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba, Brazil; Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Conn BN, Lieberman JA, Chatman P, Cotton K, Essandoh MA, Ebqa’ai M, Nelson TL, Wozniak KL. Antifungal activity of eumelanin-inspired indoylenepheyleneethynylene against Cryptococcus neoformans. Front Microbiol 2024; 14:1339303. [PMID: 38293553 PMCID: PMC10826398 DOI: 10.3389/fmicb.2023.1339303] [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: 11/15/2023] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen that causes meningitis in >152,000 immunocompromised individuals annually, leading to 112,000 yearly deaths. The four classes of existing antifungal agents target plasma membrane sterols (ergosterol), nucleic acid synthesis, and cell wall synthesis. Existing drugs are not highly effective against Cryptococcus, and antifungal drug resistance is an increasing problem. A novel antimicrobial compound, a eumelanin-inspired indoylenepheyleneethynylene, EIPE-1, was synthesized and has antimicrobial activity against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MSRA), but not towards Gram-negative organisms. Based on EIPE-1's antibacterial activity, we hypothesized that EIPE-1 could have antifungal activity. For these studies, we tested EIPE-1 against C. neoformans strain H99 and 6 additional cryptococcal clinical isolates. We examined antifungal activity, cytotoxicity, effects on fungal gene expression, and mechanism of action of EIPE-1. Results showed that EIPE-1 has fungicidal effects on seven cryptococcal strains with MICs ranging from 1.56 to 3.125 μg/mL depending on the strain, and it is non-toxic to mammalian cells. We conducted scanning and transmission electron microscopy on the exposed cells to examine structural changes to the organism following EIPE-1 treatment. Cells exposed displayed structural changes to their cell wall and membranes, with internal contents leaking out of the cells. To understand the effect of EIPE-1 on fungal gene expression, RNA sequencing was conducted. Results showed that EIPE-1 affects several processes involved stress response, ergosterol biosynthesis, capsule biosynthesis, and cell wall attachment and remodeling. Therefore, our studies demonstrate that EIPE-1 has antifungal activity against C. neoformans, which affects both cellular structure and gene expression of multiple fungal pathways involved in cell membrane stability and viability.
Collapse
Affiliation(s)
- Brittney N. Conn
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Jacob A. Lieberman
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Priscilla Chatman
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Kaitlyn Cotton
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Martha A. Essandoh
- Department of Chemistry, Oklahoma State University, Stillwater, OK, United States
| | - Mohammad Ebqa’ai
- Department of Chemistry, Oklahoma State University, Stillwater, OK, United States
| | - Toby L. Nelson
- Department of Chemistry, Oklahoma State University, Stillwater, OK, United States
| | - Karen L. Wozniak
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
17
|
Maybruck BT, Upadhya R, Lam WC, Specht CA, Lodge JK. Fluorescence and Biochemical Assessment of the Chitin and Chitosan Content of Cryptococcus. Methods Mol Biol 2024; 2775:329-347. [PMID: 38758327 PMCID: PMC11487689 DOI: 10.1007/978-1-0716-3722-7_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The cell wall of the fungal pathogens Cryptococcus neoformans and C. gattii is critical for cell wall integrity and signaling external threats to the cell, allowing it to adapt and grow in a variety of changing environments. Chitin is a polysaccharide found in the cell walls of fungi that is considered to be essential for fungal survival. Chitosan is a polysaccharide derived from chitin via deacetylation that is also essential for cryptococcal cell wall integrity, fungal pathogenicity, and virulence. Cryptococcus has evolved mechanisms to regulate the amount of chitin and chitosan during growth under laboratory conditions or during mammalian infection. Therefore, levels of chitin and chitosan have been useful phenotypes to define mutant Cryptococcus strains. As a result, we have developed and/or refined various qualitative and quantitative methods for measuring chitin and chitosan. These techniques include those that use fluorescent probes that are known to bind to chitin (e.g., calcofluor white and wheat germ agglutinin), as well as those that preferentially bind to chitosan (e.g., eosin Y and cibacron brilliant red 3B-A). Techniques that enhance the localization and quantification of chitin and chitosan in the cell wall include (i) fluorescence microscopy, (ii) flow cytometry, (iii) and spectrofluorometry. We have also modified two highly selective biochemical methods to measure cellular chitin and chitosan content: the Morgan-Elson and the 3-methyl-2-benzothiazolone hydrazine hydrochloride (MBTH) assays, respectively.
Collapse
Affiliation(s)
- Brian T Maybruck
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- AstraZeneca, South San Francisco, CA, USA
| | - Rajendra Upadhya
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
| | - Woei C Lam
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Pfizer STL, Chesterfield, MO, USA
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Charles A Specht
- Department of Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jennifer K Lodge
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
18
|
Tugume L, Ssebambulidde K, Kasibante J, Ellis J, Wake RM, Gakuru J, Lawrence DS, Abassi M, Rajasingham R, Meya DB, Boulware DR. Cryptococcal meningitis. Nat Rev Dis Primers 2023; 9:62. [PMID: 37945681 DOI: 10.1038/s41572-023-00472-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 11/12/2023]
Abstract
Cryptococcus neoformans and Cryptococcus gattii species complexes cause meningoencephalitis with high fatality rates and considerable morbidity, particularly in persons with deficient T cell-mediated immunity, most commonly affecting people living with HIV. Whereas the global incidence of HIV-associated cryptococcal meningitis (HIV-CM) has decreased over the past decade, cryptococcosis still accounts for one in five AIDS-related deaths globally due to the persistent burden of advanced HIV disease. Moreover, mortality remains high (~50%) in low-resource settings. The armamentarium to decrease cryptococcosis-associated mortality is expanding: cryptococcal antigen screening in the serum and pre-emptive azole therapy for cryptococcal antigenaemia are well established, whereas enhanced pre-emptive combination treatment regimens to improve survival of persons with cryptococcal antigenaemia are in clinical trials. Short courses (≤7 days) of amphotericin-based therapy combined with flucytosine are currently the preferred options for induction therapy of cryptococcal meningitis. Whether short-course induction regimens improve long-term morbidity such as depression, reduced neurocognitive performance and physical disability among survivors is the subject of further study. Here, we discuss underlying immunology, changing epidemiology, and updates on the management of cryptococcal meningitis with emphasis on HIV-associated disease.
Collapse
Affiliation(s)
- Lillian Tugume
- Infectious Diseases Institute, Makerere University, Kampala, Uganda.
| | - Kenneth Ssebambulidde
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John Kasibante
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Jayne Ellis
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Clinical Research Department, Faculty of Infectious and Tropical Diseases London School of Hygiene and Tropical Medicine, London, UK
| | - Rachel M Wake
- Institute for Infection and Immunity, St George's University of London, London, UK
| | - Jane Gakuru
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - David S Lawrence
- Clinical Research Department, Faculty of Infectious and Tropical Diseases London School of Hygiene and Tropical Medicine, London, UK
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Mahsa Abassi
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Radha Rajasingham
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - David B Meya
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - David R Boulware
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
19
|
Ueno K, Nagamori A, Honkyu NO, Kataoka M, Shimizu K, Chang YC, Kwon-Chung KJ, Miyazaki Y. Cryptococcus neoformans requires the TVF1 gene for thermotolerance and virulence. Med Mycol 2023; 61:myad101. [PMID: 37818721 PMCID: PMC10565887 DOI: 10.1093/mmy/myad101] [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: 07/11/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023] Open
Abstract
Cryptococcus neoformans is the primary causative agent of cryptococcosis. Since C. neoformans thrives in environments and its optimal growth temperature is 25-30°C, it needs to adapt to heat stress in order to cause infection in mammalian hosts. In this study, we aimed to investigate the role of an uncharacterized gene, CNAG_03308. Although the CNAG_03308 deletion strain grew as well as the parent strain KN99, it produced yeast cells with abnormal morphology at 37°C and failed to propagate at 39°C. Furthermore, the deletion strain exhibited slower growth at 37°C in the presence of congo red, which is a cell wall stressor. When cultured at 39°C, the deletion strain showed strong staining with fluorescent probes for cell wall chitin and chitosan, including FITC-labeled wheat germ agglutinin, Eosin Y, and calcofluor white. The transmission electron microscopy of the deletion strain revealed a thickened inner layer of the cell wall containing chitin and chitosan under heat stress. This cell-surface altered deletion strain induced dendritic cells to secrete more interleukin (IL)-6 and IL-23 than the control strains under heat stress. In a murine infection study, C57BL/6 mice infected with the deletion strain exhibited lower mortality and lower fungal burden in the lungs and brain compared to those infected with the control strains. Based on these findings, we concluded that CNAG_03308 gene is necessary for C. neoformans to adapt to heat stress both in vitro and in the host environment. Therefore, we designated the CNAG_03308 gene as TVF1, which stands for thermotolerance and virulence-related factor 1.
Collapse
Affiliation(s)
- Keigo Ueno
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Akiko Nagamori
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Nahoko Oniyama Honkyu
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kiminori Shimizu
- Department of Biological Science and Technology, Tokyo University of Science, Tokyo 125-8585, Japan
| | - Yun C Chang
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kyung J Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Yoshitsugu Miyazaki
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
20
|
Cavalcante SB, Dos Santos Biscaino C, Kreusch MG, da Silva AF, Duarte RTD, Robl D. The hidden rainbow: the extensive biotechnological potential of Antarctic fungi pigments. Braz J Microbiol 2023; 54:1675-1687. [PMID: 37286926 PMCID: PMC10484874 DOI: 10.1007/s42770-023-01011-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/19/2023] [Indexed: 06/09/2023] Open
Abstract
The Antarctic continent is an extreme environment recognized mainly by its subzero temperatures. Fungi are ubiquitous microorganisms that stand out even among Antarctic organisms, primarily due to secondary metabolites production with several biological activities. Pigments are examples of such metabolites, which mainly occur in response to hostile conditions. Various pigmented fungi have been isolated from the Antarctic continent, living in the soil, sedimentary rocks, snow, water, associated with lichens, mosses, rhizospheres, and zooplankton. Physicochemical extreme environments provide a suitable setup for microbial pigment production with unique characteristics. The biotechnological potential of extremophiles, combined with concerns over synthetic pigments, has led to a great interest in natural pigment alternatives. Besides biological activities provided by fungal pigments for surviving in extreme environments (e.g., photoprotection, antioxidant activity, and stress resistance), it may present an opportunity for biotechnological industries. This paper reviews the biotechnological potential of Antarctic fungal pigments, with a detailed discussion over the biological role of fungal pigments, potential industrial production of pigments from extremophilic fungi, pigments toxicity, current market perspective and published intellectual properties related to pigmented Antarctic fungi.
Collapse
Affiliation(s)
- Sabrina Barros Cavalcante
- Department of Microbiology, Immunology and Parasitlogy, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Carla Dos Santos Biscaino
- Department of Microbiology, Immunology and Parasitlogy, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Marianne Gabi Kreusch
- Department of Microbiology, Immunology and Parasitlogy, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - André Felipe da Silva
- Bioprocess and Biotechnology Engineering Undergraduate Program, Federal University of Tocantins (UFT), Gurupi, TO, Brazil
| | - Rubens Tadeu Delgado Duarte
- Department of Microbiology, Immunology and Parasitlogy, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Diogo Robl
- Department of Microbiology, Immunology and Parasitlogy, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil.
| |
Collapse
|
21
|
Nagy L, Vonk P, Künzler M, Földi C, Virágh M, Ohm R, Hennicke F, Bálint B, Csernetics Á, Hegedüs B, Hou Z, Liu X, Nan S, Pareek M, Sahu N, Szathmári B, Varga T, Wu H, Yang X, Merényi Z. Lessons on fruiting body morphogenesis from genomes and transcriptomes of Agaricomycetes. Stud Mycol 2023; 104:1-85. [PMID: 37351542 PMCID: PMC10282164 DOI: 10.3114/sim.2022.104.01] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/02/2022] [Indexed: 01/09/2024] Open
Abstract
Fruiting bodies (sporocarps, sporophores or basidiomata) of mushroom-forming fungi (Agaricomycetes) are among the most complex structures produced by fungi. Unlike vegetative hyphae, fruiting bodies grow determinately and follow a genetically encoded developmental program that orchestrates their growth, tissue differentiation and sexual sporulation. In spite of more than a century of research, our understanding of the molecular details of fruiting body morphogenesis is still limited and a general synthesis on the genetics of this complex process is lacking. In this paper, we aim at a comprehensive identification of conserved genes related to fruiting body morphogenesis and distil novel functional hypotheses for functionally poorly characterised ones. As a result of this analysis, we report 921 conserved developmentally expressed gene families, only a few dozens of which have previously been reported to be involved in fruiting body development. Based on literature data, conserved expression patterns and functional annotations, we provide hypotheses on the potential role of these gene families in fruiting body development, yielding the most complete description of molecular processes in fruiting body morphogenesis to date. We discuss genes related to the initiation of fruiting, differentiation, growth, cell surface and cell wall, defence, transcriptional regulation as well as signal transduction. Based on these data we derive a general model of fruiting body development, which includes an early, proliferative phase that is mostly concerned with laying out the mushroom body plan (via cell division and differentiation), and a second phase of growth via cell expansion as well as meiotic events and sporulation. Altogether, our discussions cover 1 480 genes of Coprinopsis cinerea, and their orthologs in Agaricus bisporus, Cyclocybe aegerita, Armillaria ostoyae, Auriculariopsis ampla, Laccaria bicolor, Lentinula edodes, Lentinus tigrinus, Mycena kentingensis, Phanerochaete chrysosporium, Pleurotus ostreatus, and Schizophyllum commune, providing functional hypotheses for ~10 % of genes in the genomes of these species. Although experimental evidence for the role of these genes will need to be established in the future, our data provide a roadmap for guiding functional analyses of fruiting related genes in the Agaricomycetes. We anticipate that the gene compendium presented here, combined with developments in functional genomics approaches will contribute to uncovering the genetic bases of one of the most spectacular multicellular developmental processes in fungi. Citation: Nagy LG, Vonk PJ, Künzler M, Földi C, Virágh M, Ohm RA, Hennicke F, Bálint B, Csernetics Á, Hegedüs B, Hou Z, Liu XB, Nan S, M. Pareek M, Sahu N, Szathmári B, Varga T, Wu W, Yang X, Merényi Z (2023). Lessons on fruiting body morphogenesis from genomes and transcriptomes of Agaricomycetes. Studies in Mycology 104: 1-85. doi: 10.3114/sim.2022.104.01.
Collapse
Affiliation(s)
- L.G. Nagy
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - P.J. Vonk
- Microbiology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands;
| | - M. Künzler
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland;
| | - C. Földi
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - M. Virágh
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - R.A. Ohm
- Microbiology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands;
| | - F. Hennicke
- Project Group Genetics and Genomics of Fungi, Chair Evolution of Plants and Fungi, Ruhr-University Bochum, 44780, Bochum, North Rhine-Westphalia, Germany;
| | - B. Bálint
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - Á. Csernetics
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - B. Hegedüs
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - Z. Hou
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - X.B. Liu
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - S. Nan
- Institute of Applied Mycology, Huazhong Agricultural University, 430070 Hubei Province, PR China
| | - M. Pareek
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - N. Sahu
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - B. Szathmári
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - T. Varga
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - H. Wu
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - X. Yang
- Institute of Applied Mycology, Huazhong Agricultural University, 430070 Hubei Province, PR China
| | - Z. Merényi
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| |
Collapse
|
22
|
Rocafort M, Srivastava V, Bowen JK, Díaz-Moreno SM, Guo Y, Bulone V, Plummer KM, Sutherland PW, Anderson MA, Bradshaw RE, Mesarich CH. Cell Wall Carbohydrate Dynamics during the Differentiation of Infection Structures by the Apple Scab Fungus, Venturia inaequalis. Microbiol Spectr 2023; 11:e0421922. [PMID: 37039647 PMCID: PMC10269774 DOI: 10.1128/spectrum.04219-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/15/2023] [Indexed: 04/12/2023] Open
Abstract
Scab, caused by the biotrophic fungal pathogen Venturia inaequalis, is the most economically important disease of apples. During infection, V. inaequalis colonizes the subcuticular host environment, where it develops specialized infection structures called runner hyphae and stromata. These structures are thought to be involved in nutrient acquisition and effector (virulence factor) delivery, but also give rise to conidia that further the infection cycle. Despite their importance, very little is known about how these structures are differentiated. Likewise, nothing is known about how these structures are protected from host defenses or recognition by the host immune system. To better understand these processes, we first performed a glycosidic linkage analysis of sporulating tubular hyphae from V. inaequalis developed in culture. This analysis revealed that the V. inaequalis cell wall is mostly composed of glucans (44%) and mannans (37%), whereas chitin represents a much smaller proportion (4%). Next, we used transcriptomics and confocal laser scanning microscopy to provide insights into the cell wall carbohydrate composition of runner hyphae and stromata. These analyses revealed that, during subcuticular host colonization, genes of V. inaequalis putatively associated with the biosynthesis of immunogenic carbohydrates, such as chitin and β-1,6-glucan, are downregulated relative to growth in culture, while on the surface of runner hyphae and stromata, chitin is deacetylated to the less-immunogenic carbohydrate chitosan. These changes are anticipated to enable the subcuticular differentiation of runner hyphae and stromata by V. inaequalis, as well as to protect these structures from host defenses and recognition by the host immune system. IMPORTANCE Plant-pathogenic fungi are a major threat to food security. Among these are subcuticular pathogens, which often cause latent asymptomatic infections, making them difficult to control. A key feature of these pathogens is their ability to differentiate specialized subcuticular infection structures that, to date, remain largely understudied. This is typified by Venturia inaequalis, which causes scab, the most economically important disease of apples. In this study, we show that, during subcuticular host colonization, V. inaequalis downregulates genes associated with the biosynthesis of two immunogenic cell wall carbohydrates, chitin and β-1,6-glucan, and coats its subcuticular infection structures with a less-immunogenic carbohydrate, chitosan. These changes are anticipated to enable host colonization by V. inaequalis and provide a foundation for understanding subcuticular host colonization by other plant-pathogenic fungi. Such an understanding is important, as it may inform the development of novel control strategies against subcuticular plant-pathogenic fungi.
Collapse
Affiliation(s)
- Mercedes Rocafort
- Laboratory of Molecular Plant Pathology, School of Agriculture and Environment, Massey University, Palmerston North, New Zealand
| | - Vaibhav Srivastava
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Joanna K. Bowen
- The New Zealand Institute for Plant and Food Research Limited, Mount Albert Research Centre, Auckland, New Zealand
| | - Sara M. Díaz-Moreno
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Yanan Guo
- Laboratory of Molecular Plant Pathology, School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Vincent Bulone
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
- School of Food, Agriculture and Wine, The University of Adelaide, Waite Campus, Adelaide, South Australia, Australia
| | - Kim M. Plummer
- Department of Animal, Plant and Soil Sciences, AgriBio, Centre for AgriBiosciences, La Trobe University, Bundoora, Melbourne, Victoria, Australia
| | - Paul W. Sutherland
- The New Zealand Institute for Plant and Food Research Limited, Mount Albert Research Centre, Auckland, New Zealand
| | - Marilyn A. Anderson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, Victoria, Australia
| | - Rosie E. Bradshaw
- Laboratory of Molecular Plant Pathology, School of Natural Sciences, Massey University, Palmerston North, New Zealand
- Bioprotection Aotearoa, Massey University, Palmerston North, New Zealand
| | - Carl H. Mesarich
- Laboratory of Molecular Plant Pathology, School of Agriculture and Environment, Massey University, Palmerston North, New Zealand
- Bioprotection Aotearoa, Massey University, Palmerston North, New Zealand
| |
Collapse
|
23
|
The ER Protein Translocation Channel Subunit Sbh1 Controls Virulence of Cryptococcus neoformans. mBio 2023; 14:e0338422. [PMID: 36749043 PMCID: PMC9973365 DOI: 10.1128/mbio.03384-22] [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] [Indexed: 02/08/2023] Open
Abstract
The fungal pathogen Cryptococcus neoformans is distinguished by a cell-wall-anchored polysaccharide capsule that is critical for virulence. Biogenesis of both cell wall and capsule relies on the secretory pathway. Protein secretion begins with polypeptide translocation across the endoplasmic reticulum (ER) membrane through a highly conserved channel formed by three proteins: Sec61, Sbh1, and Sss1. Sbh1, the most divergent, contains multiple phosphorylation sites, which may allow it to regulate entry into the secretory pathway in a species- and protein-specific manner. Absence of SBH1 causes a cell-wall defect in both Saccharomyces cerevisiae and C. neoformans, although other phenotypes differ. Notably, proteomic analysis showed that when cryptococci are grown in conditions that mimic aspects of the mammalian host environment (tissue culture medium, 37°C, 5% CO2), a set of secretory and transmembrane proteins is upregulated in wild-type, but not in Δsbh1 mutant cells. The Sbh1-dependent proteins show specific features of their ER targeting sequences that likely cause them to transit less efficiently into the secretory pathway. Many also act in cell-wall biogenesis, while several are known virulence factors. Consistent with these observations, the C. neoformans Δsbh1 mutant is avirulent in a mouse infection model. We conclude that, in the context of conditions encountered during infection, Sbh1 controls the entry of virulence factors into the secretory pathway of C. neoformans, and thereby regulates fungal pathogenicity. IMPORTANCE Cryptococcus neoformans is a yeast that causes almost 200,000 deaths worldwide each year, mainly of immunocompromised individuals. The surface structures of this pathogen, a protective cell wall surrounded by a polysaccharide capsule, are made and maintained by proteins that are synthesized inside the cell and travel outwards through the secretory pathway. A protein called Sbh1 is part of the machinery that determines which polypeptides enter this export pathway. We found that when Sbh1 is absent, both C. neoformans and the model yeast S. cerevisiae show cell-wall defects. Lack of Sbh1 also changes the pattern of secretion of both transmembrane and soluble proteins, in a manner that depends on characteristics of their sequences. Notably, multiple proteins that are normally upregulated in conditions similar to those encountered during infection, including several needed for cryptococcal virulence, are no longer increased. Sbh1 thereby regulates the ability of this important pathogen to cause disease.
Collapse
|
24
|
|
25
|
Chitin Synthesis in Yeast: A Matter of Trafficking. Int J Mol Sci 2022; 23:ijms232012251. [PMID: 36293107 PMCID: PMC9603707 DOI: 10.3390/ijms232012251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
Chitin synthesis has attracted scientific interest for decades as an essential part of fungal biology and for its potential as a target for antifungal therapies. While this interest remains, three decades ago, pioneering molecular studies on chitin synthesis regulation identified the major chitin synthase in yeast, Chs3, as an authentic paradigm in the field of the intracellular trafficking of integral membrane proteins. Over the years, researchers have shown how the intracellular trafficking of Chs3 recapitulates all the steps in the intracellular trafficking of integral membrane proteins, from their synthesis in the endoplasmic reticulum to their degradation in the vacuole. This trafficking includes specific mechanisms for sorting in the trans-Golgi network, regulated endocytosis, and endosomal recycling at different levels. This review summarizes the work carried out on chitin synthesis regulation, mostly focusing on Chs3 as a molecular model to study the mechanisms involved in the control of the intracellular trafficking of proteins.
Collapse
|
26
|
Cryptococcus neoformans Infection in the Central Nervous System: The Battle between Host and Pathogen. J Fungi (Basel) 2022; 8:jof8101069. [PMID: 36294634 PMCID: PMC9605252 DOI: 10.3390/jof8101069] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Cryptococcus neoformans (C. neoformans) is a pathogenic fungus with a global distribution. Humans become infected by inhaling the fungus from the environment, and the fungus initially colonizes the lungs. If the immune system fails to contain C. neoformans in the lungs, the fungus can disseminate to the blood and invade the central nervous system, resulting in fatal meningoencephalitis particularly in immunocompromised individuals including HIV/AIDS patients. Following brain invasion, C. neoformans will encounter host defenses involving resident as well as recruited immune cells in the brain. To overcome host defenses, C. neoformans possesses multiple virulence factors capable of modulating immune responses. The outcome of the interactions between the host and C. neoformans will determine the disease progression. In this review, we describe the current understanding of how C. neoformans migrates to the brain across the blood–brain barrier, and how the host immune system responds to the invading organism in the brain. We will also discuss the virulence factors that C. neoformans uses to modulate host immune responses.
Collapse
|
27
|
Moreira-Walsh B, Ragsdale A, Lam W, Upadhya R, Xu E, Lodge JK, Donlin MJ. Membrane Integrity Contributes to Resistance of Cryptococcus neoformans to the Cell Wall Inhibitor Caspofungin. mSphere 2022; 7:e0013422. [PMID: 35758672 PMCID: PMC9429927 DOI: 10.1128/msphere.00134-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/01/2022] [Indexed: 01/20/2023] Open
Abstract
The fungal pathogen Cryptococcus neoformans causes up to 278 000 infections each year globally, resulting in up to 180,000 deaths annually, mostly impacting immunocompromised people. Therapeutic options for C. neoformans infections are very limited. Caspofungin, a member of the echinocandin class of antifungals, is generally well tolerated but clinically ineffective against C. neoformans. We sought to identify biological processes that can be targeted to render the cell more susceptible to echinocandins by screening the available libraries of gene deletion mutants made in the KN99α background for caspofungin sensitivity. We adapted a Candida albicans fungal biofilm assay for the growth characteristics of C. neoformans and systematically screened 4,030 individual gene deletion mutants in triplicate plate assays. We identified 25 strains that showed caspofungin sensitivity. We followed up with a dose dependence assay, and 17 of the 25 were confirmed sensitive, 5 of which were also sensitive in an agar plate assay. We made new deletion mutant strains for four of these genes: CFT1, encoding an iron transporter; ERG4, encoding a sterol desaturase; MYO1, encoding a myosin heavy chain; and YSP2, encoding a sterol transporter. All were more sensitive to membrane stress and showed significantly increased sensitivity to caspofungin at higher temperatures. Surprisingly, none showed any obvious cell wall defects such as would be expected for caspofungin-sensitive strains. Our microscopy analyses suggested that loss of membrane integrity contributed to the caspofungin sensitivity, either by allowing more caspofungin to enter or remain in the cell or by altering the location or orientation of the enzyme target to render it more susceptible to inhibition. IMPORTANCE The intrinsic resistance of Cryptococcus neoformans to the cell wall inhibitor caspofungin limits the available therapies for treating cryptococcal infections. We screened a collection of more than 4,000 gene deletion strains for altered caspofungin sensitivity to identify biological processes that could be targeted to render the cell more susceptible to caspofungin. We identified multiple genes with an effect on caspofungin susceptibility and found that they were associated with altered membrane permeability rather than the expected cell wall defects. This suggests that targeting these genes or other genes affecting membrane permeability is a viable path for developing novel therapies for treating this global fungal pathogen.
Collapse
Affiliation(s)
- Brenda Moreira-Walsh
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Abigail Ragsdale
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Woei Lam
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajendra Upadhya
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Evan Xu
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer K. Lodge
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maureen J. Donlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
28
|
Chitosan Is Necessary for the Structure of the Cell Wall, and Full Virulence of Ustilago maydis. J Fungi (Basel) 2022; 8:jof8080813. [PMID: 36012801 PMCID: PMC9409902 DOI: 10.3390/jof8080813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
Smut fungi comprise a large group of biotrophic phytopathogens infecting important crops, such as wheat and corn. U. maydis is a plant pathogenic fungus responsible for common smut in maize and teocintle. Through our analysis of the transcriptome of the yeast-to-mycelium dimorphic transition at acid pH, we determined the number of genes encoding chitin deacetylases of the fungus, and observed that the gene encoding one of them (UMAG_11922; CDA1) was the only one up-regulated. The mutation of this gene and the analysis of the mutants revealed that they contained reduced amounts of chitosan, were severely affected in their virulence, and showed aberrant mycelial morphology when grown at acid pH. When the CDA1 gene was reinserted into the mutants by the use of an autonomous replication plasmid, virulence and chitosan levels were recovered in the retro mutant strains, indicating that the CDA1 gene was involved in these features. These data revealed that chitosan plays a crucial role in the structure and morphogenesis of the cell wall during mycelial development of the fungus, and that in its absence, the cell wall becomes altered and is unable to support the stress imposed by the defense mechanism mounted on by the plant host during the infection process.
Collapse
|
29
|
Abstract
Cryptococcosis is a disease caused by the pathogenic fungi Cryptococcus neoformans and Cryptococcus gattii, both environmental fungi that cause severe pneumonia and may even lead to cryptococcal meningoencephalitis. Although C. neoformans affects more fragile individuals, such as immunocompromised hosts through opportunistic infections, C. gattii causes a serious indiscriminate primary infection in immunocompetent individuals. Typically seen in tropical and subtropical environments, C. gattii has increased its endemic area over recent years, largely due to climatic factors that favor contagion in warmer climates. It is important to point out that not only C. gattii, but the Cryptococcus species complex produces a polysaccharidic capsule with immunomodulatory properties, enabling the pathogenic species of Cryptococccus to subvert the host immune response during the establishment of cryptococcosis, facilitating its dissemination in the infected organism. C. gattii causes a more severe and difficult-to-treat infection, with few antifungals eliciting an effective response during chronic treatment. Much of the immunopathology of this cryptococcosis is still poorly understood, with most studies focusing on cryptococcosis caused by the species C. neoformans. C. gattii became more important in the epidemiological scenario with the outbreaks in the Pacific Northwest of the United States, which resulted in phylogenetic studies of the virulent variant responsible for the severe infection in the region. Since then, the study of cryptococcosis caused by C. gattii has helped researchers understand the immunopathological aspects of different variants of this pathogen.
Collapse
|
30
|
Diniz-Lima I, da Fonseca LM, dos Reis JS, Rodrigues da Costa Santos MA, da Costa KM, do Nascimento Santos CA, Barcelos PM, Guimarães-Pinto K, Filardy AA, Freire-de-Lima ME, Decote-Ricardo D, Morrot A, Freire-de-Lima CG, Freire-de-Lima L. The Sweet Side of Fungal Infections: Structural Glycan Diversity and Its Importance for Pathogenic Adaptation. MEDICINES (BASEL, SWITZERLAND) 2022; 9:medicines9060037. [PMID: 35736250 PMCID: PMC9230512 DOI: 10.3390/medicines9060037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022]
Abstract
Fungal infections are the most common secondary infections in debilitated individuals in a state of chronic disease or immunosuppression. Despite this, most fungal infections are neglected, mainly due to the lower frequency of their more severe clinical forms in immunocompetent individuals with a healthy background. However, over the past few years, several cases of severe fungal infections in healthy individuals have provoked a change in the epidemiological dynamics of fungal infections around the world, both due to recurrent outbreaks in previously infrequent regions and the greater emergence of more pathogenic fungal variants affecting healthy individuals, such as in the Cryptococcus genus. Therefore, before the arrival of a scenario of prevalent severe fungal infections, it is necessary to assess more carefully what are the real reasons for the increased incidence of fungal infection globally. What are the factors that are currently contributing to this new possible epidemiological dynamic? Could these be of a structural nature? Herein, we propose a discussion based on the importance of the virulence factors of glycoconjugate composition in the adaptation of pathogenic fungal species into the current scenario of increasing severity of these infections.
Collapse
Affiliation(s)
- Israel Diniz-Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Leonardo Marques da Fonseca
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Jhenifer Santos dos Reis
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Marcos André Rodrigues da Costa Santos
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Kelli Monteiro da Costa
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Carlos Antonio do Nascimento Santos
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Pedro Marçal Barcelos
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| | - Kamila Guimarães-Pinto
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (K.G.-P.); (A.A.F.)
| | - Alessandra Almeida Filardy
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (K.G.-P.); (A.A.F.)
| | - Marco Edilson Freire-de-Lima
- Instituto de Química, Departamento de Química Orgânica, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro 23890-000, Brazil;
| | - Debora Decote-Ricardo
- Departamento de Microbiologia e Imunologia Veterinária, Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro 23890-000, Brazil;
| | - Alexandre Morrot
- Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, Brazil;
| | - Celio Geraldo Freire-de-Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
- Correspondence: ; Tel./Fax: +55-21-3938-6646
| | - Leonardo Freire-de-Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil; (I.D.-L.); (L.M.d.F.); (J.S.d.R.); (M.A.R.d.C.S.); (K.M.d.C.); (C.A.d.N.S.); (P.M.B.); (L.F.-d.-L.)
| |
Collapse
|
31
|
Meshram S, Gogoi R, Bashyal BM, Kumar A, Mandal PK, Hossain F. Comparative Transcriptome Analysis of Fungal Pathogen Bipolaris maydis to Understand Pathogenicity Behavior on Resistant and Susceptible Non-CMS Maize Genotypes. Front Microbiol 2022; 13:837056. [PMID: 35572625 PMCID: PMC9100685 DOI: 10.3389/fmicb.2022.837056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Bipolaris maydis is pathogen of maize which causes maydis leaf blight disease. In India major losses occur due to the B. maydis race “O” pathogen, whereas in other parts of the world, major losses are due to the race “T” pathogen. In the present study, we conducted an in planta transcriptomics study of the B. maydis race “O” pathogen after infection on non-CMS maize resistant and susceptible genotypes by mRNA sequencing to understand the molecular basis of pathogenicity for better management of the pathogen. Approximately 23.4 GB of mRNA-seq data of B. maydis were obtained from both resistant and susceptible maize backgrounds for fungus. Differentially expressed genes (DEGs) analysis of B. maydis in two different genetic backgrounds suggested that the majority of highly DEGs were associated with mitochondrial, cell wall and chitin synthesis, sugar metabolism, peroxidase activity, mitogen-activated protein kinase (MAPK) activity, and shikimate dehydrogenase. KEGG analysis showed that the biosynthetic pathways for secondary metabolism, antibiotics, and carbon metabolism of fungus were highly enriched, respectively, in susceptible backgrounds during infection. Previous studies in other host pathogen systems suggest that these genes play a vital role in causing disease in their host plants. Our study is probably the first transcriptome study of the B. maydis race “O” pathogen and provides in-depth insight of pathogenicity on the host.
Collapse
Affiliation(s)
- Shweta Meshram
- Division of Plant Pathology, Indian Council of Agricultural Research (ICAR)-Indian Agricultural Research Institute, New Delhi, India
| | - Robin Gogoi
- Division of Plant Pathology, Indian Council of Agricultural Research (ICAR)-Indian Agricultural Research Institute, New Delhi, India
- *Correspondence: Robin Gogoi,
| | - Bishnu Maya Bashyal
- Division of Plant Pathology, Indian Council of Agricultural Research (ICAR)-Indian Agricultural Research Institute, New Delhi, India
| | - Aundy Kumar
- Division of Plant Pathology, Indian Council of Agricultural Research (ICAR)-Indian Agricultural Research Institute, New Delhi, India
| | - Pranab Kumar Mandal
- Indian Council of Agricultural Research (ICAR)-National Institute for Plant Biotechnology, New Delhi, India
| | - Firoz Hossain
- Division of Genetics, Indian Council of Agricultural Research (ICAR)-Indian Agricultural Research Institute, New Delhi, India
| |
Collapse
|
32
|
Berguson HP, Caulfield LW, Price MS. Influence of Pathogen Carbon Metabolism on Interactions With Host Immunity. Front Cell Infect Microbiol 2022; 12:861405. [PMID: 35372116 PMCID: PMC8968422 DOI: 10.3389/fcimb.2022.861405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
Cryptococcus neoformans is a ubiquitous opportunistic fungal pathogen typically causing disease in immunocompromised individuals and is globally responsible for about 15% of AIDS-related deaths annually. C. neoformans first causes pulmonary infection in the host and then disseminates to the brain, causing meningoencephalitis. The yeast must obtain and metabolize carbon within the host in order to survive in the central nervous system and cause disease. Communication between pathogen and host involves recognition of multiple carbon-containing compounds on the yeast surface: polysaccharide capsule, fungal cell wall, and glycosylated proteins comprising the major immune modulators. The structure and function of polysaccharide capsule has been studied for the past 70 years, emphasizing its role in virulence. While protected by the capsule, fungal cell wall has likewise been a focus of study for several decades for its role in cell integrity and host recognition. Associated with both of these major structures are glycosylated proteins, which exhibit known immunomodulatory effects. While many studies have investigated the role of carbon metabolism on virulence and survival within the host, the precise mechanism(s) affecting host-pathogen communication remain ill-defined. This review summarizes the current knowledge on mutants in carbon metabolism and their effect on the host immune response that leads to changes in pathogen recognition and virulence. Understanding these critical interactions will provide fresh perspectives on potential treatments and the natural history of cryptococcal disease.
Collapse
Affiliation(s)
- Hannah P. Berguson
- Department of Anatomical Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, VA, United States
| | - Lauren W. Caulfield
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, United States
| | - Michael S. Price
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, VA, United States
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States
- *Correspondence: Michael S. Price,
| |
Collapse
|
33
|
Vélez N, Vega-Vela N, Muñoz M, Gómez P, Escandón P, Ramírez JD, Zaragoza O, Monteoliva Diaz L, Parra-Giraldo CM. Deciphering the Association among Pathogenicity, Production and Polymorphisms of Capsule/Melanin in Clinical Isolates of Cryptococcus neoformans var. grubii VNI. J Fungi (Basel) 2022; 8:245. [PMID: 35330247 PMCID: PMC8950468 DOI: 10.3390/jof8030245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Cryptococcus neoformans is an opportunistic fungal pathogen that can cause meningitis in immunocompromised individuals. The objective of this work was to study the relationship between the phenotypes and genotypes of isolates of clinical origin from different cities in Colombia. METHODS Genome classification of 29 clinical isolates of C. neoformans var. grubii was performed using multilocus sequence typing (MLST), and genomic sequencing was used to genotype protein-coding genes. Pathogenicity was assessed in a larval model, and melanin production and capsule size were evaluated in vitro and in vivo. RESULTS Eleven MLST sequence types (STs) were found, the most frequent being ST69 (n = 9), ST2, ST93, and ST377 (each with n = 4). In the 29 isolates, different levels of pigmentation, capsule size and pathogenicity were observed. Isolates classified as highly pathogenic showed a tendency to exhibit larger increases in capsule size. In the analysis of polymorphisms, 48 non-synonymous variants located in the predicted functional domains of 39 genes were found to be associated with capsule size change, melanin, or pathogenicity. CONCLUSIONS No clear patterns were found in the analysis of the phenotype and genotype of Cryptococcus. However, the data suggest that the increase in capsule size is a key variable for the differentiation of pathogenic isolates, regardless of the method used for its induction.
Collapse
Affiliation(s)
- Nórida Vélez
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Nelson Vega-Vela
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia
| | - Paola Gómez
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Patricia Escandón
- Grupo de Microbiología, Instituto Nacional de Salud, Bogotá 111321, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Oscar Zaragoza
- Mycology Reference Laboratory National Centre for Microbiology, Instituto de Salud Carlos III, 28222 Madrid, Spain
| | - Lucía Monteoliva Diaz
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Claudia-Marcela Parra-Giraldo
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| |
Collapse
|
34
|
Chitosan Production by Fungi: Current State of Knowledge, Future Opportunities and Constraints. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8020076] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Conventionally, the commercial supply of chitin and chitosan relies on shellfish wastes as the extraction sources. However, the fungal sources constitute a valuable option, especially for biomedical and pharmaceutical applications, due to the batch-to-batch unsteady properties of chitin and chitosan from conventional ones. Fungal production of these glycans is not affected by seasonality enables accurate process control and, consequently, more uniform properties of the obtained product. Moreover, liquid and solid production media often are derived from wastes, thus enabling the application of circular economy criteria and improving the process economics. The present review deals with fungal chitosan production processes focusing on waste-oriented and integrated production processes. In doing so, contrary to other reviews that used a genus-specific approach for organizing the available information, the present one bases the discussion on the bioprocess typology. Finally, the main process parameters affecting chitosan production and their interactions are critically discussed.
Collapse
|
35
|
Upadhya R, Lam WC, Hole CR, Parchment D, Lee CK, Specht CA, Levitz SM, Lodge JK. Cryptococcus neoformans Cda1 and Cda2 coordinate deacetylation of chitin during infection to control fungal virulence. ACTA ACUST UNITED AC 2021; 7:100066. [PMID: 34712865 PMCID: PMC8529172 DOI: 10.1016/j.tcsw.2021.100066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022]
Abstract
Chitosan, a deacetylated form of chitin, is required for the virulence of Cryptococcus neoformans. There are three chitin deacetylase genes (CDA) that are essential for chitosan production, and deletion of all three genes results in the absence of chitosan, loss of virulence, and induction of a protective host response when used as a vaccine. Cda1 plays a major role in deacetylating chitin during pulmonary infection of CBA/J mice. Inoculation with the cda1Δ strain did not lead to a lethal infection. However, the infection was not cleared. The persistence of the fungus in the host suggests that chitin is still being deacetylated by Cda2 and/or Cda3. To test this hypothesis, we subjected strains deleted of two CDA genes to fungal virulence in CBA/J, C57BL/6 and BALB/c and found that cda1Δcda2Δ was avirulent in all mouse lines, as evidenced by its complete clearance. Consistent with the major role of Cda1 in CBA/J, we found that cda2Δcda3Δ was as virulent as its wild-type progenitor KN99. On the other hand, cda1Δcda3Δ displayed virulence comparable to that of cda1Δ. The virulence of each mutant correlates with the amount of chitosan produced when grown under host-mimicking culture conditions. In addition, the avirulence of cda1Δcda2Δ was followed by the induction of a protective immune response in C57BL/6 and CBA/J mice, when a live or heat-killed form of the mutant was used as a vaccine respectively. Taken together, these data imply that, in C. neoformans, coordinated activity of both Cda1 and Cda2 is essential for mediating fungal virulence.
Collapse
Affiliation(s)
- Rajendra Upadhya
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Woei C Lam
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Camaron R Hole
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Danealle Parchment
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chrono K Lee
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Charles A Specht
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jennifer K Lodge
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
36
|
Fernandes C, Mota M, Barros L, Dias MI, Ferreira ICFR, Piedade AP, Casadevall A, Gonçalves T. Pyomelanin Synthesis in Alternaria alternata Inhibits DHN-Melanin Synthesis and Decreases Cell Wall Chitin Content and Thickness. Front Microbiol 2021; 12:691433. [PMID: 34512569 PMCID: PMC8430343 DOI: 10.3389/fmicb.2021.691433] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
The genus Alternaria includes several of fungi that are darkly pigmented by DHN-melanin. These are pathogenic to plants but are also associated with human respiratory allergic diseases and with serious infections in immunocompromised individuals. The present work focuses on the alterations of the composition and structure of the hyphal cell wall of Alternaria alternata occuring under the catabolism of L-tyrosine and L-phenylalanine when cultured in minimal salt medium (MM). Under these growing conditions, we observed the released of a brown pigment into the culture medium. FTIR analysis demonstrates that the produced pigment is chemically identical to the pigment released when the fungus is grown in MM with homogentisate acid (HGA), the intermediate of pyomelanin, confirming that this pigment is pyomelanin. In contrast to other fungi that also synthesize pyomelanin under tyrosine metabolism, A. alternata inhibits DHN-melanin cell wall accumulation when pyomelanin is produced, and this is associated with reduced chitin cell wall content. When A. alternata is grown in MM containing L-phenylalanine, a L-tyrosine percursor, pyomelanin is synthesized but only at trace concentrations and A. alternata mycelia display an albino-like phenotype since DHN-melanin accumulation is inhibited. CmrA, the transcription regulator for the genes coding for the DHN-melanin pathway, is involved in the down-regulation of DHN-melanin synthesis when pyomelanin is being synthetized, since the CMRA gene and genes of the enzymes involved in DHN-melanin synthesis pathway showed a decreased expression. Other amino acids do not trigger pyomelanin synthesis and DHN-melanin accumulation in the cell wall is not affected. Transmission and scanning electron microscopy show that the cell wall structure and surface decorations are altered in L-tyrosine- and L-phenylalanine-grown fungi, depending on the pigment produced. In summary, growth in presence of L-tyrosine and L-phenylalanine leads to pigmentation and cell wall changes, which could be relevant to infection conditions where these amino acids are expected to be available.
Collapse
Affiliation(s)
- Chantal Fernandes
- CNC—Center for Neuroscience and Cell Biology of Coimbra, Coimbra, Portugal
| | - Marta Mota
- CNC—Center for Neuroscience and Cell Biology of Coimbra, Coimbra, Portugal
- FMUC—Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lillian Barros
- Mountain Research Center (CIMO), Polytechnic Institute of Bragança, Bragança, Portugal
- Laboratory of Separation and Reaction Engineering - Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Bragança, Bragança, Portugal
| | - Maria Inês Dias
- Mountain Research Center (CIMO), Polytechnic Institute of Bragança, Bragança, Portugal
- Laboratory of Separation and Reaction Engineering - Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Bragança, Bragança, Portugal
| | - Isabel C. F. R. Ferreira
- Mountain Research Center (CIMO), Polytechnic Institute of Bragança, Bragança, Portugal
- Laboratory of Separation and Reaction Engineering - Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Bragança, Bragança, Portugal
| | - Ana P. Piedade
- Centre for Mechanical Engineering, Materials and Processes, Department of Mechanical Engineering, University of Coimbra, Coimbra, Portugal
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Teresa Gonçalves
- CNC—Center for Neuroscience and Cell Biology of Coimbra, Coimbra, Portugal
- FMUC—Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
37
|
Imanishi-Shimizu Y, Kamogawa Y, Shimada Y, Shimizu K. A capsule-associated gene of Cryptococcus neoformans, CAP64, is involved in pH homeostasis. MICROBIOLOGY-SGM 2021; 167. [PMID: 34125663 PMCID: PMC8374607 DOI: 10.1099/mic.0.001029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The CAP64 gene is known to be involved in capsule formation in the basidiomycete yeast Cryptococcus neoformans. A null mutant of CAP64, Δcap64, lacks a capsule around the cell wall and its acidic organelles are not stained with quinacrine. In order to clarify whether the Cap64 protein indeed maintains vacuole or vesicle acidification, so that the vesicle containing the capsule polysaccharide or DBB substrate are transported to the cell membrane side, the relationship between CAP64 and intracellular transport genes and between CAP64 and enzyme-secretion activity were analysed. Laccase activity was higher in the Δcap64 strain than in the wild-type strain, and the transcriptional levels of SAV1 and VPH1 were also higher in the Δcap64 strain than in the wild-type strain. The intracellular localization of the Cap64 protein was analysed by overexpressing an mCherry-tagged Cap64 and observing its fluorescence. The Cap64 protein was accumulated within cells in a patch-like manner. The quinacrine-stained cells were observed to analyse the acidified cell compartments; quinacrine was found to be accumulated in a patch-like manner, with the patches overlapping the fluorescence of CAP64-mCherry fusion protein. Quinacrine was thus accumulated in a patch-like fashion in the cells, and the mCherry-tagged Cap64 protein position was consistent with the position of quinacrine accumulation in cells. These results suggest that CAP64 might be involved in intracellular acidification and vesicle secretion via exocytosis.
Collapse
Affiliation(s)
- Yumi Imanishi-Shimizu
- Department of Bioscience, College of Science and Engineering, Kanto Gakuin University, 1-50-1 Mutsuura-higashi, Kanazawa-ku, Yokohama 236-8501, Japan
| | - Yukina Kamogawa
- Department of Bioscience, College of Science and Engineering, Kanto Gakuin University, 1-50-1 Mutsuura-higashi, Kanazawa-ku, Yokohama 236-8501, Japan
| | - Yukino Shimada
- Department of Bioscience, College of Science and Engineering, Kanto Gakuin University, 1-50-1 Mutsuura-higashi, Kanazawa-ku, Yokohama 236-8501, Japan
| | - Kiminori Shimizu
- Department of Biological Science and Technology, Tokyo University of Science, Niijuku 6-3-1, Katsushika-ku, Tokyo 125-8585, Japan
| |
Collapse
|
38
|
Liu M, Tang H, Jiang H, Li J, Yan S, Wang Q. Effects of air discharge on surface charges and cell walls of Fusarium oxysporum. Int Microbiol 2021; 24:415-425. [PMID: 33963940 DOI: 10.1007/s10123-020-00157-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/28/2020] [Accepted: 12/29/2020] [Indexed: 10/21/2022]
Abstract
Air discharge showed significant inhibition on mycelial growth and spore germination of Fusarium oxysporum, one of the main spoilage fungi in post-harvest lotus roots which is an important economic aquatic vegetable in China. However, the antimicrobial mechanism of air discharge is not clear yet. In the present study, the effects of air discharge on F. oxysporum separated from post-harvest rotten lotus roots were characterized by analyzing surface charges, cell wall permeability, and changes in chitin and chitosan including surface morphology, functional groups, degree of deacetylation, crystallinity, and C/N ratio. After air discharge treatments, alkaline phosphatase leak assay revealed that cell wall permeability of F. oxysporum was magnified. What's more, zeta potentials of F. oxysporum increased and negative charges on cell surfaces decreased. The ordered and compact molecular arrangements of chitin and chitosan in cell walls of F. oxysporum were reduced. The deacetylation degree of chitin and chitosan increased, and the C/N ratios of chitin and chitosan decreased. It was concluded from these results that air discharge caused the transformation in structures of chitin and chitosan, resulting in the exposure of positively charged amino groups and decrease of negative charges on cell surfaces which brought damage to the structure and function of F. oxysporum's cell walls.
Collapse
Affiliation(s)
- Mengdie Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Hui Tang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huiwen Jiang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jie Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China. .,Aquatic Vegetable Preservation&Processing Technology Engineering Center of Hubei Province, Wuhan, Hubei, China. .,Engineering Research Center of Ministry of Education for Green Development of Aquatic Biological Industry in Yangtze River Economic Belt, Wuhan, Hubei, China.
| | - Shoulei Yan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.,Aquatic Vegetable Preservation&Processing Technology Engineering Center of Hubei Province, Wuhan, Hubei, China.,Engineering Research Center of Ministry of Education for Green Development of Aquatic Biological Industry in Yangtze River Economic Belt, Wuhan, Hubei, China
| | - Qingzhang Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.,Aquatic Vegetable Preservation&Processing Technology Engineering Center of Hubei Province, Wuhan, Hubei, China
| |
Collapse
|
39
|
Geetha N, Bhavya G, Abhijith P, Shekhar R, Dayananda K, Jogaiah S. Insights into nanomycoremediation: Secretomics and mycogenic biopolymer nanocomposites for heavy metal detoxification. JOURNAL OF HAZARDOUS MATERIALS 2021; 409:124541. [PMID: 33223321 DOI: 10.1016/j.jhazmat.2020.124541] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/02/2020] [Accepted: 11/06/2020] [Indexed: 05/21/2023]
Abstract
Our environment thrives on the subtle balance achieved by the forever cyclical nature of building and rebuilding life through natural processes. Fungi, being the evident armor of bioremediation, is the indispensable element of the soil food web, contribute to be the nature's most dynamic arsenal with non-specific enzymes like peroxidase (POX), glutathione peroxidase (GPx), catalase (CAT), superoxide dismutase (SOD), non-enzymatic compounds like thiol (-SH) groups and non-protein compounds such as glutathione (GSH) and metallothionein (MT). Recently, the area of nanomycoremediation has been gaining momentum as a powerful tool for environmental clean-up strategies with its ability to detoxify heavy metals with its unique characteristics to adapt mechanisms such as biosorption, bioconversion, and biodegradation to harmless end products. The insight into the elaborate secretomic processes provides us with huge opportunities for creating a magnificent living bioremediation apparatus. This review discusses the scope and recent advances in the lesser understood area, nanomycoremediation, the state-of-the-art, innovative, cost-effective and promising tool for detoxification of heavy metal pollutants and focuses on the metabolic capabilities and secretomics with nanobiotechnological interventions.
Collapse
Affiliation(s)
- Nagaraja Geetha
- Nanobiotechnology Laboratory, Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India
| | - Gurulingaiah Bhavya
- Nanobiotechnology Laboratory, Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India
| | - Padukana Abhijith
- Nanobiotechnology Laboratory, Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India
| | - Ravikant Shekhar
- Nanobiotechnology Laboratory, Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India
| | - Karigowda Dayananda
- Nanobiotechnology Laboratory, Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India
| | - Sudisha Jogaiah
- Laboratory of Plant Healthcare and Diagnostics, P.G. Department of Biotechnology and Microbiology, Karnatak University, Dharwad 580003, Karnataka, India.
| |
Collapse
|
40
|
Liu S, Youngchim S, Zamith-Miranda D, Nosanchuk JD. Fungal Melanin and the Mammalian Immune System. J Fungi (Basel) 2021; 7:jof7040264. [PMID: 33807336 PMCID: PMC8066723 DOI: 10.3390/jof7040264] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Melanins are ubiquitous complex polymers that are commonly known in humans to cause pigmentation of our skin. Melanins are also present in bacteria, fungi, and helminths. In this review, we will describe the diverse interactions of fungal melanin with the mammalian immune system. We will particularly focus on Cryptococcus neoformans and also discuss other major melanotic pathogenic fungi. Melanin interacts with the immune system through diverse pathways, reducing the effectiveness of phagocytic cells, binding effector molecules and antifungals, and modifying complement and antibody responses.
Collapse
Affiliation(s)
- Sichen Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.L.); (D.Z.-M.)
| | - Sirida Youngchim
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Daniel Zamith-Miranda
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.L.); (D.Z.-M.)
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joshua D. Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.L.); (D.Z.-M.)
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence:
| |
Collapse
|
41
|
Araújo GRDS, Alcantara CDL, Rodrigues N, de Souza W, Pontes B, Frases S. Ultrastructural Study of Cryptococcus neoformans Surface During Budding Events. Front Microbiol 2021; 12:609244. [PMID: 33732220 PMCID: PMC7957021 DOI: 10.3389/fmicb.2021.609244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/10/2021] [Indexed: 11/25/2022] Open
Abstract
Cryptococcus neoformans is a fungal pathogen that causes life-threatening infections in immunocompromised individuals. It is surrounded by three concentric structures that separate the cell from the extracellular space: the plasma membrane, the cell wall and the polysaccharide (PS) capsule. Although several studies have revealed the chemical composition of these structures, little is known about their ultrastructural organization and remodeling during C. neoformans budding events. Here, by combining the latest and most accurate light and electron microscopy techniques, we describe the morphological remodeling that occurs among the capsule, cell wall and plasma membrane during budding in C. neoformans. Our results show that the cell wall deforms to generate a specialized region at one of the cell’s poles. This region subsequently begins to break into layers that are slightly separated from each other and with thick tips. We also observe a reorganization of the capsular PS around the specialized regions. While daughter cells present their PS fibers aligned in the direction of budding, mother cells show a similar pattern but in the opposite direction. Also, daughter cells form multilamellar membrane structures covering the continuous opening between both cells. Together, our findings provide compelling ultrastructural evidence for C. neoformans surface remodeling during budding, which may have important implications for future studies exploring these remodeled specialized regions as drug-targets against cryptococcosis.
Collapse
Affiliation(s)
- Glauber R de S Araújo
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina de L Alcantara
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Noêmia Rodrigues
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Pontes
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Pinças Óticas (LPO-COPEA), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Stempinski PR, Zielinski JM, Dbouk NH, Huey ES, McCormack EC, Rubin AM, Chandrasekaran S, Kozubowski L. Genetic contribution to high temperature tolerance in Cryptococcus neoformans. Genetics 2021; 217:1-15. [PMID: 33683363 PMCID: PMC8045695 DOI: 10.1093/genetics/iyaa009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/24/2020] [Indexed: 11/12/2022] Open
Abstract
The human fungal pathogen Cryptococcus neoformans relies on a complex signaling network for the adaptation and survival at the host temperature. Protein phosphatase calcineurin is central to proliferation at 37°C but its exact contributions remain ill-defined. To better define genetic contributions to the C. neoformans temperature tolerance, 4031 gene knockouts were screened for genes essential at 37°C and under conditions that keep calcineurin inactive. Identified 83 candidate strains, potentially sensitive to 37°C, were subsequently subject to technologically simple yet robust assay, in which cells are exposed to a temperature gradient. This has resulted in identification of 46 genes contributing to the maximum temperature at which C. neoformans can proliferate (Tmax). The 46 mutants, characterized by a range of Tmax on drug-free media, were further assessed for Tmax under conditions that inhibit calcineurin, which led to identification of several previously uncharacterized knockouts exhibiting synthetic interaction with the inhibition of calcineurin. A mutant that lacked septin Cdc11 was among those with the lowest Tmax and failed to proliferate in the absence of calcineurin activity. To further define connections with calcineurin and the role for septins in high temperature growth, the 46 mutants were tested for cell morphology at 37°C and growth in the presence of agents disrupting cell wall and cell membrane. Mutants sensitive to calcineurin inhibition were tested for synthetic lethal interaction with deletion of the septin-encoding CDC12 and the localization of the septin Cdc3-mCherry. The analysis described here pointed to previously uncharacterized genes that were missed in standard growth assays indicating that the temperature gradient assay is a valuable complementary tool for elucidating the genetic basis of temperature range at which microorganisms proliferate.
Collapse
Affiliation(s)
- Piotr R Stempinski
- Department of Genetics & Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, SC 29634, USA
| | - Jessica M Zielinski
- Department of Genetics & Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, SC 29634, USA
| | - Nadir H Dbouk
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Elizabeth S Huey
- Department of Genetics & Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, SC 29634, USA
| | - Ellen C McCormack
- Department of Genetics & Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, SC 29634, USA
| | - Alexander M Rubin
- Department of Genetics & Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, SC 29634, USA
| | | | - Lukasz Kozubowski
- Department of Genetics & Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
43
|
Silva VKA, Bhattacharya S, Oliveira NK, Savitt AG, Zamith-Miranda D, Nosanchuk JD, Fries BC. Replicative Aging Remodels the Cell Wall and Is Associated with Increased Intracellular Trafficking in Human Pathogenic Yeasts. mBio 2021; 13:e0019022. [PMID: 35164553 PMCID: PMC8844920 DOI: 10.1128/mbio.00190-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 01/08/2023] Open
Abstract
Replicative aging is an underexplored field of research in medical mycology. Cryptococcus neoformans (Cn) and Candida glabrata (Cg) are dreaded fungal pathogens that cause fatal invasive infections. The fungal cell wall is essential for yeast viability and pathogenesis. In this study, we provide data characterizing age-associated modifications to the cell wall of Cn and Cg. Here, we report that old yeast cells upregulate genes of cell wall biosynthesis, leading to cell wall reorganization and increased levels of all major components, including glucan, chitin, and its derivatives, as well as mannan. This results in a significant thickening of the cell wall in aged cells. Old-generation yeast cells exhibited drastic ultrastructural changes, including the presence of abundant vesicle-like particles in the cytoplasm, and enlarged vacuoles with altered pH homeostasis. Our findings suggest that the cell wall modifications could be enabled by augmented intracellular trafficking. This work furthers our understanding of the cell phenotype that emerges during aging. It highlights differences in these two fungal pathogens and elucidates mechanisms that explain the enhanced resistance of old cells to antifungals and phagocytic attacks. IMPORTANCE Cryptococcus neoformans and Candida glabrata are two opportunistic human fungal pathogens that cause life-threatening diseases. During infection, both microorganisms have the ability to persist for long periods, and treatment failure can occur even if standard testing identifies the yeasts to be sensitive to antifungals. Replicative life span is a trait that is measured by the number of divisions a cell undergoes before death. Aging in fungi is associated with enhanced tolerance to antifungals and resistance to phagocytosis, and characterization of old cells may help identify novel antifungal targets. The cell wall remains an attractive target for new therapies because it is essential for fungi and is not present in humans. This study shows that the organization of the fungal cell wall changes remarkably during aging and becomes thicker and is associated with increased intracellular trafficking as well as the alteration of vacuole morphology and pH homeostasis.
Collapse
Affiliation(s)
- Vanessa K. A. Silva
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Somanon Bhattacharya
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Natalia Kronbauer Oliveira
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Anne G. Savitt
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Daniel Zamith-Miranda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Joshua D. Nosanchuk
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Bettina C. Fries
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Veterans Administration Medical Center, Northport, New York, USA
| |
Collapse
|
44
|
Li C, Hou S, Ma X, Li J, Huo L, Zhang P, Hao X, Zhu X. Epigenetic regulation of virulence and the transcription of ribosomal protein genes involves a YEATS family protein in Cryptococcus deneoformans. FEMS Yeast Res 2021; 21:6095727. [PMID: 33440003 DOI: 10.1093/femsyr/foab001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/11/2021] [Indexed: 01/18/2023] Open
Abstract
Epigenetic marks or post-translational modifications on histones have important regulatory roles in gene expression in eukaryotic organisms. The epigenetic regulation of gene expression in the pathogenic yeast Cryptococcus deneoformans remains largely undetermined. The YEATS domain proteins are readers of crotonylated lysine residues in histones. Here, we reported the identification of a single-copy gene putatively coding for a YEATS domain protein (Yst1) in C. deneoformans. To define its function, we created a mutant strain, yst1Δ, using CRISPR-Cas9 editing. yst1Δ exhibited defects in phenotype, for instance, it was hypersensitive to osmotic stress in the presence of 1.3 M NaCl or KCl. Furthermore, it was hypersensitive to 1% Congo red, suggesting defects in the cell wall. Interestingly, RNA-seq data revealed that Yst1p was critical for the expression of genes encoding the ribosomal proteins, that is, most were expressed with significantly lower levels of mRNA in yst1Δ than in the wild-type strain. The mutant strain was hypersensitive to low temperature and anti-ribosomal drugs, which we putatively attribute to the impairment in ribosomal function. In addition, the yst1Δ strain was less virulent to Galleria mellonella. These results generally suggest that Yst1, as a histone modification reader, might be a key coordinator of the transcriptome of this human pathogen. Yst1 could be a potential target for novel antifungal drugs, which might lead to significant developments in the clinical treatment of cryptococcosis.
Collapse
Affiliation(s)
- Chenxi Li
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Shaonan Hou
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Xiaoyu Ma
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Jiajia Li
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Liang Huo
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Ping Zhang
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Xiaoran Hao
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Xudong Zhu
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| |
Collapse
|
45
|
Selvasekaran P, Mahalakshmi, Roshini F, Angalene LA, Chandini, Sunil T, Chidambaram R. Fungal Exopolysaccharides: Production and Biotechnological Industrial Applications in Food and Allied Sectors. Fungal Biol 2021. [DOI: 10.1007/978-3-030-68260-6_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
46
|
Treatment strategies for cryptococcal infection: challenges, advances and future outlook. Nat Rev Microbiol 2021; 19:454-466. [PMID: 33558691 PMCID: PMC7868659 DOI: 10.1038/s41579-021-00511-0] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 01/31/2023]
Abstract
Cryptococcus spp., in particular Cryptococcus neoformans and Cryptococcus gattii, have an enormous impact on human health worldwide. The global burden of cryptococcal meningitis is almost a quarter of a million cases and 181,000 deaths annually, with mortality rates of 100% if infections remain untreated. Despite these alarming statistics, treatment options for cryptococcosis remain limited, with only three major classes of drugs approved for clinical use. Exacerbating the public health burden is the fact that the only new class of antifungal drugs developed in decades, the echinocandins, displays negligible antifungal activity against Cryptococcus spp., and the efficacy of the remaining therapeutics is hampered by host toxicity and pathogen resistance. Here, we describe the current arsenal of antifungal agents and the treatment strategies employed to manage cryptococcal disease. We further elaborate on the recent advances in our understanding of the intrinsic and adaptive resistance mechanisms that are utilized by Cryptococcus spp. to evade therapeutic treatments. Finally, we review potential therapeutic strategies, including combination therapy, the targeting of virulence traits, impairing stress response pathways and modulating host immunity, to effectively treat infections caused by Cryptococcus spp. Overall, understanding of the mechanisms that regulate anti-cryptococcal drug resistance, coupled with advances in genomics technologies and high-throughput screening methodologies, will catalyse innovation and accelerate antifungal drug discovery.
Collapse
|
47
|
Chrissian C, Lin CPC, Camacho E, Casadevall A, Neiman AM, Stark RE. Unconventional Constituents and Shared Molecular Architecture of the Melanized Cell Wall of C. neoformans and Spore Wall of S. cerevisiae. J Fungi (Basel) 2020; 6:E329. [PMID: 33271921 PMCID: PMC7712904 DOI: 10.3390/jof6040329] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
The fungal cell wall serves as the interface between the cell and the environment. Fungal cell walls are composed largely of polysaccharides, primarily glucans and chitin, though in many fungi stress-resistant cell types elaborate additional cell wall structures. Here, we use solid-state nuclear magnetic resonance spectroscopy to compare the architecture of cell wall fractions isolated from Saccharomyces cerevisiae spores and Cryptococcus neoformans melanized cells. The specialized cell walls of these two divergent fungi are highly similar in composition. Both use chitosan, the deacetylated derivative of chitin, as a scaffold on which a polyaromatic polymer, dityrosine and melanin, respectively, is assembled. Additionally, we demonstrate that a previously identified but uncharacterized component of the S. cerevisiae spore wall is composed of triglycerides, which are also present in the C. neoformans melanized cell wall. Moreover, we identify a tyrosine-derived constituent in the C. neoformans wall that, although it is not dityrosine, is a non-pigment constituent of the cell wall. The similar composition of the walls of these two phylogenetically distant species suggests that triglycerides, polyaromatics, and chitosan are basic building blocks used to assemble highly stress-resistant cell walls and the use of these constituents may be broadly conserved in other fungal species.
Collapse
Affiliation(s)
- Christine Chrissian
- CUNY Institute for Macromolecular Assemblies, City University of New York, New York, NY 10031, USA;
- Department of Chemistry and Biochemistry, The City College of New York, New York, NY 10031, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Coney Pei-Chen Lin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Emma Camacho
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (E.C.); (A.C.)
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (E.C.); (A.C.)
| | - Aaron M. Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Ruth E. Stark
- CUNY Institute for Macromolecular Assemblies, City University of New York, New York, NY 10031, USA;
- Department of Chemistry and Biochemistry, The City College of New York, New York, NY 10031, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
48
|
de Oliveira HC, Castelli RF, Reis FCG, Rizzo J, Rodrigues ML. Pathogenic Delivery: The Biological Roles of Cryptococcal Extracellular Vesicles. Pathogens 2020; 9:pathogens9090754. [PMID: 32948010 PMCID: PMC7557404 DOI: 10.3390/pathogens9090754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are produced by all domains of life. In fungi, these structures were first described in Cryptococcus neoformans and, since then, they were characterized in several pathogenic and non-pathogenic fungal species. Cryptococcal EVs participate in the export of virulence factors that directly impact the Cryptococcus-host interaction. Our knowledge of the biogenesis and pathogenic roles of Cryptococcus EVs is still limited, but recent methodological and scientific advances have improved our understanding of how cryptococcal EVs participate in both physiological and pathogenic events. In this review, we will discuss the importance of cryptococcal EVs, including early historical studies suggesting their existence in Cryptococcus, their putative mechanisms of biogenesis, methods of isolation, and possible roles in the interaction with host cells.
Collapse
Affiliation(s)
- Haroldo C. de Oliveira
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775 CIC Curitiba/PR, Curitiba 81350-010, Brasil; (H.C.d.O.); (R.F.C.); (F.C.G.R.)
| | - Rafael F. Castelli
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775 CIC Curitiba/PR, Curitiba 81350-010, Brasil; (H.C.d.O.); (R.F.C.); (F.C.G.R.)
- Programa de Pós-Graduação em Biologia Parasitária, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4.365, Pavilhão Arthur Neiva–Manguinhos, Rio de Janeiro 21040-360, Brasil
| | - Flavia C. G. Reis
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775 CIC Curitiba/PR, Curitiba 81350-010, Brasil; (H.C.d.O.); (R.F.C.); (F.C.G.R.)
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS), Fundação Oswaldo Cruz (Fiocruz), Av. Brasil, 4036–Prédio Da Expansão–8˚ Andar–Sala 814, Rio De Janeiro 21040-361, Brasil
| | - Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France;
| | - Marcio L. Rodrigues
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775 CIC Curitiba/PR, Curitiba 81350-010, Brasil; (H.C.d.O.); (R.F.C.); (F.C.G.R.)
- Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, Brasil
- Correspondence:
| |
Collapse
|
49
|
Evolutionary analysis and protein family classification of chitin deacetylases in Cryptococcus neoformans. J Microbiol 2020; 58:805-811. [PMID: 32870486 DOI: 10.1007/s12275-020-0288-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/23/2020] [Indexed: 10/23/2022]
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen causing cryptococcal meningoencephalitis. Interestingly, the cell wall of C. neoformans contains chitosan, which is critical for its virulence and persistence in the mammalian host. C. neoformans (H99) has three chitin deacetylases (CDAs), which convert chitin to chitosan. Herein, the classification of the chitin-related protein (CRP) family focused on cryptococcal CDAs was analyzed by phylogenetics, evolutionary pressure (dN/dS), and 3D modeling. A phylogenetic tree of 110 CRPs revealed that they can be divided into two clades, CRP I and II with bootstrap values (> 99%). CRP I clade comprises five groups (Groups 1-5) with a total of 20 genes, while CRP II clade comprises sixteen groups (Groups 6-21) with a total of 90 genes. CRP I comprises only fungal CDAs, including all three C. neoformans CDAs, whereas CRP II comprises diverse CDAs from fungi, bacteria, and amoeba, along with other carbohydrate esterase 4 family proteins. All CDAs have the signal peptide, except those from group 11. Notably, CDAs with the putative O-gycosylation site possess either the glycosylphosphatidylinositol (GPI)-anchor motif for CRP I or the chitin-binding domain (CBD) for CRP II, respectively. This evolutionary conservation strongly indicates that the O-glycosylation modification and the presence of either the GPI-anchor motif or the chitin-binding domain is important for fungal CDAs to function efficiently at the cell surface. This study reveals that C. neoformans CDAs carrying GPI anchors have evolved divergently from fungal and bacterial CDAs, providing new insights into evolution and classification of CRP family.
Collapse
|
50
|
Chrissian C, Camacho E, Kelly JE, Wang H, Casadevall A, Stark RE. Solid-state NMR spectroscopy identifies three classes of lipids in Cryptococcus neoformans melanized cell walls and whole fungal cells. J Biol Chem 2020; 295:15083-15096. [PMID: 32859751 DOI: 10.1074/jbc.ra120.015201] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/20/2020] [Indexed: 12/19/2022] Open
Abstract
A primary virulence-associated trait of the opportunistic fungal pathogen Cryptococcus neoformans is the production of melanin pigments that are deposited into the cell wall and interfere with the host immune response. Previously, our solid-state NMR studies of isolated melanized cell walls (melanin "ghosts") revealed that the pigments are strongly associated with lipids, but their identities, origins, and potential roles were undetermined. Herein, we exploited spectral editing techniques to identify and quantify the lipid molecules associated with pigments in melanin ghosts. The lipid profiles were remarkably similar in whole C. neoformans cells, grown under either melanizing or nonmelanizing conditions; triglycerides (TGs), sterol esters (SEs), and polyisoprenoids (PPs) were the major constituents. Although no quantitative differences were found between melanized and nonmelanized cells, melanin ghosts were relatively enriched in SEs and PPs. In contrast to lipid structures reported during early stages of fungal growth in nutrient-rich media, variants found herein could be linked to nutrient stress, cell aging, and subsequent production of substances that promote chronic fungal infections. The fact that TGs and SEs are the typical cargo of lipid droplets suggests that these organelles could be connected to C. neoformans melanin synthesis. Moreover, the discovery of PPs is intriguing because dolichol is a well-established constituent of human neuromelanin. The presence of these lipid species even in nonmelanized cells suggests that they could be produced constitutively under stress conditions in anticipation of melanin synthesis. These findings demonstrate that C. neoformans lipids are more varied compositionally and functionally than previously recognized.
Collapse
Affiliation(s)
- Christine Chrissian
- Department of Chemistry and Biochemistry and CUNY Institute for Macromolecular Assemblies, City College of New York, New York, New York, USA; Ph.D. Program in Biochemistry, Graduate Center of the City University of New York, New York, New York, USA
| | - Emma Camacho
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - John E Kelly
- Department of Chemistry and Biochemistry and CUNY Institute for Macromolecular Assemblies, City College of New York, New York, New York, USA
| | - Hsin Wang
- Department of Chemistry and Biochemistry and CUNY Institute for Macromolecular Assemblies, City College of New York, New York, New York, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ruth E Stark
- Department of Chemistry and Biochemistry and CUNY Institute for Macromolecular Assemblies, City College of New York, New York, New York, USA; Ph.D. Program in Biochemistry, Graduate Center of the City University of New York, New York, New York, USA; Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York, USA.
| |
Collapse
|