1
|
Valente MR, Martins Alcântara L, Cintra DS, Mendoza SR, Medeiros EG, Gomes KX, Honorato L, Almeida MDA, Vieira CB, Nosanchuk JD, Sgarbi DBDG, Pinto MR, Nimrichter L, Guimarães AJ. Interactions of the emerging fungus Candida auris with Acanthamoeba castellanii reveal phenotypic changes with direct implications on the response to stress and virulence. Microbiol Spectr 2025; 13:e0174624. [PMID: 39688412 PMCID: PMC11792492 DOI: 10.1128/spectrum.01746-24] [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: 07/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Candida auris is an emerging fungal pathogen notable for its resistance to multiple antifungals and ability to survive in various environments. Understanding the interactions between C. auris and environmental protozoa, such as Acanthamoeba castellanii, could provide insights into fungal adaptability and pathogenicity. Two C. auris isolates (MMC1 and MMC2) were co-cultured with A. castellanii to examine interaction dynamics, survival, stress responses, growth, virulence, biofilm formation, and antifungal susceptibility. The association of C. auris-A. castellanii varied with a multiplicity of infection (MOI), with MMC2 exhibiting higher association rates at increased MOI than MMC1. Both isolates survived distinctly within A. castellanii, as the MMC1 showed an initial decline and subsequent increase in viability, while MMC2 maintained higher viability for up to 24 h, decreasing afterward. Both isolates exhibited accelerated growth when recovered from A. castellanii. The MMC2 isolate displayed increased resistance to oxidative, osmotic, and thermal stresses upon interaction with A. castellanii, whereas MMC1 showed limited changes. Exposure to A. castellanii also influenced the expression of virulence factors differently, with MMC1 increasing phospholipase and peptidase, while MMC2 upregulated phytase, esterase, hemolysin, and siderophores. Upon contact with A. castellanii, MMC2 enhanced biofilm formation, unlike MMC1. Both isolates increased ergosterol upon interactions, enhancing susceptibility to amphotericin B. However, both isolates were more tolerant to itraconazole and caspofungin, particularly MMC2, which showed differential expression of ergosterol biosynthesis enzymes and increased cell wall polysaccharides. This study reveals that interactions with A. castellanii modulate C. auris physiology and virulence, contributing to its environmental adaptability and resistance to antifungals. IMPORTANCE Candida auris has emerged as a critical public health concern due to its resistance to multiple antifungal drugs and ability to survive on surfaces under harsh conditions, mainly due to biofilm formation. The precise origin of this emerging pathogen still awaits elucidation, but interactions with environmental protozoa may have helped C. auris to develop such virulence and resistance traits. In this work, we precisely characterize the interactions of C. auris with the free-living amoeba Acanthamoeba castellanii and how these protozoa may alter the fungal behavior in terms of virulence, thermotolerance, biofilm formation capacity, and drug resistance. It may be essential to understand the various interactions C. auris could perform in the environment, directly impacting the outcome of human infections under the One Health approach.
Collapse
Affiliation(s)
- Michele Ramos Valente
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Lucas Martins Alcântara
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Deborah Santos Cintra
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Susana Ruiz Mendoza
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Elisa Gonçalves Medeiros
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Kamilla Xavier Gomes
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marcos de Abreu Almeida
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Carmen Baur Vieira
- Núcleo de Pesquisa de Virologia, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Joshua Daniel Nosanchuk
- Infectious Diseases, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, USA
| | - Diana Bridon da Graça Sgarbi
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Marcia Ribeiro Pinto
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Programa de Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Rede Micologia RJ – Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology (INCT) in Human Pathogenic Fungi, São Paulo, Brazil
| | - Allan Jefferson Guimarães
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Rede Micologia RJ – Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology (INCT) in Human Pathogenic Fungi, São Paulo, Brazil
| |
Collapse
|
2
|
Medeiros EG, Valente MR, Honorato L, Ferreira MDS, Mendoza SR, Gonçalves DDS, Martins Alcântara L, Gomes KX, Pinto MR, Nakayasu ES, Clair G, da Rocha IFM, dos Reis FCG, Rodrigues ML, Alves LR, Nimrichter L, Casadevall A, Guimarães AJ. Comprehensive characterization of extracellular vesicles produced by environmental (Neff) and clinical (T4) strains of Acanthamoeba castellanii. mSystems 2024; 9:e0122623. [PMID: 38717186 PMCID: PMC11237502 DOI: 10.1128/msystems.01226-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/01/2024] [Indexed: 06/19/2024] Open
Abstract
We conducted a comprehensive comparative analysis of extracellular vesicles (EVs) from two Acanthamoeba castellanii strains, Neff (environmental) and T4 (clinical). Morphological analysis via transmission electron microscopy revealed slightly larger Neff EVs (average = 194.5 nm) compared to more polydisperse T4 EVs (average = 168.4 nm). Nanoparticle tracking analysis (NTA) and dynamic light scattering validated these differences. Proteomic analysis of the EVs identified 1,352 proteins, with 1,107 common, 161 exclusive in Neff, and 84 exclusively in T4 EVs. Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) mapping revealed distinct molecular functions and biological processes and notably, the T4 EVs enrichment in serine proteases, aligned with its pathogenicity. Lipidomic analysis revealed a prevalence of unsaturated lipid species in Neff EVs, particularly triacylglycerols, phosphatidylethanolamines (PEs), and phosphatidylserine, while T4 EVs were enriched in diacylglycerols and diacylglyceryl trimethylhomoserine, phosphatidylcholine and less unsaturated PEs, suggesting differences in lipid metabolism and membrane permeability. Metabolomic analysis indicated Neff EVs enrichment in glycerolipid metabolism, glycolysis, and nucleotide synthesis, while T4 EVs, methionine metabolism. Furthermore, RNA-seq of EVs revealed differential transcript between the strains, with Neff EVs enriched in transcripts related to gluconeogenesis and translation, suggesting gene regulation and metabolic shift, while in the T4 EVs transcripts were associated with signal transduction and protein kinase activity, indicating rapid responses to environmental changes. In this novel study, data integration highlighted the differences in enzyme profiles, metabolic processes, and potential origins of EVs in the two strains shedding light on the diversity and complexity of A. castellanii EVs and having implications for understanding host-pathogen interactions and developing targeted interventions for Acanthamoeba-related diseases.IMPORTANCEA comprehensive and fully comparative analysis of extracellular vesicles (EVs) from two Acanthamoeba castellanii strains of distinct virulence, a Neff (environmental) and T4 (clinical), revealed striking differences in their morphology and protein, lipid, metabolites, and transcripts levels. Data integration highlighted the differences in enzyme profiles, metabolic processes, and potential distinct origin of EVs from both strains, shedding light on the diversity and complexity of A. castellanii EVs, with direct implications for understanding host-pathogen interactions, disease mechanisms, and developing new therapies for the clinical intervention of Acanthamoeba-related diseases.
Collapse
Affiliation(s)
- Elisa Gonçalves Medeiros
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Michele Ramos Valente
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Leandro Honorato
- Departamento de Microbiologia Geral, Laboratório de Glicobiologia de Eucariotos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marina da Silva Ferreira
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Susana Ruiz Mendoza
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Diego de Souza Gonçalves
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lucas Martins Alcântara
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Kamilla Xavier Gomes
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Departamento de Microbiologia Geral, Laboratório de Glicobiologia de Eucariotos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marcia Ribeiro Pinto
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Ernesto S. Nakayasu
- Biological Science Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | | | - Flavia C. G. dos Reis
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Fiocruz, Curitiba, Paraná, Brazil
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS), Fiocruz, Rio de Janeiro, Brazil
| | - Marcio L. Rodrigues
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Fiocruz, Curitiba, Paraná, Brazil
- Instituto de Microbiologia Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
| | - Lysangela R. Alves
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Fiocruz, Curitiba, Paraná, Brazil
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Laboratório de Glicobiologia de Eucariotos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Rede Micologia RJ–Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Rede Micologia RJ–Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Chen L, Han W, Jing W, Feng M, Zhou Q, Cheng X. Novel anti- Acanthamoeba effects elicited by a repurposed poly (ADP-ribose) polymerase inhibitor AZ9482. Front Cell Infect Microbiol 2024; 14:1414135. [PMID: 38863831 PMCID: PMC11165085 DOI: 10.3389/fcimb.2024.1414135] [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: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
Introduction Acanthamoeba infection is a serious public health concern, necessitating the development of effective and safe anti-Acanthamoeba chemotherapies. Poly (ADP-ribose) polymerases (PARPs) govern a colossal amount of biological processes, such as DNA damage repair, protein degradation and apoptosis. Multiple PARP-targeted compounds have been approved for cancer treatment. However, repurposing of PARP inhibitors to treat Acanthamoeba is poorly understood. Methods In the present study, we attempted to fill these knowledge gaps by performing anti-Acanthamoeba efficacy assays, cell biology experiments, bioinformatics, and transcriptomic analyses. Results Using a homology model of Acanthamoeba poly (ADP-ribose) polymerases (PARPs), molecular docking of approved drugs revealed three potential inhibitory compounds: olaparib, venadaparib and AZ9482. In particular, venadaparib exhibited superior docking scores (-13.71) and favorable predicted binding free energy (-89.28 kcal/mol), followed by AZ9482, which showed a docking score of -13.20 and a binding free energy of -92.13 kcal/mol. Notably, the positively charged cyclopropylamine in venadaparib established a salt bridge (through E535) and a hydrogen bond (via N531) within the binding pocket. For comparison, AZ9482 was well stacked by the surrounding aromatic residues including H625, Y652, Y659 and Y670. In an assessment of trophozoites viability, AZ9482 exhibited a dose-and time-dependent anti-trophozoite effect by suppressing Acanthamoeba PARP activity, unlike olaparib and venadaparib. An Annexin V-fluorescein isothiocyanate/propidium iodide apoptosis assay revealed AZ9482 induced trophozoite necrotic cell death rather than apoptosis. Transcriptomics analyses conducted on Acanthamoeba trophozoites treated with AZ9482 demonstrated an atlas of differentially regulated proteins and genes, and found that AZ9482 rapidly upregulates a multitude of DNA damage repair pathways in trophozoites, and intriguingly downregulates several virulent genes. Analyzing gene expression related to DNA damage repair pathway and the rate of apurinic/apyrimidinic (AP) sites indicated DNA damage efficacy and repair modulation in Acanthamoeba trophozoites following AZ9482 treatment. Discussion Collectively, these findings highlight AZ9482, as a structurally unique PARP inhibitor, provides a promising prototype for advancing anti-Acanthamoeba drug research.
Collapse
Affiliation(s)
- Lijun Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Han
- Research Center for Intelligent Computing Platforms, Zhejiang Lab, Hangzhou, China
| | - Wenwen Jing
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meng Feng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qingtong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Hargrove T, Lamb DC, Wawrzak Z, Hull M, Kelly SL, Guengerich FP, Lepesheva GI. Identification of Potent and Selective Inhibitors of Acanthamoeba: Structural Insights into Sterol 14α-Demethylase as a Key Drug Target. J Med Chem 2024; 67:7443-7457. [PMID: 38683753 PMCID: PMC11089504 DOI: 10.1021/acs.jmedchem.4c00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Acanthamoeba are free-living pathogenic protozoa that cause blinding keratitis, disseminated infection, and granulomatous amebic encephalitis, which is generally fatal. The development of efficient and safe drugs is a critical unmet need. Acanthamoeba sterol 14α-demethylase (CYP51) is an essential enzyme of the sterol biosynthetic pathway. Repurposing antifungal azoles for amoebic infections has been reported, but their inhibitory effects on Acanthamoeba CYP51 enzymatic activity have not been studied. Here, we report catalytic properties, inhibition, and structural characterization of CYP51 from Acanthamoeba castellanii. The enzyme displays a 100-fold substrate preference for obtusifoliol over lanosterol, supporting the plant-like cycloartenol-based pathway in the pathogen. The strongest inhibition was observed with voriconazole (1 h IC50 0.45 μM), VT1598 (0.25 μM), and VT1161 (0.20 μM). The crystal structures of A. castellanii CYP51 with bound VT1161 (2.24 Å) and without an inhibitor (1.95 Å), presented here, can be used in the development of azole-based scaffolds to achieve optimal amoebicidal effectiveness.
Collapse
Affiliation(s)
- Tatiana
Y. Hargrove
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - David C. Lamb
- Faculty
of Medicine, Health and Life Science, Swansea
University, Swansea SA2 8PP, U.K.
| | - Zdzislaw Wawrzak
- Synchrotron
Research Center, Life Science Collaborative Access Team, Northwestern University, Argonne, Illinois 60439, United States
| | - Marcus Hull
- Faculty
of Medicine, Health and Life Science, Swansea
University, Swansea SA2 8PP, U.K.
| | - Steven L. Kelly
- Faculty
of Medicine, Health and Life Science, Swansea
University, Swansea SA2 8PP, U.K.
| | - F. Peter Guengerich
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Galina I. Lepesheva
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Chemical Biology, Nashville, Tennessee 37232, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
5
|
Ferreira MDS, Gonçalves DDS, Mendoza SR, de Oliveira GA, Pontes B, la Noval CRD, Honorato L, Ramos LFC, Nogueira FCS, Domont GB, Casadevall A, Nimrichter L, Peralta JM, Guimaraes AJ. β-1,3-Glucan recognition by Acanthamoeba castellanii as a putative mechanism of amoeba-fungal interactions. Appl Environ Microbiol 2024; 90:e0173623. [PMID: 38259076 PMCID: PMC10880599 DOI: 10.1128/aem.01736-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
In this study, we conducted an in-depth analysis to characterize potential Acanthamoeba castellanii (Ac) proteins capable of recognizing fungal β-1,3-glucans. Ac specifically anchors curdlan or laminarin, indicating the presence of surface β-1,3-glucan-binding molecules. Using optical tweezers, strong adhesion of laminarin- or curdlan-coated beads to Ac was observed, highlighting their adhesive properties compared to controls (characteristic time τ of 46.9 and 43.9 s, respectively). Furthermore, Histoplasma capsulatum (Hc) G217B, possessing a β-1,3-glucan outer layer, showed significant adhesion to Ac compared to a Hc G186 strain with an α-1,3-glucan outer layer (τ of 5.3 s vs τ 83.6 s). The addition of soluble β-1,3-glucan substantially inhibited this adhesion, indicating the involvement of β-1,3-glucan recognition. Biotinylated β-1,3-glucan-binding proteins from Ac exhibited higher binding to Hc G217B, suggesting distinct recognition mechanisms for laminarin and curdlan, akin to macrophages. These observations hinted at the β-1,3-glucan recognition pathway's role in fungal entrance and survival within phagocytes, supported by decreased fungal viability upon laminarin or curdlan addition in both phagocytes. Proteomic analysis identified several Ac proteins capable of binding β-1,3-glucans, including those with lectin/glucanase superfamily domains, carbohydrate-binding domains, and glycosyl transferase and glycosyl hydrolase domains. Notably, some identified proteins were overexpressed upon curdlan/laminarin challenge and also demonstrated high affinity to β-1,3-glucans. These findings underscore the complexity of binding via β-1,3-glucan and suggest the existence of alternative fungal recognition pathways in Ac.IMPORTANCEAcanthamoeba castellanii (Ac) and macrophages both exhibit the remarkable ability to phagocytose various extracellular microorganisms in their respective environments. While substantial knowledge exists on this phenomenon for macrophages, the understanding of Ac's phagocytic mechanisms remains elusive. Recently, our group identified mannose-binding receptors on the surface of Ac that exhibit the capacity to bind/recognize fungi. However, the process was not entirely inhibited by soluble mannose, suggesting the possibility of other interactions. Herein, we describe the mechanism of β-1,3-glucan binding by A. castellanii and its role in fungal phagocytosis and survival within trophozoites, also using macrophages as a model for comparison, as they possess a well-established mechanism involving the Dectin-1 receptor for β-1,3-glucan recognition. These shed light on a potential parallel evolution of pathways involved in the recognition of fungal surface polysaccharides.
Collapse
Affiliation(s)
- Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Diego de Souza Gonçalves
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Susana Ruiz Mendoza
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Gabriel Afonso de Oliveira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas e Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Claudia Rodríguez-de la Noval
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Luis Felipe Costa Ramos
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Fábio C. S. Nogueira
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Gilberto B. Domont
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Leonardo Nimrichter
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
- Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Niterói, Rio de Janeiro, Brazil
| | - Jose Mauro Peralta
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Allan J. Guimaraes
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
- Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Niterói, Rio de Janeiro, Brazil
- Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Carvalho JHDS, Nascimento JKC, Silva KGV, Silveira Neto S, Macedo ATD, Lima França H, Ferreira LDR, Silva RDS, Sa JC, Ramos DG, Marques DDAV, Furst C, Santos DA, Santos JRA, Holanda RA. Yeast-amoeba interaction influences murine cryptococcosis. Microbes Infect 2023; 25:105153. [PMID: 37244475 DOI: 10.1016/j.micinf.2023.105153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 05/21/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
The virulence of Cryptococcus spp. is modulated in the natural environment through interaction with abiotic and biotic factors, and this can occasionally have implications for the progression of cryptococcosis in mammals. Hence, we evaluated whether the prior interaction of highly virulent Cryptococcus gattii strain R265 with Acanthamoeba castellanii influenced the progression of cryptococcosis. The influence of the capsule on endocytosis was evaluated using amoeba and yeast morphometrics. Mice were intratracheally infected with yeast re-isolated from the amoeba (Interaction), yeast without prior contact with the amoeba (Non-Interaction), or sterile phosphate-buffered saline (SHAM). Morbidity signs and symptoms were monitored during the survival curve, while cytokine and fungal burden measurements and histopathological analysis were performed on the 10th day post infection. Morbidity and mortality parameters in experimental cryptococcosis were influenced by the prior interaction of yeast with amoeba, which led to phenotypic changes in the cryptococcal cells, polysaccharide secretion, and their tolerance to oxidative stress. Our results suggest that a prior yeast-amoeba interaction modulates yeast virulence, which is associated with a greater tolerance to oxidative stress related to the exo-polysaccharide content and influences the progression of cryptococcal infection.
Collapse
Affiliation(s)
| | | | | | - Sebastiao Silveira Neto
- Laboratório de Biologia Molecular de Microrganismos Patogênicos, Universidade CEUMA, São Luís, Maranhão, Brazil
| | | | - Hermeson Lima França
- Laboratório de Biologia Molecular de Microrganismos Patogênicos, Universidade CEUMA, São Luís, Maranhão, Brazil
| | - Larissa Dos Reis Ferreira
- Laboratório de Biologia Molecular de Microrganismos Patogênicos, Universidade CEUMA, São Luís, Maranhão, Brazil
| | - Rayssa de Sousa Silva
- Laboratório de Biologia Molecular de Microrganismos Patogênicos, Universidade CEUMA, São Luís, Maranhão, Brazil
| | - Joicy Cortez Sa
- Laboratório de Imunologia, Universidade CEUMA, São Luís, Maranhão, Brazil
| | - Diego Gomes Ramos
- Laboratório Integrado de Biotecnologia Aplicada, Universidade de Pernambuco, Recife, Pernambuco, Brazil
| | | | - Cinthia Furst
- Departamento de Patologia, Universidade Federal do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Daniel Assis Santos
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Rodrigo Assuncao Holanda
- Laboratório de Biologia Molecular de Microrganismos Patogênicos, Universidade CEUMA, São Luís, Maranhão, Brazil.
| |
Collapse
|
7
|
Tsai CM, Chen CH, Cheng WH, Stelma FF, Li SC, Lin WC. Homeostasis of cellular amino acids in Acanthamoeba castellanii exposed to different media under amoeba-bacteria coculture conditions. BMC Microbiol 2023; 23:198. [PMID: 37495951 PMCID: PMC10373360 DOI: 10.1186/s12866-023-02942-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Acanthamoeba castellanii is a free-living protist that feeds on diverse bacteria. A. castellanii has frequently been utilized in studies on microbial interactions. Grazing bacteria also exhibit diverse effects on the physiological characteristics of amoebae, such as their growth, encystation, and cytotoxicity. Since the composition of amoebae amino acids is closely related to cellular activities, it can indicate the overall responses of A. castellanii to various stimuli. METHOD A. castellanii was exposed to different culture conditions in low-nutrient medium with heat-killed DH5α to clarify their effects. A targeted metabolomic technique was utilized to evaluate the concentration of cellular amino acids. The amino acid composition and pathways were analyzed by two web-based tools: MetaboAnalyst and Pathview. Then, long-term exposure to A. castellanii was investigated through in silico and in vitro methods to elucidate the homeostasis of amino acids and the growth of A. castellanii. RESULTS Under short-term exposure, all kinds of amino acids were enriched in all exposed groups. In contrast to the presence of heat-killed bacteria, the medium exhibited obvious effects on the amino acid composition of A. castellanii. After long-term exposure, the amino acid composition was more similar to that of the control group. A. castellanii may achieve amino acid homeostasis through pathways related to alanine, aspartate, citrulline, and serine. DISCUSSION Under short-term exposure, compared to the presence of bacteria, the type of medium exerted a more powerful effect on the amino acid composition of the amoeba. Previous studies focused on the interaction of the amoeba and bacteria with effective secretion systems and effectors. This may have caused the effects of low-nutrient environments to be overlooked. CONCLUSION When A. castellanii was stimulated in the coculture system through various methods, such as the presence of bacteria and a low-nutrient environment, it accumulated intracellular amino acids within a short period. However, different stimulations correspond to different amino acid compositions. After long-term exposure, A. castellanii achieved an amino acid equilibrium by downregulating the biosynthesis of several amino acids.
Collapse
Affiliation(s)
- Chih-Ming Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Hsien Chen
- Department of Parasitology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Hung Cheng
- Department of Parasitology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Foekje F. Stelma
- Department of Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Sung-Chou Li
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Nursing, Meiho University, Pingtung, Taiwan
| | - Wei-Chen Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Parasitology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
8
|
Wang Y, Jiang L, Zhao Y, Ju X, Wang L, Jin L, Fine RD, Li M. Biological characteristics and pathogenicity of Acanthamoeba. Front Microbiol 2023; 14:1147077. [PMID: 37089530 PMCID: PMC10113681 DOI: 10.3389/fmicb.2023.1147077] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
Acanthamoeba is an opportunistic protozoa, which exists widely in nature and is mainly distributed in soil and water. Acanthamoeba usually exists in two forms, trophozoites and cysts. The trophozoite stage is one of growth and reproduction while the cyst stage is characterized by cellular quiescence, commonly resulting in human infection, and the lack of effective monotherapy after initial infection leads to chronic disease. Acanthamoeba can infect several human body tissues such as the skin, cornea, conjunctiva, respiratory tract, and reproductive tract, especially when the tissue barriers are damaged. Furthermore, serious infections can cause Acanthamoeba keratitis, granulomatous amoebic encephalitis, skin, and lung infections. With an increasing number of Acanthamoeba infections in recent years, the pathogenicity of Acanthamoeba is becoming more relevant to mainstream clinical care. This review article will describe the etiological characteristics of Acanthamoeba infection in detail from the aspects of biological characteristic, classification, disease, and pathogenic mechanism in order to provide scientific basis for the diagnosis, treatment, and prevention of Acanthamoeba infection.
Collapse
Affiliation(s)
- Yuehua Wang
- College of Laboratory Medicine, Jilin Medical University, Jilin City, China
| | - Linzhe Jiang
- General Surgery, Jilin People’s Hospital, Jilin City, China
| | - Yitong Zhao
- College of Laboratory Medicine, Jilin Medical University, Jilin City, China
| | - Xiaohong Ju
- College of Laboratory Medicine, Jilin Medical University, Jilin City, China
| | - Le Wang
- Department of Laboratory Medicine, Jilin Hospital of Integrated Chinese and Western Medicine, Jilin City, China
| | - Liang Jin
- Department of Laboratory Medicine, Jilin Hospital of Integrated Chinese and Western Medicine, Jilin City, China
| | - Ryan D. Fine
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York City, NY, United States
| | - Mingguang Li
- College of Laboratory Medicine, Jilin Medical University, Jilin City, China
- *Correspondence: Mingguang Li,
| |
Collapse
|
9
|
Rayamajhee B, Willcox MDP, Henriquez FL, Petsoglou C, Subedi D, Carnt N. Acanthamoeba, an environmental phagocyte enhancing survival and transmission of human pathogens. Trends Parasitol 2022; 38:975-990. [PMID: 36109313 DOI: 10.1016/j.pt.2022.08.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/13/2023]
Abstract
The opportunistic protist Acanthamoeba, which interacts with other microbes such as bacteria, fungi, and viruses, shows significant similarity in cellular and functional aspects to human macrophages. Intracellular survival of microbes in this microbivorous amoebal host may be a crucial step for initiation of infection in higher eukaryotic cells. Therefore, Acanthamoeba-microbe adaptations are considered an evolutionary model of macrophage-pathogen interactions. This paper reviews Acanthamoeba as an emerging human pathogen and different ecological interactions between Acanthamoeba and microbes that may serve as environmental training grounds and a genetic melting pot for the evolution, persistence, and transmission of potential human pathogens.
Collapse
Affiliation(s)
- Binod Rayamajhee
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia.
| | - Mark D P Willcox
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia.
| | - Fiona L Henriquez
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Blantyre, South Lanarkshire, G72 0LH, UK
| | - Constantinos Petsoglou
- Sydney and Sydney Eye Hospital, Southeastern Sydney Local Health District, Sydney, Australia; Save Sight Institute, University of Sydney, Sydney, Australia
| | - Dinesh Subedi
- School of Biological Sciences, Monash University, Clayton, Australia
| | - Nicole Carnt
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia
| |
Collapse
|
10
|
Corsaro D. Acanthamoeba Mannose and Laminin Binding Proteins Variation across Species and Genotypes. Microorganisms 2022; 10:2162. [PMID: 36363753 PMCID: PMC9692275 DOI: 10.3390/microorganisms10112162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/26/2022] [Indexed: 11/26/2023] Open
Abstract
Acanthamoeba is a ubiquitous free-living amoeba capable of being an opportunistic pathogen in humans and animals. A critical step in infection is the adhesion of the amoeba to host cells and tissues, and two major parasite adhesins, mannose-binding protein (MBP) and laminin-binding protein (LBP), are known to recognize the cell surface glycoproteins and those of the extracellular matrix, respectively. In this study, the available genomes of Acanthamoeba were analysed to recover the sequences of MBP and LBP using previously published genetic data. Genes for both proteins were successfully obtained from strains belonging to various genotypes (T4A, T4D, T4G, T4F, T2, T5, T10, T22, T7 and T18), resulting in a single gene for LBP but identifying two types of MBP, MBP1 and MBP2. Phylogenetic analysis based on deduced amino acid sequences shows that both MBP and LBP have a branching pattern that is consistent with that based on 18S rDNA, indicating that changes in both proteins occurred during diversification of Acanthamoeba lines. Notably, all MBPs possess a conserved motif, shared with some bacterial C-type lectins, which could be the recognition site for mannose binding.
Collapse
|
11
|
Pruksaphon K, Nosanchuk JD, Thammasit P, Pongpom M, Youngchim S. Interaction of Talaromyces marneffei with free living soil amoeba as a model of fungal pathogenesis. Front Cell Infect Microbiol 2022; 12:1023067. [PMID: 36262181 PMCID: PMC9574045 DOI: 10.3389/fcimb.2022.1023067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Talaromyces (Penicillium) marneffei is an important dimorphic mycosis endemic in Southeast Asia and Southern China, but the origin and maintenance of virulence traits in this organism remains obscure. Several pathogenic fungi, including Cryptococcus neoformans, Aspergillus fumigatus, Blastomyces dermatitidis, Sporothrix schenckii, Histoplasma capsulatum and Paracoccidioides spp. interact with free living soil amoebae and data suggests that fungal pathogenic strategies may emerge from environmental interactions of these fungi with ubiquitous phagocytic microorganisms. In this study, we examined the interactions of T. marneffei with the soil amoeba Acanthamoeba castellanii. T. marneffei was rapidly ingested by A. castellanii and phagocytosis of fungal cells resulted in amoeba death after 24 h of contact. Co-culture also resulted in a rapid transition for conidia to the fission-yeast form. In addition, well-established virulence factors such as melanin and a yeast specific mannoprotein of T. marneffei were expressed during interaction with A. castellanii at 37°C. Our findings support the assumption that soil amoebae environmental predators play a role in the selection and maintenance of particular features in T. marneffei that impart virulence to this clinically important dimorphic fungus in mammalian hosts.
Collapse
Affiliation(s)
- Kritsada Pruksaphon
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Joshua D. Nosanchuk
- Departments of Microbiology and Immunology and Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Patcharin Thammasit
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Monsicha Pongpom
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirida Youngchim
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- *Correspondence: Sirida Youngchim,
| |
Collapse
|
12
|
Fukunaga T, Ohashi T, Tanaka Y, Yoshimatsu T, Higuchi Y, Maekawa H, Takegawa K. Galactosylation of cell-surface glycoprotein required for hyphal growth and cell wall integrity in Schizosaccharomyces japonicus. J Biosci Bioeng 2022; 134:384-392. [DOI: 10.1016/j.jbiosc.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/01/2022]
|
13
|
Mendoza SR, Liedke SC, de La Noval CR, da Silva Ferreira M, Gomes KX, Honorato L, Nimrichter L, Peralta JM, Guimarães AJ. In vitro and in vivo efficacies of Dectin-1-Fc(IgG)(s) fusion proteins against invasive fungal infections. Med Mycol 2022; 60:6648754. [PMID: 35867978 DOI: 10.1093/mmy/myac050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/22/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Fungal infections have increased in the last years, particularly associated to an increment in the number of immunocompromised individuals and the emergence of known or new resistant species, despite the difficulties in the often time-consuming diagnosis. The controversial efficacy of the currently available strategies for their clinical management, apart from their high toxicity and severe side effects, have renewed the interest in the research and development of new broad antifungal alternatives. These encompass vaccines and passive immunization strategies with monoclonal antibodies (mAbs), recognizing ubiquitous fungal targets, such as fungal cell wall β-1,3-glucan polysaccharides, which could be used in early therapeutic intervention without the need for the diagnosis at species-level. As additional alternatives, based on the Dectin-1 great affinity to β-1,3-glucan, our group developed broad antibody-like Dectin1-Fc(IgG)(s) from distinct subclasses (IgG2a and IgG2b) and compared their antifungal in vitro and passive immunizations in vivo performances. Dectin1-Fc(IgG2a) and Dectin1-Fc(IgG2b) demonstrated high affinity to laminarin and the fungal cell wall by ELISA, flow cytometry and microscopy. Both Dectin-1-Fc(IgG)(s) inhibited H. capsulatum and C. neoformans growth in a dose-dependent fashion. For C. albicans, such inhibitory effect was observed with concentrations as low as 0.098 and 0.049 µg/mL, respectively, which correlated with the impairment of the kinetics and lengths of germ tubes in comparison to controls. Previous opsonization with Dectin-1-Fc(IgG)(s) enhanced considerably the macrophage antifungal effector functions, increasing the fungi macrophages-interactions and significantly reducing the intraphagosome fungal survival, as lower CFUs were observed. The administration of both Dectin1-Fc(IgG)(s) reduced the fungal burden and mortality in murine histoplasmosis and candidiasis models, in accordance with previous evaluations in aspergillosis model. These results altogether strongly suggested that therapeutic interventions with Dectin-1-Fc(IgG)(s) fusion proteins could directly impact the innate immunity and disease outcome in favor of the host, by direct neutralization, opsonization, phagocytosis, and fungal elimination, providing interesting information on the potential of these new strategies for the control of invasive fungal infections.
Collapse
Affiliation(s)
- S R Mendoza
- Laboratório de Bioquímica e Imunologia das Micoses, Instituto Biomédico, Fluminense Federal University, Brazil.,Programa de Pós-Graduação em Imunologia e Inflamação, Federal University of Rio de Janeiro, Brazil
| | - S C Liedke
- Laboratório de Diagnóstico Imunológico e Molecular de Doenças Infecciosas e Parasitárias, Federal University of Rio de Janeiro, Brazil
| | - C R de La Noval
- Laboratório de Bioquímica e Imunologia das Micoses, Instituto Biomédico, Fluminense Federal University, Brazil.,Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Paulo de Góes, Federal University of Rio de Janeiro, Brazil
| | - M da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Instituto Biomédico, Fluminense Federal University, Brazil.,Programa de Pós-Graduação em Imunologia e Inflamação, Federal University of Rio de Janeiro, Brazil
| | - K X Gomes
- Laboratório de Bioquímica e Imunologia das Micoses, Instituto Biomédico, Fluminense Federal University, Brazil.,Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), RJ, Brazil
| | - L Honorato
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Paulo de Góes, Federal University of Rio de Janeiro, Brazil
| | - L Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Paulo de Góes, Federal University of Rio de Janeiro, Brazil.,Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), RJ, Brazil
| | - J M Peralta
- Laboratório de Diagnóstico Imunológico e Molecular de Doenças Infecciosas e Parasitárias, Federal University of Rio de Janeiro, Brazil
| | - A J Guimarães
- Laboratório de Bioquímica e Imunologia das Micoses, Instituto Biomédico, Fluminense Federal University, Brazil.,Programa de Pós-Graduação em Imunologia e Inflamação, Federal University of Rio de Janeiro, Brazil.,Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), RJ, Brazil.,Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Fluminense Federal University, Brazil
| |
Collapse
|
14
|
Bellini NK, Thiemann OH, Reyes-Batlle M, Lorenzo-Morales J, Costa AO. A history of over 40 years of potentially pathogenic free-living amoeba studies in Brazil - a systematic review. Mem Inst Oswaldo Cruz 2022; 117:e210373. [PMID: 35792751 PMCID: PMC9252135 DOI: 10.1590/0074-02760210373] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/28/2022] [Indexed: 12/17/2022] Open
Abstract
Free-living amoeba (FLA) group includes the potentially pathogenic genera Acanthamoeba, Naegleria, Balamuthia, Sappinia, and Vermamoeba, causative agents of human infections (encephalitis, keratitis, and disseminated diseases). In Brazil, the first report on pathogenic FLA was published in the 70s and showed meningoencephalitis caused by Naegleria spp. FLA studies are emerging, but no literature review is available to investigate this trend in Brazil critically. Thus, the present work aims to integrate and discuss these data. Scopus, PubMed, and Web of Science were searched, retrieving studies from 1974 to 2020. The screening process resulted in 178 papers, which were clustered into core and auxiliary classes and sorted into five categories: wet-bench studies, dry-bench studies, clinical reports, environmental identifications, and literature reviews. The papers dating from the last ten years account for 75% (134/178) of the total publications, indicating the FLA topic has gained Brazilian interest. Moreover, 81% (144/178) address Acanthamoeba-related matter, revealing this genus as the most prevalent in all categories. Brazil’s Southeast, South, and Midwest geographic regions accounted for 96% (171/178) of the publications studied in the present work. To the best of our knowledge, this review is the pioneer in summarising the FLA research history in Brazil.
Collapse
Affiliation(s)
- Natália Karla Bellini
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brasil
| | - Otavio Henrique Thiemann
- Universidade de São Paulo, Instituto de Física de São Carlos, São Carlos, SP, Brasil.,Universidade Federal de São Carlos, Departamento de Genética e Evolução, São Carlos, SP, Brasil
| | - María Reyes-Batlle
- Universidad de La Laguna, Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Departamento de Obstetricia, Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Red de Investigación Cooperativa en Enfermedades Tropicales, Tenerife, Islas Canarias, Spain
| | - Jacob Lorenzo-Morales
- Universidad de La Laguna, Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Departamento de Obstetricia, Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Red de Investigación Cooperativa en Enfermedades Tropicales, Tenerife, Islas Canarias, Spain.,Instituto de Salud Carlos III, Consorcio Centro de Investigación Biomédica en Red MP de Enfermedades Infecciosas, Madrid, Spain
| | - Adriana Oliveira Costa
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brasil
| |
Collapse
|
15
|
Ferreira MDS, Mendoza SR, Gonçalves DDS, Rodríguez-de la Noval C, Honorato L, Nimrichter L, Ramos LFC, Nogueira FCS, Domont GB, Peralta JM, Guimarães AJ. Recognition of Cell Wall Mannosylated Components as a Conserved Feature for Fungal Entrance, Adaptation and Survival Within Trophozoites of Acanthamoeba castellanii and Murine Macrophages. Front Cell Infect Microbiol 2022; 12:858979. [PMID: 35711659 PMCID: PMC9194641 DOI: 10.3389/fcimb.2022.858979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023] Open
Abstract
Acanthamoeba castellanii (Ac) is a species of free-living amoebae (FLAs) that has been widely applied as a model for the study of host-parasite interactions and characterization of environmental symbionts. The sharing of niches between Ac and potential pathogens, such as fungi, favors associations between these organisms. Through predatory behavior, Ac enhances fungal survival, dissemination, and virulence in their intracellular milieu, training these pathogens and granting subsequent success in events of infections to more evolved hosts. In recent studies, our group characterized the amoeboid mannose binding proteins (MBPs) as one of the main fungal recognition pathways. Similarly, mannose-binding lectins play a key role in activating antifungal responses by immune cells. Even in the face of similarities, the distinct impacts and degrees of affinity of fungal recognition for mannose receptors in amoeboid and animal hosts are poorly understood. In this work, we have identified high-affinity ligands for mannosylated fungal cell wall residues expressed on the surface of amoebas and macrophages and determined the relative importance of these pathways in the antifungal responses comparing both phagocytic models. Mannose-purified surface proteins (MPPs) from both phagocytes showed binding to isolated mannose/mannans and mannosylated fungal cell wall targets. Although macrophage MPPs had more intense binding when compared to the amoeba receptors, the inhibition of this pathway affects fungal internalization and survival in both phagocytes. Mass spectrometry identified several MPPs in both models, and in silico alignment showed highly conserved regions between spotted amoeboid receptors (MBP and MBP1) and immune receptors (Mrc1 and Mrc2) and potential molecular mimicry, pointing to a possible convergent evolution of pathogen recognition mechanisms.
Collapse
Affiliation(s)
- Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
- Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Ruiz Mendoza
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
- Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego de Souza Gonçalves
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
- Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Rodríguez-de la Noval
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| | - Leandro Honorato
- Programa de Pós-Graduação em Ciências (Microbiologia), Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia RJ - FAPERJ, Rio de Janeiro, Brazil
| | - Luís Felipe Costa Ramos
- Laboratório de Química de Proteínas, Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio C. S. Nogueira
- Laboratório de Química de Proteínas, Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto B. Domont
- Laboratório de Química de Proteínas, Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Mauro Peralta
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Allan J. Guimarães
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
- Pós-Graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia RJ - FAPERJ, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
16
|
War of the microbial world: Acanthamoeba spp. interactions with microorganisms. Folia Microbiol (Praha) 2021; 66:689-699. [PMID: 34145552 PMCID: PMC8212903 DOI: 10.1007/s12223-021-00889-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/09/2021] [Indexed: 12/15/2022]
Abstract
Acanthamoeba is known to interact with a plethora of microorganisms such as bacteria, fungi and viruses. In these interactions, the amoebae can be predatory in nature, transmission vehicle or an incubator. Amoebae consume microorganisms, especially bacteria, as food source to fulfil their nutritional needs by taking up bacteria through phagocytosis and lysing them in phagolysosomes and hence play an eminent role in the regulation of bacterial density in the nature and accountable for eradication of around 60% of the bacterial population in the environment. Acanthamoeba can also act as a “Trojan horse” for microbial transmission in the environment. Additionally, Acanthamoeba may serve as an incubator-like reservoir for microorganisms, including those that are pathogenic to humans, where the microorganisms use amoebae’s defences to resist harsh environment and evade host defences and drugs, whilst growing in numbers inside the amoebae. Furthermore, amoebae can also be used as a “genetic melting pot” where exchange of genes as well as adaptation of microorganisms, leading to higher pathogenicity, may arise. Here, we describe bacteria, fungi and viruses that are known to interact with Acanthamoeba spp.
Collapse
|
17
|
Fukunaga T, Tanaka N, Furumoto T, Nakakita S, Ohashi T, Higuchi Y, Maekawa H, Takegawa K. Substrate specificities of α1,2- and α1,3-galactosyltransferases and characterization of Gmh1p and Otg1p in Schizosaccharomyces pombe. Glycobiology 2021; 31:1037-1045. [PMID: 33909078 DOI: 10.1093/glycob/cwab028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/26/2022] Open
Abstract
In the fission yeast Schizosaccharomyces pombe, α1,2- and α1,3-linked D-galactose (Gal) residues are transferred to N- and O-linked oligosaccharides of glycoproteins by galactosyltransferases. Although the galactomannans are important for cell-cell communication in S. pombe (e.g., in non-sexual aggregation), the mechanisms underlying galactosylation in cells remain unclear. S. pombe has 10 galactosyltransferase-related genes: seven belonging to glycosyltransferase (GT) family 34 and three belonging GT family 8. Disruption of all 10 α-galactosyltransferases (strain Δ10GalT) has been shown to result in a complete lack of α-Gal residues. Here, we have investigated the function and substrate specificities of galactosyltransferases in S pombe by using strains expressing single α-galactosyltransferases in the Δ10GalT background. High-performance liquid chromatography (HPLC) analysis of pyridylaminated O-linked oligosaccharides showed that two GT family 34 α1,2-galactosyltransferases (Gma12p and Gmh6p) and two GT family 8 α1,3-galactosyltransferases (Otg2p and Otg3p) are involved in galactosylation of O-linked oligosaccharide. Moreover, 1H-NMR of N-glycans revealed that three GT family 34 α1,2-galactosyltransferases (Gmh1p, Gmh2p, and Gmh3p) are required for galactosylation of N-linked oligosaccharides. Furthermore, HPLC and lectin-blot analysis revealed that Otg1p showed α1,3-galactosyltransferase activity under conditions of co-expression with Gmh6p, indicating that α-1,2-linked galactose is required for the galactosylation activity of Otg1p in S. pombe. In conclusion, eight galactosyltransferases have been shown to have activity in S. pombe with different substrate specificities. These findings will be useful for genetically tailoring the galactosylation of both N- and O- glycans in fission yeast.
Collapse
Affiliation(s)
- Takamasa Fukunaga
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Naotaka Tanaka
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Toshio Furumoto
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Shinichi Nakakita
- Department of Endocrinology; Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Kagawa, Japan; and Life Science Research Center, Kagawa University, Kagawa, Japan
| | - Takao Ohashi
- Faculty of Science and Engineering Department of Life Science, Setsunan University, Osaka, Japan
| | - Yujiro Higuchi
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Hiromi Maekawa
- Centre for Promotion of International Education and Research, Faculty of Agriculture, Kyushu university, Fukuoka, Japan
| | - Kaoru Takegawa
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
18
|
Fu MS, Liporagi-Lopes LC, Dos Santos SR, Tenor JL, Perfect JR, Cuomo CA, Casadevall A. Amoeba Predation of Cryptococcus neoformans Results in Pleiotropic Changes to Traits Associated with Virulence. mBio 2021; 12:e00567-21. [PMID: 33906924 PMCID: PMC8092252 DOI: 10.1128/mbio.00567-21] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/30/2021] [Indexed: 11/20/2022] Open
Abstract
Amoeboid predators, such as amoebae, are proposed to select for survival traits in soil microbes such as Cryptococcus neoformans; these traits can also function in animal virulence by defeating phagocytic immune cells, such as macrophages. Consistent with this notion, incubation of various fungal species with amoebae enhanced their virulence, but the mechanisms involved are unknown. In this study, we exposed three strains of C. neoformans (1 clinical and 2 environmental) to predation by Acanthamoeba castellanii for prolonged times and then analyzed surviving colonies phenotypically and genetically. Surviving colonies comprised cells that expressed either pseudohyphal or yeast phenotypes, which demonstrated variable expression of traits associated with virulence, such as capsule size, urease production, and melanization. Phenotypic changes were associated with aneuploidy and DNA sequence mutations in some amoeba-passaged isolates, but not in others. Mutations in the gene encoding the oligopeptide transporter (CNAG_03013; OPT1) were observed among amoeba-passaged isolates from each of the three strains. Isolates derived from environmental strains gained the capacity for enhanced macrophage toxicity after amoeba selection and carried mutations on the CNAG_00570 gene encoding Pkr1 (AMP-dependent protein kinase regulator) but manifested reduced virulence in mice because they elicited more effective fungal-clearing immune responses. Our results indicate that C. neoformans survival under constant amoeba predation involves the generation of strains expressing pleiotropic phenotypic and genetic changes. Given the myriad potential predators in soils, the diversity observed among amoeba-selected strains suggests a bet-hedging strategy whereby variant diversity increases the likelihood that some will survive predation.IMPORTANCECryptococcus neoformans is a ubiquitous environmental fungus that is also a leading cause of fatal fungal infection in humans, especially among immunocompromised patients. A major question in the field is how an environmental yeast such as C. neoformans becomes a human pathogen when it has no need for an animal host in its life cycle. Previous studies showed that C. neoformans increases its pathogenicity after interacting with its environmental predator amoebae. Amoebae, like macrophages, are phagocytic cells that are considered an environmental training ground for pathogens to resist macrophages, but the mechanism by which C. neoformans changes its virulence through interactions with protozoa is unknown. Our study indicates that fungal survival in the face of amoeba predation is associated with the emergence of pleiotropic phenotypic and genomic changes that increase the chance of fungal survival, with this diversity suggesting a bet-hedging strategy to ensure that some forms survive.
Collapse
Affiliation(s)
- Man Shun Fu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Livia C Liporagi-Lopes
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Samuel R Dos Santos
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Jennifer L Tenor
- Division of Infectious Diseases, Department of Medicine and Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - John R Perfect
- Division of Infectious Diseases, Department of Medicine and Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
19
|
“Feast-Fit-Fist-Feat”: Overview of Free-living Amoeba Interactions with Fungi and Virulence as a Foundation for Success in Battle. CURRENT TROPICAL MEDICINE REPORTS 2021. [DOI: 10.1007/s40475-020-00220-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Hubert F, Rodier MH, Minoza A, Portet-Sulla V, Cateau E, Brunet K. Free-living amoebae promote Candida auris survival and proliferation in water. Lett Appl Microbiol 2020; 72:82-89. [PMID: 32978979 DOI: 10.1111/lam.13395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022]
Abstract
Candida auris is an emerging species responsible for life-threatening infections. Its ability to be resistant to most systemic antifungal classes and its capacity to persist in a hospital environment have led to health concerns. Currently, data about environmental reservoirs are limited but remain essential in control of C. auris spread. The aim of our study was to explore the interactions between C. auris and two free-living amoeba (FLA) species, Vermamoeba vermiformis and Acanthamoeba castellanii, potentially found in the same water environment. Candida auris was incubated with FLA trophozoites or their culture supernatants. The number of FLA and yeasts was determined at different times and transmission electron microscopy (TEM) was performed. Supernatants of FLAs promoted yeast survival and proliferation. Internalization of viable C. auris within both FLA species was also evidenced by TEM. A water environmental reservoir of C. auris can therefore be considered through FLAs and contamination of the hospital water networks would consequently be possible.
Collapse
Affiliation(s)
- F Hubert
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Département des agents infectieux, Service de Mycologie-Parasitologie, CHU Poitiers, Poitiers, France
| | - M-H Rodier
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Département des agents infectieux, Service de Mycologie-Parasitologie, CHU Poitiers, Poitiers, France.,UMR CNRS 7267, Poitiers, France
| | - A Minoza
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Département des agents infectieux, Service de Mycologie-Parasitologie, CHU Poitiers, Poitiers, France
| | - V Portet-Sulla
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Département des agents infectieux, Service de Mycologie-Parasitologie, CHU Poitiers, Poitiers, France
| | - E Cateau
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Département des agents infectieux, Service de Mycologie-Parasitologie, CHU Poitiers, Poitiers, France.,UMR CNRS 7267, Poitiers, France
| | - K Brunet
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Département des agents infectieux, Service de Mycologie-Parasitologie, CHU Poitiers, Poitiers, France.,INSERM U1070, Poitiers, France
| |
Collapse
|
21
|
de Faria LV, do Carmo PHF, da Costa MC, Peres NTA, Rodrigues Chagas IA, Furst C, Ferreira GF, Costa AO, Santos DA. Acanthamoeba castellanii as an alternative interaction model for the dermatophyte Trichophyton rubrum. Mycoses 2020; 63:1331-1340. [PMID: 32869415 DOI: 10.1111/myc.13173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Trichophyton rubrum (Tr) is the main aetiological agent of human dermatophytosis, being isolated from the environment and keratinised tissues. In the environment, Tr can interact with other organisms, such as free-living amoebas (FLA), which can act as an alternative host system to study the interaction between microbes and phagocytic cells. OBJECTIVES To characterise the Acanthamoeba castellanii (ALX)-Tr interaction. METHODS Interaction was characterised in three conditions: trophozoites (PYG), late (PYG/NES) and early (NES) encystation stimulus, evaluating encystation kinetics, phagocytosis, exocytosis and fungicidal activity dynamics. RESULTS Tr was able to induce ALX encystation and be internalised by ALX. The number of internalised conidia was high at 1 hour, and ALX presented fungicidal activity with increased intracellular ROS production and exocytosis. In PYG/NES, phagocytosis and ROS production were reduced, with decreased ALX's fungicidal activity. However, in NES there was an increased fungal engulfment, and a reduced ROS production and higher fungal burden. Furthermore, exogenous mannose decreased phagocytosis of Tr conidia, and divalent cations induced ROS production and increased ALX's fungicidal activity. Interestingly, phagocytosis was reduced in the presence of cytoskeleton inhibitor, but exocytosis was increased, suggesting that Tr conidia may have alternative pathways to escape ALX's cells. CONCLUSION A castellanii is a proper model for studying Tr-FLA interaction, since ALX can engulf, produce ROS and kill Tr, and all these parameters are influenced by an encystation stimulus and divalent cations. Moreover, this interaction is likely to occur in the environment implicating in the adaptation to environmental stressful conditions in both organisms.
Collapse
Affiliation(s)
- Lucas V de Faria
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paulo H F do Carmo
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marliete C da Costa
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nalu T A Peres
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela A Rodrigues Chagas
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cinthia Furst
- Departamento de Patologia, Centro Ciências da Saúde, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | - Gabriella F Ferreira
- Programa Multicêntrico de Pós Graduação em Bioquímica e Biologia Molecular, Universidade Federal de Juiz de Fora, Governador Valadares, Brazil
| | - Adriana O Costa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel A Santos
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
22
|
Fisher MC, Gurr SJ, Cuomo CA, Blehert DS, Jin H, Stukenbrock EH, Stajich JE, Kahmann R, Boone C, Denning DW, Gow NAR, Klein BS, Kronstad JW, Sheppard DC, Taylor JW, Wright GD, Heitman J, Casadevall A, Cowen LE. Threats Posed by the Fungal Kingdom to Humans, Wildlife, and Agriculture. mBio 2020; 11:e00449-20. [PMID: 32371596 PMCID: PMC7403777 DOI: 10.1128/mbio.00449-20] [Citation(s) in RCA: 264] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The fungal kingdom includes at least 6 million eukaryotic species and is remarkable with respect to its profound impact on global health, biodiversity, ecology, agriculture, manufacturing, and biomedical research. Approximately 625 fungal species have been reported to infect vertebrates, 200 of which can be human associated, either as commensals and members of our microbiome or as pathogens that cause infectious diseases. These organisms pose a growing threat to human health with the global increase in the incidence of invasive fungal infections, prevalence of fungal allergy, and the evolution of fungal pathogens resistant to some or all current classes of antifungals. More broadly, there has been an unprecedented and worldwide emergence of fungal pathogens affecting animal and plant biodiversity. Approximately 8,000 species of fungi and Oomycetes are associated with plant disease. Indeed, across agriculture, such fungal diseases of plants include new devastating epidemics of trees and jeopardize food security worldwide by causing epidemics in staple and commodity crops that feed billions. Further, ingestion of mycotoxins contributes to ill health and causes cancer. Coordinated international research efforts, enhanced technology translation, and greater policy outreach by scientists are needed to more fully understand the biology and drivers that underlie the emergence of fungal diseases and to mitigate against their impacts. Here, we focus on poignant examples of emerging fungal threats in each of three areas: human health, wildlife biodiversity, and food security.
Collapse
Affiliation(s)
- Matthew C Fisher
- MRC Centre for Global Infectious Disease Analysis, Imperial College, London, United Kingdom
| | - Sarah J Gurr
- Department of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - David S Blehert
- U.S. Geological Survey, National Wildlife Health Center, Madison, Wisconsin, USA
| | - Hailing Jin
- Department of Microbiology and Plant Pathology, Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California-Riverside, Riverside, California, USA
| | - Eva H Stukenbrock
- Max Planck Fellow Group Environmental Genomics, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Environmental Genomics, Christian-Albrechts University, Kiel, Germany
| | - Jason E Stajich
- Department of Microbiology and Plant Pathology, Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California-Riverside, Riverside, California, USA
| | - Regine Kahmann
- Max Planck Institute for Terrestrial Microbiology, Department of Organismic Interactions, Marburg, Germany
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - David W Denning
- The National Aspergillosis Centre, Wythenshawe Hospital, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Neil A R Gow
- Department of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Bruce S Klein
- Department of Pediatrics, Department of Internal Medicine, and Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - James W Kronstad
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Donald C Sheppard
- McGill Interdisciplinary Initiative in Infection and Immunology, Departments of Medicine, Microbiology & Immunology, McGill University, Montreal, Canada
| | - John W Taylor
- University of California-Berkeley, Department of Plant and Microbial Biology, Berkeley, California, USA
| | - Gerard D Wright
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Azzam SZ, Cayme GJ, Martinez LR. Polymicrobial interactions involving fungi and their importance for the environment and in human disease. Microb Pathog 2020; 140:103942. [PMID: 31881258 PMCID: PMC7105396 DOI: 10.1016/j.micpath.2019.103942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/08/2023]
Abstract
Understanding polymicrobial interactions involving fungi in the environment and the human mycobiome is necessary to address environmental and medically related problems such as drought or antimicrobial resistance. The diversity of these interactions highlights the complexity of fungi, considering how some interactions can be antagonistic, while others synergistic. Over the years, an increase in studies on the mycobiome have revealed similarities between the human and environmental hosts. More recently, studies have focused on microbial commensal relationships and identifying causative agents of human disease. The overlap of some of these interactions is impossible to ignore, indicating that there are areas for medical exploitation that need to be further investigated. This review provides the latest advances in polymicrobial interactions involving fungi and discusses the importance of the fungal lifestyle in the environment and in human disease.
Collapse
Affiliation(s)
- Seham Z Azzam
- Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, TX, USA
| | - Ginelle J Cayme
- Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, TX, USA
| | - Luis R Martinez
- Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, TX, USA; Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA.
| |
Collapse
|