1
|
Zhu WY, Segeren L, Chan N, Ball B, Geddes-McAlister J. Proteome remodeling upon disruption of virulence-associated CipC from Cryptococcus neoformans. Microbiol Resour Announc 2025; 14:e0113824. [PMID: 40047507 PMCID: PMC11984179 DOI: 10.1128/mra.01138-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/17/2025] [Indexed: 04/11/2025] Open
Abstract
The human fungal pathogen, Cryptococcus neoformans, produces an array of virulence factors that regulate the production of a polysaccharide capsule and melanin, thermotolerance, and the release of extracellular enzymes. Here, we applied quantitative proteomics to investigate the role of CipC, a known virulence-associated protein, in the fungus.
Collapse
Affiliation(s)
- Wan Yun Zhu
- Molecular and Cellular Biology Department, University Of Guelph, Guelph, Ontario, Canada
| | - Lauren Segeren
- Molecular and Cellular Biology Department, University Of Guelph, Guelph, Ontario, Canada
| | - Norris Chan
- Molecular and Cellular Biology Department, University Of Guelph, Guelph, Ontario, Canada
| | - Brianna Ball
- Molecular and Cellular Biology Department, University Of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
2
|
Ramakrishnan G, Miskolci V, Hunter M, Giese MA, Münch D, Hou Y, Eliceiri KW, Lasarev MR, White RM, Huttenlocher A. Real-time imaging reveals a role for macrophage protrusive motility in melanoma invasion. J Cell Biol 2025; 224:e202403096. [PMID: 39570286 PMCID: PMC11586626 DOI: 10.1083/jcb.202403096] [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: 03/15/2024] [Revised: 08/26/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024] Open
Abstract
Macrophages are primary cells of the innate immune system that mediate tumor progression. However, the motile behavior of macrophages and interactions with tumor cells are not well understood. Here, we exploit the optical transparency of larval zebrafish and perform real-time imaging of macrophage-melanoma interactions. We found that macrophages are highly motile in the tumor microenvironment. Macrophages extend dynamic projections between tumor cells that precede invasive melanoma migration. Modulating macrophage motility with a dominant inhibitory mutation in Rac2 inhibits recruitment to the tumor and impairs tumor invasion. However, a hyperactivating mutation in Rac2 does not affect macrophage recruitment but limits macrophage projections into the melanoma mass and reduces invasive melanoma cell migration. Taken together, these findings reveal a role for Rac2-mediated macrophage protrusive motility in melanoma invasion.
Collapse
Affiliation(s)
- Gayathri Ramakrishnan
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Veronika Miskolci
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Miranda Hunter
- Cancer Biology and Genetics and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Morgan A. Giese
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Daniela Münch
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Kevin W. Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Michael R. Lasarev
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Richard M. White
- Cancer Biology and Genetics and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
3
|
Yamaguchi N, Otsuna H, Eisenberg-Bord M, Ramakrishnan L. An Image Processing Tool for Automated Quantification of Bacterial Burdens in Zebrafish Larvae. Zebrafish 2025; 22:11-14. [PMID: 39718816 PMCID: PMC11971606 DOI: 10.1089/zeb.2024.0170] [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: 12/25/2024] Open
Abstract
Zebrafish larvae are used to model the pathogenesis of multiple bacteria. This transparent model offers the unique advantage of allowing quantification of fluorescent bacterial burdens (fluorescent pixel counts [FPC]) in vivo by facile microscopical methods, replacing enumeration of bacteria using time-intensive plating of lysates on bacteriological media. Accurate FPC measurements require laborious manual image processing to mark the outside borders of the animals so as to delineate the bacteria inside the animals from those in the culture medium that they are in. Here, we have developed an automated ImageJ/Fiji-based macro that accurately detects the outside borders of Mycobacterium marinum-infected larvae.
Collapse
Affiliation(s)
- Naoya Yamaguchi
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michal Eisenberg-Bord
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Lalita Ramakrishnan
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
4
|
Sami MRS, Rani NA, Elahi MME, Hossain MS, Al Mueid MA, Rahim Z, Patil RB, Moin AT, Bithi IJ, Nahar S, Konika IJ, Roy S, Preya JA, Ahmed J. An immunoinformatics and extensive molecular dynamics study to develop a polyvalent multi-epitope vaccine against cryptococcosis. PLoS One 2024; 19:e0315105. [PMID: 39739919 PMCID: PMC11687922 DOI: 10.1371/journal.pone.0315105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
Cryptococcosis is a lethal mycosis instigated by the pathogenic species Cryptococcus neoformans and Cryptococcus gattii, primarily affects the lungs, manifesting as pneumonia, and the brain, where it presents as meningitis. Mortality rate could reach 100% if infections remain untreated in cryptococcal meningitis. Treatment options for cryptococcosis are limited and and there are no licensed vaccines clinically available to treat or prevent cryptococcosis. Our study utilizes an integrated bioinformatics approaches to develop a polyvalent multiepitope subunit vaccine focusing on the key virulent proteins Heat shock transcription factor and Chaperone DnaK of both C. neoformans and C. gatti. Then in silico analysis was done to predict highly antigenic epitopes by assessing antigenicity, transmembrane topology screening, allergenecity, toxicity, and molecular docking approaches. Following this analysis, we designed two vaccine constructs integrating a compatible adjuvant and suitable linkers. These constructs exhibited notable characteristics including high antigenicity, non-toxicity, solubility, stability, and compatibility with Toll-like receptors (TLRs). The interaction between both vaccine constructs and TLR2, TLR3, and TLR9 was assessed through molecular docking analysis. Molecular dynamics simulations and MM-PBSA calculations suggest the substantial stabilizing property and binding affinity of Vaccine Construct V1 against TLR9. Both the vaccines revealed to have a higher number of interchain hydrogen bond with TLR9. These findings serve as a crucial stepping stone towards a comprehensive solution for combating cryptococcus infections induced by both C. neoformans and C. gattii. Further validation through in vivo studies is crucial to confirm the effectiveness and potential of the vaccine to curb the spread of cryptococcosis. Subsequent validation through in vivo studies is paramount to confirm the effectiveness and potential of the vaccine in reducing the spread of cryptococcosis.
Collapse
Affiliation(s)
- Md. Razwan Sardar Sami
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Nurul Amin Rani
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | | | - Mohammad Sajjad Hossain
- Department of Theoretical and Computational Chemistry, Faculty of Science, University of Dhaka, Dhaka, Bangladesh
| | - Minhaz Abdullah Al Mueid
- Department of Pharmacy, Faculty of Biological Science, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Zahidur Rahim
- Department of Zoology, Jahangirnagar University, Dhaka, Bangladesh
| | - Rajesh B. Patil
- Sinhgad Technical Education Societys, Sinhgad College of Pharmacy, Department of Pharmaceutical Chemistry, Pune, Maharashtra, India
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science, University of Chittagong, Chattogram, Bangladesh
| | - Israt Jahan Bithi
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Sabekun Nahar
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Israt Jahan Konika
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Sneha Roy
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Jannatul Aleya Preya
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Jamil Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Biochemistry and Chemistry, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| |
Collapse
|
5
|
Trecourt A, Rabodonirina M, Donzel M, Chapey-Picq E, Bentaher A, Dupont D, Miossec C, Persat F, Wallon M, Lemoine JP, Tirard-Collet P, Baltrès A, Alanio A, Devouassoux-Shisheboran M, Menotti J. Cryptococcus neoformans/gattii and Histoplasma capsulatum var. capsulatum infections on tissue sections: Diagnostic pitfalls and relevance of an integrated histomolecular diagnosis. Med Mycol 2024; 63:myae126. [PMID: 39732625 PMCID: PMC11735191 DOI: 10.1093/mmy/myae126] [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: 10/27/2024] [Revised: 12/18/2024] [Accepted: 12/27/2024] [Indexed: 12/30/2024] Open
Abstract
Cryptococcus neoformans/gattii and Histoplasma capsulatum var. capsulatum may present atypical histopathological features inducing diagnostic errors. We aimed to estimate the frequency of these atypical features in formalin-fixed tissue (FT) samples and to assess the relevance of an integrated histomolecular diagnosis using specific H. capsulatum PCR and panfungal PCR followed by Sanger sequencing and/or targeted massive parallel sequencing (MPS). A total of 27 FT from 23 patients with a histopathological diagnosis of cryptococcosis (n = 16 FT from 13 patients) or histoplasmosis (n = 11 FT from 10 patients) were retrospectively included. All FT were consultation cases. Mycological identifications on equivalent fresh tissue were available for 11/23 (47.8%) patients. The expert pathologist review modified the diagnosis suggested by the initial pathologist in 7/27 (25.9%) FT. Fungal morphology and tissue inflammation were compared between both mycoses. The most discriminant atypical criterion was the presence of dented-looking yeasts, observed in 68.75% (11/16) of C. neoformans/gattii and none (0/11) of H. capsulatum var. capsulatum (P = .002). For the 12/23 (52.2%) patients without mycological identification on fresh tissue, an integrated histomolecular diagnosis on FT using specific PCR or panfungal PCR followed by Sanger sequencing and/or MPS led to fungal identification in 9/12 (75%) cases; for cryptococcosis, the targeted MPS sensitivity was higher than that of Sanger sequencing (P = .041). Thus, because atypical histopathological features may be tricky, integrated histomolecular diagnosis is essential for optimal patient care.
Collapse
Affiliation(s)
- Alexis Trecourt
- Service de Pathologie Multi-Site - Site Sud, Centre Hospitalier Lyon Sud, Hospices Civil de Lyons, Lyon, France
- Faculté de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, UR 3738 – CICLY – Equipe Inflammation et immunité de l′épithélium respiratoire, Lyon, France
| | - Meja Rabodonirina
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Sud Charles Mérieux, Université Claude Bernard Lyon-1, Lyon, France
| | - Marie Donzel
- Service de Pathologie Multi-Site - Site Sud, Centre Hospitalier Lyon Sud, Hospices Civil de Lyons, Lyon, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052, CNRS UMR 5286, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon 1, Lyon, France
| | - Emmanuelle Chapey-Picq
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Sud Charles Mérieux, Université Claude Bernard Lyon-1, Lyon, France
| | - Abderrazzak Bentaher
- Faculté de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, UR 3738 – CICLY – Equipe Inflammation et immunité de l′épithélium respiratoire, Lyon, France
| | - Damien Dupont
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon, France
| | - Charline Miossec
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Florence Persat
- Faculté de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, UR 3738 – CICLY – Equipe Inflammation et immunité de l′épithélium respiratoire, Lyon, France
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon, France
| | - Martine Wallon
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Sud Charles Mérieux, Université Claude Bernard Lyon-1, Lyon, France
| | - Jean-Philippe Lemoine
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Pauline Tirard-Collet
- Faculté de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, UR 3738 – CICLY – Equipe Inflammation et immunité de l′épithélium respiratoire, Lyon, France
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon, France
- Génomique épidémiologique des maladies infectieuses (GENEPII), Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Aline Baltrès
- Service de Pathologie, Centre Léon Berard, Lyon, France
| | - Alexandre Alanio
- Groupe Hospitalier Saint-Louis-Lariboisière-Fernand-Widal, Assistance Publique-Hôpitaux de Paris, Paris, France
- Molecular Mycology Unit, CNRS UMR2000, Institut Pasteur, Paris, France
| | - Mojgan Devouassoux-Shisheboran
- Service de Pathologie Multi-Site - Site Sud, Centre Hospitalier Lyon Sud, Hospices Civil de Lyons, Lyon, France
- Faculté de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, UR 3738 – CICLY – Equipe Inflammation et immunité de l′épithélium respiratoire, Lyon, France
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon, France
| | - Jean Menotti
- Faculté de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, UR 3738 – CICLY – Equipe Inflammation et immunité de l′épithélium respiratoire, Lyon, France
- Service de Parasitologie et Mycologie Médicale, Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon, France
- Génomique épidémiologique des maladies infectieuses (GENEPII), Institut des Agents Infectieux, Hôpital Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
6
|
Martinez Barrera S, Hatchell E, Byrum SD, Mackintosh SG, Kozubowski L. Quantitative analysis of septin Cdc10 & Cdc3-associated proteome during stress response in the fungal pathogen Cryptococcus neoformans. PLoS One 2024; 19:e0313444. [PMID: 39689097 DOI: 10.1371/journal.pone.0313444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/23/2024] [Indexed: 12/19/2024] Open
Abstract
Cryptococcus neoformans is a pathogenic basidiomycetous yeast that primarily infects immunocompromised individuals. Fatal outcome of cryptococcosis depends on the ability of C. neoformans to sense and adapt to 37°C. A complex of conserved filament forming GTPases, called septins, composed of Cdc3, Cdc10, Cdc11, and Cdc12, assembles at the mother-bud neck in C. neoformans. Septins Cdc3 and Cdc12 are essential for proliferation of C. neoformans at 37°C and for virulence in the Galleria mellonella model of infection, presumably due to their requirement for septin complex formation, and the involvement in cytokinesis. However, how exactly Cdc3, and Cdc12 contribute to C. neoformans growth at 37°C remains unknown. Based on studies investigating roles of septins in Saccharomyces cerevisiae, septin complex at the mother-bud neck of C. neoformans is predicted to interact with proteins involved in cell cycle control, morphogenesis, and cytokinesis, but the septin-associated proteome in C. neoformans has not been investigated. Here, we utilized tandem mass spectrometry to define C. neoformans proteins that associate with either Cdc3 or Cdc10 at ∼25°C or after the shift to 37°C. Our findings unveil a diverse array of septin-associated proteins, highlighting potential roles of septins in cell division, and stress response. Two proteins, identified as associated with both Cdc3 and Cdc10, the actin-binding protein profilin, which was detected at both temperatures, and ATP-binding multi-drug transporter Afr1, which was detected exclusively at 37°C, were further confirmed by co-immunoprecipitation. We also confirmed that association of Cdc3 with Afr1 was enhanced at 37°C. Upon shift to 37°C, septins Cdc3 and Cdc10 exhibited altered localization and Cdc3 partially co-localized with Afr1. In addition, we also investigated changes to levels of individual C. neoformans proteins upon shift from ∼25 to 37°C in exponentially grown culture and when cells entered stationary phase at ∼25°C. Our study reveals changes to C. neoformans proteome associated with heat and nutrient deprivation stresses and provides a landscape of septin-associated C. neoformans proteome, which will facilitate elucidating the biology of septins and mechanisms of stress response in this fungal pathogen.
Collapse
Affiliation(s)
- Stephani Martinez Barrera
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, United States of America
| | - Emma Hatchell
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, United States of America
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Lukasz Kozubowski
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, United States of America
| |
Collapse
|
7
|
Bednarek JM, Brown JCS. Elements of dissemination in cryptococcosis. mBio 2024; 15:e0215523. [PMID: 39470312 PMCID: PMC11633103 DOI: 10.1128/mbio.02155-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Abstract
As healthcare improves and our ability to support patients with compromised immune systems increases, such patients become more vulnerable to microbes in the environment. These include fungal pathogens such as Cryptococcus neoformans, the primary cause of fungal meningitis and a top priority pathogen on the World Health Organization fungal pathogen list. Like many other environmental pathogens, C. neoformans must adapt to and thrive in diverse environments in order to cause disease: (i) the environmental niche, (ii) the lungs following inhalation of infectious particles, (iii) the bloodstream and/or lymphatic system during dissemination, and (iv) the central nervous system (CNS), where it causes a deadly cryptococcal meningoencephalitis. Because CNS infection is the driver of mortality and the presenting illness, understanding the dissemination process from both host and fungal perspectives is important for treating these infections. In this review, we discuss the different stages of dissemination, how fungal cells interact with host cells during disease, and the ability to adapt to different environments within hosts.
Collapse
Affiliation(s)
- Joseph M. Bednarek
- School of Biological Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Jessica C. S. Brown
- School of Biological Sciences, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Brown GD, Ballou ER, Bates S, Bignell EM, Borman AM, Brand AC, Brown AJP, Coelho C, Cook PC, Farrer RA, Govender NP, Gow NAR, Hope W, Hoving JC, Dangarembizi R, Harrison TS, Johnson EM, Mukaremera L, Ramsdale M, Thornton CR, Usher J, Warris A, Wilson D. The pathobiology of human fungal infections. Nat Rev Microbiol 2024; 22:687-704. [PMID: 38918447 DOI: 10.1038/s41579-024-01062-w] [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] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
Human fungal infections are a historically neglected area of disease research, yet they cause more than 1.5 million deaths every year. Our understanding of the pathophysiology of these infections has increased considerably over the past decade, through major insights into both the host and pathogen factors that contribute to the phenotype and severity of these diseases. Recent studies are revealing multiple mechanisms by which fungi modify and manipulate the host, escape immune surveillance and generate complex comorbidities. Although the emergence of fungal strains that are less susceptible to antifungal drugs or that rapidly evolve drug resistance is posing new threats, greater understanding of immune mechanisms and host susceptibility factors is beginning to offer novel immunotherapeutic options for the future. In this Review, we provide a broad and comprehensive overview of the pathobiology of human fungal infections, focusing specifically on pathogens that can cause invasive life-threatening infections, highlighting recent discoveries from the pathogen, host and clinical perspectives. We conclude by discussing key future challenges including antifungal drug resistance, the emergence of new pathogens and new developments in modern medicine that are promoting susceptibility to infection.
Collapse
Affiliation(s)
- Gordon D Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Elizabeth R Ballou
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Steven Bates
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elaine M Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Andrew M Borman
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alexandra C Brand
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Carolina Coelho
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Peter C Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rhys A Farrer
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Nelesh P Govender
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - William Hope
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - J Claire Hoving
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rachael Dangarembizi
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Thomas S Harrison
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elizabeth M Johnson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Liliane Mukaremera
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark Ramsdale
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
9
|
Bosch C, Toplis B, Van Staden AD, Volschenk H, Smith C, Dicks L, Botha A. Exposure of Cryptococcus neoformans to low nitrogen levels enhances virulence. Int Microbiol 2024; 27:1587-1595. [PMID: 38483745 PMCID: PMC11452460 DOI: 10.1007/s10123-024-00504-y] [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: 10/21/2023] [Revised: 02/18/2024] [Accepted: 03/07/2024] [Indexed: 10/05/2024]
Abstract
Previous studies have shown a correlation between nitrogen levels and Cryptococcus neoformans pathogenicity. Here we report on the in vivo effects of cryptococcal pre-exposure to ecologically relevant nitrogen levels. C. neoformans H99 was cultured in yeast carbon base (YCB) supplemented with 0.53 g/L NH4Cl and 0.21 g/L NH4Cl, respectively, and used to infect larvae of the Greater Wax moth, Galleria mellonella. Cells cultured in low nitrogen YCB (LN) were more virulent compared to cells cultured in high nitrogen YCB (HN). Microscopic examination of haemolymph collected from infected larvae revealed that cells cultured in LN were larger than cells cultured in HN, with the majority of LN cells exceeding 10 µm and possibly entering titanisation. Additionally, compared to HN-cultured cells, fewer LN-cultured cells were engulfed by macrophages. The enhanced virulence of LN-cultured cells was attributed to the increased cell size in vivo. In contrast, reduced macrophage uptake was attributed to increased capsule thickness of in vitro cells. Not only do these findings demonstrate the effects of culture conditions, specifically nitrogen levels, on C. neoformans virulence, but they also highlight the importance of isolate background in the cryptococcal-host interaction.
Collapse
Affiliation(s)
- Caylin Bosch
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Barbra Toplis
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Anton DuPreez Van Staden
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Heinrich Volschenk
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Carine Smith
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Leon Dicks
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Alfred Botha
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
10
|
Yamaguchi N, Otsuna H, Eisenberg-Bord M, Ramakrishnan L. An Image Processing Tool for Automated Quantification of Bacterial Burdens in Zebrafish Larvae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608298. [PMID: 39229075 PMCID: PMC11370481 DOI: 10.1101/2024.08.16.608298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Zebrafish larvae are used to model the pathogenesis of multiple bacteria. This transparent model offers the unique advantage of allowing quantification of fluorescent bacterial burdens (fluorescent pixel counts: FPC) in vivo by facile microscopical methods, replacing enumeration of bacteria using time-intensive plating of lysates on bacteriological media. Accurate FPC measurements require laborious manual image processing to mark the outside borders of the animals so as to delineate the bacteria inside the animals from those in the culture medium that they are in. Here, we have developed an automated ImageJ/Fiji-based macro that accurately detect the outside borders of Mycobacterium marinum-infected larvae.
Collapse
Affiliation(s)
- Naoya Yamaguchi
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, CB2 0AW Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
- Equal contributions
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
- Equal contributions
| | - Michal Eisenberg-Bord
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, CB2 0AW Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, CB2 0AW Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
| |
Collapse
|
11
|
Al-Huthaifi AM, Radman BA, Al-Alawi AA, Mahmood F, Liu TB. Mechanisms and Virulence Factors of Cryptococcus neoformans Dissemination to the Central Nervous System. J Fungi (Basel) 2024; 10:586. [PMID: 39194911 DOI: 10.3390/jof10080586] [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: 07/09/2024] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Cryptococcosis is a prevalent fungal infection of the central nervous system (CNS) caused by Cryptococcus neoformans, a yeast with a polysaccharide capsule in the basidiomycete group. Normally, C. neoformans infects the respiratory tract and then breaches the blood-brain barrier (BBB), leading to meningitis or meningoencephalitis, which leads to hundreds of thousands of deaths each year. Although the mechanism by which C. neoformans infiltrates the BBB to invade the brain has yet to be fully understood, research has revealed that C. neoformans can cross the BBB using transcellular penetration, paracellular traversal, and infected phagocytes (the "Trojan horse" mechanism). The secretion of multiple virulence factors by C. neoformans is crucial in facilitating the spread of infection after breaching the BBB and causing brain infections. Extensive research has shown that various virulence factors play a significant role in the dissemination of infection beyond the lungs. This review explores the mechanisms of C. neoformans entering the CNS and explains how it bypasses the BBB. Additionally, it aims to understand the interplay between the regulatory mechanisms and virulence factors of C. neoformans.
Collapse
Affiliation(s)
| | - Bakeel A Radman
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | | | - Fawad Mahmood
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Tong-Bao Liu
- Medical Research Institute, Southwest University, Chongqing 400715, China
- Jinfeng Laboratory, Chongqing 401329, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, China
| |
Collapse
|
12
|
Gaylord EA, Choy HL, Chen G, Briner SL, Doering TL. Sac1 links phosphoinositide turnover to cryptococcal virulence. mBio 2024; 15:e0149624. [PMID: 38953635 PMCID: PMC11323556 DOI: 10.1128/mbio.01496-24] [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: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
Cryptococcus neoformans is an environmentally acquired fungal pathogen that causes over 140,000 deaths per year. Cryptococcal infection occurs when infectious particles are deposited into the lung, where they encounter host phagocytic cells. C. neoformans may be engulfed by these phagocytes, an important step of infection that leads to outcomes ranging from termination of infection to cryptococcal dissemination. To study this critical process, we screened approximately 4,700 cryptococcal gene deletion mutants for altered uptake, using primary mouse and human phagocytic cells. Among the hits of these two screens, we identified 93 mutants with perturbed uptake in both systems, as well as others with differences in uptake by only one cell type. We further screened the hits for changes in thickness of the capsule, a protective polysaccharide layer around the cell which is an important cryptococcal virulence factor. The combination of our three screens yielded 45 mutants, including one lacking the phosphatidylinositol-4-phosphate phosphatase Sac1. In this work, we implicate Sac1 in both host cell uptake and capsule production. We found that sac1 mutants exhibit lipid trafficking defects, reductions in secretory system function, and changes in capsule size and composition. Many of these changes occur specifically in tissue culture media, highlighting the role of Sac1 phosphatase activity in responding to the stress of host-like conditions. Overall, these findings show how genome-scale screening can identify cellular factors that contribute to our understanding of cryptococcal biology and demonstrate the role of Sac1 in determining fungal virulence.IMPORTANCECryptococcus neoformans is a fungal pathogen with significant impact on global health. Cryptococcal cells inhaled from the environment are deposited into the lungs, where they first contact the human immune system. The interaction between C. neoformans and host cells is critical because this step of infection can determine whether the fungal cells die or proliferate within the human host. Despite the importance of this stage of infection, we have limited knowledge of cryptococcal factors that influence its outcome. In this study, we identify cryptococcal genes that affect uptake by both human and mouse cells. We also identify mutants with altered capsule, a protective coating that surrounds the cells to shield them from the host immune system. Finally, we characterize the role of one gene, SAC1, in these processes. Overall, this study contributes to our understanding of how C. neoformans interacts with and protects itself from host cells.
Collapse
Affiliation(s)
- Elizabeth A. Gaylord
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hau Lam Choy
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Guohua Chen
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sydney L. Briner
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Lee CWJ, Brisland A, Qu X, Horianopoulos LC, Hu G, Mayer FL, Kronstad JW. Loss of Opi3 causes a lipid imbalance that influences the virulence traits of Cryptococcus neoformans but not cryptococcosis. Front Cell Infect Microbiol 2024; 14:1448229. [PMID: 39193507 PMCID: PMC11347413 DOI: 10.3389/fcimb.2024.1448229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
The basidiomycete fungus Cryptococcus neoformans is a useful model for investigating mechanisms of fungal pathogenesis in mammalian hosts. This pathogen is the causative agent of cryptococcal meningitis in immunocompromised patients and is in the critical priority group of the World Health Organization fungal priority pathogens list. In this study, we employed a mutant lacking the OPI3 gene encoding a methylene-fatty-acyl-phospholipid synthase to characterize the role of phosphatidylcholine (PC) and lipid homeostasis in the virulence of C. neoformans. We first confirmed that OPI3 was required for growth in nutrient limiting conditions, a phenotype that could be rescued with exogenous choline and PC. Additionally, we established that loss of Opi3 and the lack of PC lead to an accumulation of neutral lipids in lipid droplets and alterations in major lipid classes. The growth defect of the opi3Δ mutant was also rescued by sorbitol and polyethylene glycol (PEG), a result consistent with protection of ER function from the stress caused by lipid imbalance. We then examined the impact of Opi3 on virulence and found that the dependence of PC synthesis on Opi3 caused reduced capsule size and this was accompanied by an increase in shed capsule polysaccharide and changes in cell wall composition. Further tests of virulence demonstrated that survival in alveolar macrophages and the ability to cause disease in mice were not impacted by loss of Opi3 despite the choline auxotrophy of the mutant in vitro. Overall, this work establishes the contribution of lipid balance to virulence factor elaboration by C. neoformans and suggests that host choline is sufficient to support proliferation during disease.
Collapse
Affiliation(s)
- Christopher W. J. Lee
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Anna Brisland
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Xianya Qu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Linda C. Horianopoulos
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Guanggan Hu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - François L. Mayer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - James W. Kronstad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Li X, Paccoud O, Chan KH, Yuen KY, Manchon R, Lanternier F, Slavin MA, van de Veerdonk FL, Bicanic T, Lortholary O. Cryptococcosis Associated With Biologic Therapy: A Narrative Review. Open Forum Infect Dis 2024; 11:ofae316. [PMID: 38947739 PMCID: PMC11212009 DOI: 10.1093/ofid/ofae316] [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: 03/05/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
Cryptococcus is an opportunistic fungal pathogen that can cause disseminated infection with predominant central nervous system involvement in patients with compromised immunity. Biologics are increasingly used in the treatment of neoplasms and autoimmune/inflammatory conditions and the prevention of transplant rejection, which may affect human defense mechanisms against cryptococcosis. In this review, we comprehensively investigate the association between cryptococcosis and various biologics, highlighting their risks of infection, clinical manifestations, and clinical outcomes. Clinicians should remain vigilant for the risk of cryptococcosis in patients receiving biologics that affect the Th1/macrophage activation pathways, such as tumor necrosis factor α antagonists, Bruton tyrosine kinase inhibitors, fingolimod, JAK/STAT inhibitors (Janus kinase/signal transducer and activator of transcription), and monoclonal antibody against CD52. Other risk factors-such as age, underlying condition, and concurrent immunosuppressants, especially corticosteroids-should also be taken into account during risk stratification.
Collapse
Affiliation(s)
- Xin Li
- Department of Infectious Diseases and Tropical Medicine, Université Paris Cité, Necker-Enfants Malades University Hospital, Assistance Publique–Hôpitaux de Paris, IHU Imagine, Paris, France
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Olivier Paccoud
- Department of Infectious Diseases and Tropical Medicine, Université Paris Cité, Necker-Enfants Malades University Hospital, Assistance Publique–Hôpitaux de Paris, IHU Imagine, Paris, France
| | - Koon-Ho Chan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Romain Manchon
- Department of Infectious Diseases and Tropical Medicine, Université Paris Cité, Necker-Enfants Malades University Hospital, Assistance Publique–Hôpitaux de Paris, IHU Imagine, Paris, France
| | - Fanny Lanternier
- Department of Infectious Diseases and Tropical Medicine, Université Paris Cité, Necker-Enfants Malades University Hospital, Assistance Publique–Hôpitaux de Paris, IHU Imagine, Paris, France
- Institut Pasteur, National Reference Center for Invasive Mycoses and Antifungals, Mycology Translational Research Group, Mycology Department, Université Paris Cité, Paris, France
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, Australia
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, the Netherlands
| | - Tihana Bicanic
- Institute of Infection and Immunity, St George's University of London, London, UK
| | - Olivier Lortholary
- Department of Infectious Diseases and Tropical Medicine, Université Paris Cité, Necker-Enfants Malades University Hospital, Assistance Publique–Hôpitaux de Paris, IHU Imagine, Paris, France
- Institut Pasteur, National Reference Center for Invasive Mycoses and Antifungals, Mycology Translational Research Group, Mycology Department, Université Paris Cité, Paris, France
| |
Collapse
|
15
|
Hammond FR, Lewis A, Pollara G, Tomlinson GS, Noursadeghi M, Kiss-Toth E, Elks PM. Tribbles1 is host protective during in vivo mycobacterial infection. eLife 2024; 13:e95980. [PMID: 38896446 PMCID: PMC11186633 DOI: 10.7554/elife.95980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Tuberculosis is a major global health problem and is one of the top 10 causes of death worldwide. There is a pressing need for new treatments that circumvent emerging antibiotic resistance. Mycobacterium tuberculosis parasitises macrophages, reprogramming them to establish a niche in which to proliferate, therefore macrophage manipulation is a potential host-directed therapy if druggable molecular targets could be identified. The pseudokinase Tribbles1 (Trib1) regulates multiple innate immune processes and inflammatory profiles making it a potential drug target in infections. Trib1 controls macrophage function, cytokine production, and macrophage polarisation. Despite wide-ranging effects on leukocyte biology, data exploring the roles of Tribbles in infection in vivo are limited. Here, we identify that human Tribbles1 is expressed in monocytes and is upregulated at the transcript level after stimulation with mycobacterial antigen. To investigate the mechanistic roles of Tribbles in the host response to mycobacteria in vivo, we used a zebrafish Mycobacterium marinum (Mm) infection tuberculosis model. Zebrafish Tribbles family members were characterised and shown to have substantial mRNA and protein sequence homology to their human orthologues. trib1 overexpression was host-protective against Mm infection, reducing burden by approximately 50%. Conversely, trib1 knockdown/knockout exhibited increased infection. Mechanistically, trib1 overexpression significantly increased the levels of proinflammatory factors il-1β and nitric oxide. The host-protective effect of trib1 was found to be dependent on the E3 ubiquitin kinase Cop1. These findings highlight the importance of Trib1 and Cop1 as immune regulators during infection in vivo and suggest that enhancing macrophage TRIB1 levels may provide a tractable therapeutic intervention to improve bacterial infection outcomes in tuberculosis.
Collapse
Affiliation(s)
- Ffion R Hammond
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Amy Lewis
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Gabriele Pollara
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Gillian S Tomlinson
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Mahdad Noursadeghi
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Endre Kiss-Toth
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
16
|
Rutherford HA, Candeias D, Duncan CJA, Renshaw SA, Hamilton N. Macrophage transplantation rescues RNASET2-deficient leukodystrophy by replacing deficient microglia in a zebrafish model. Proc Natl Acad Sci U S A 2024; 121:e2321496121. [PMID: 38753517 PMCID: PMC11126979 DOI: 10.1073/pnas.2321496121] [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: 12/20/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
RNASET2-deficient leukodystrophy is a rare infantile white matter disorder mimicking a viral infection and resulting in severe psychomotor impairments. Despite its severity, there is little understanding of cellular mechanisms of pathogenesis and no treatments. Recent research using the rnaset2 mutant zebrafish model has suggested that microglia may be the drivers of the neuropathology, due to their failure to digest apoptotic debris during neurodevelopment. Therefore, we developed a strategy for microglial replacement through transplantation of adult whole kidney marrow-derived macrophages into embryonic hosts. Using live imaging, we revealed that transplant-derived macrophages can engraft within host brains and express microglia-specific markers, suggesting the adoption of a microglial phenotype. Tissue-clearing strategies revealed the persistence of transplanted cells in host brains beyond embryonic stages. We demonstrated that transplanted cells clear apoptotic cells within the brain, as well as rescue overactivation of the antiviral response otherwise seen in mutant larvae. RNA sequencing at the point of peak transplant-derived cell engraftment confirms that transplantation can reduce the brain-wide immune response and particularly, the antiviral response, in rnaset2-deficient brains. Crucially, this reduction in neuroinflammation resulted in behavioral rescue-restoring rnaset2 mutant motor activity to wild-type (WT) levels in embryonic and juvenile stages. Together, these findings demonstrate the role of microglia as the cellular drivers of neuropathology in rnaset2 mutants and that macrophage transplantation is a viable strategy for microglial replacement in the zebrafish. Therefore, microglia-targeted interventions may have therapeutic benefits in RNASET2-deficient leukodystrophy.
Collapse
Affiliation(s)
- Holly A. Rutherford
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Diogo Candeias
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| | - Christopher J. A. Duncan
- Immunology and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, NewcastleNE2 4HH, United Kingdom
- Department of Infection and Tropical Medicine, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals National Health Services Foundation Trust, NewcastleNE2 4HH, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Noémie Hamilton
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| |
Collapse
|
17
|
Speirs ZC, Loynes CA, Mathiessen H, Elks PM, Renshaw SA, Jørgensen LVG. What can we learn about fish neutrophil and macrophage response to immune challenge from studies in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109490. [PMID: 38471626 DOI: 10.1016/j.fsi.2024.109490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
Fish rely, to a high degree, on the innate immune system to protect them against the constant exposure to potential pathogenic invasion from the surrounding water during homeostasis and injury. Zebrafish larvae have emerged as an outstanding model organism for immunity. The cellular component of zebrafish innate immunity is similar to the mammalian innate immune system and has a high degree of sophistication due to the needs of living in an aquatic environment from early embryonic stages of life. Innate immune cells (leukocytes), including neutrophils and macrophages, have major roles in protecting zebrafish against pathogens, as well as being essential for proper wound healing and regeneration. Zebrafish larvae are visually transparent, with unprecedented in vivo microscopy opportunities that, in combination with transgenic immune reporter lines, have permitted visualisation of the functions of these cells when zebrafish are exposed to bacterial, viral and parasitic infections, as well as during injury and healing. Recent findings indicate that leukocytes are even more complex than previously anticipated and are essential for inflammation, infection control, and subsequent wound healing and regeneration.
Collapse
Affiliation(s)
- Zoë C Speirs
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Catherine A Loynes
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Heidi Mathiessen
- Laboratory of Experimental Fish Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C., Denmark
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Stephen A Renshaw
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Louise von Gersdorff Jørgensen
- Laboratory of Experimental Fish Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C., Denmark.
| |
Collapse
|
18
|
Shen S, Miskolci V, Dewey CN, Sauer JD, Huttenlocher A. Infection induced inflammation impairs wound healing through IL-1β signaling. iScience 2024; 27:109532. [PMID: 38577110 PMCID: PMC10993181 DOI: 10.1016/j.isci.2024.109532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/14/2024] [Accepted: 03/16/2024] [Indexed: 04/06/2024] Open
Abstract
Wound healing is impaired by infection; however, how microbe-induced inflammation modulates tissue repair remains unclear. We took advantage of the optical transparency of zebrafish and a genetically tractable microbe, Listeria monocytogenes, to probe the role of infection and inflammation in wound healing. Infection with bacteria engineered to activate the inflammasome, Lm-Pyro, induced persistent inflammation and impaired healing despite low bacterial burden. Inflammatory infections induced il1b expression and blocking IL-1R signaling partially rescued wound healing in the presence of persistent infection. We found a critical window of microbial clearance necessary to limit persistent inflammation and enable efficient wound repair. Taken together, our findings suggest that the dynamics of microbe-induced tissue inflammation impacts repair in complex tissue damage independent of bacterial load, with a critical early window for efficient tissue repair.
Collapse
Affiliation(s)
- Simone Shen
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Veronika Miskolci
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Colin N. Dewey
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
19
|
Hester MM, Carlson D, Lodge JK, Levitz SM, Specht CA. Immune evasion by Cryptococcus gattii in vaccinated mice coinfected with C. neoformans. Front Immunol 2024; 15:1356651. [PMID: 38469300 PMCID: PMC10925662 DOI: 10.3389/fimmu.2024.1356651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/05/2024] [Indexed: 03/13/2024] Open
Abstract
Cryptococcus neoformans and C. gattii, the etiologic agents of cryptococcosis, cause over 100,000 deaths worldwide every year, yet no cryptococcal vaccine has progressed to clinical trials. In preclinical studies, mice vaccinated with an attenuated strain of C. neoformans deleted of three cryptococcal chitin deacetylases (Cn-cda1Δ2Δ3Δ) were protected against a lethal challenge with C. neoformans strain KN99. While Cn-cda1Δ2Δ3Δ extended the survival of mice infected with C. gattii strain R265 compared to unvaccinated groups, we were unable to demonstrate fungal clearance as robust as that seen following KN99 challenge. In stark contrast to vaccinated mice challenged with KN99, we also found that R265-challenged mice failed to induce the production of protection-associated cytokines and chemokines in the lungs. To investigate deficiencies in the vaccine response to R265 infection, we developed a KN99-R265 coinfection model. In unvaccinated mice, the strains behaved in a manner which mirrored single infections, wherein only KN99 disseminated to the brain and spleen. We expanded the coinfection model to Cn-cda1Δ2Δ3Δ-vaccinated mice. Fungal burden, cytokine production, and immune cell infiltration in the lungs of vaccinated, coinfected mice were indicative of immune evasion by C. gattii R265 as the presence of R265 neither compromised the immunophenotype established in response to KN99 nor inhibited clearance of KN99. Collectively, these data indicate that R265 does not dampen a protective vaccine response, but rather suggest that R265 remains largely undetected by the immune system.
Collapse
Affiliation(s)
- Maureen M. Hester
- Department of Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Diana Carlson
- Department of Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Jennifer K. Lodge
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Stuart M. Levitz
- Department of Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Charles A. Specht
- Department of Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
20
|
Roosen L, Maes D, Musetta L, Himmelreich U. Preclinical Models for Cryptococcosis of the CNS and Their Characterization Using In Vivo Imaging Techniques. J Fungi (Basel) 2024; 10:146. [PMID: 38392818 PMCID: PMC10890286 DOI: 10.3390/jof10020146] [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: 11/30/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
Infections caused by Cryptococcus neoformans and Cryptococcus gattii remain a challenge to our healthcare systems as they are still difficult to treat. In order to improve treatment success, in particular for infections that have disseminated to the central nervous system, a better understanding of the disease is needed, addressing questions like how it evolves from a pulmonary to a brain disease and how novel treatment approaches can be developed and validated. This requires not only clinical research and research on the microorganisms in a laboratory environment but also preclinical models in order to study cryptococci in the host. We provide an overview of available preclinical models, with particular emphasis on models of cryptococcosis in rodents. In order to further improve the characterization of rodent models, in particular the dynamic aspects of disease manifestation, development, and ultimate treatment, preclinical in vivo imaging methods are increasingly used, mainly in research for oncological, neurological, and cardiac diseases. In vivo imaging applications for fungal infections are rather sparse. A second aspect of this review is how research on models of cryptococcosis can benefit from in vivo imaging methods that not only provide information on morphology and tissue structure but also on function, metabolism, and cellular properties in a non-invasive way.
Collapse
Affiliation(s)
- Lara Roosen
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Dries Maes
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Luigi Musetta
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
21
|
Boodwa-Ko D, Doering TL. A Quick reCAP: Discovering Cryptococcus neoformans Capsule Mutants. J Fungi (Basel) 2024; 10:114. [PMID: 38392786 PMCID: PMC10889740 DOI: 10.3390/jof10020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/24/2024] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen that can cause severe meningoencephalitis in immunocompromised hosts and is a leading cause of death in HIV/AIDS patients. This pathogenic yeast is surrounded by a polysaccharide capsule that is critical for virulence and plays important roles in host-pathogen interactions. Understanding capsule biosynthesis is therefore key to defining the biology of C. neoformans and potentially discovering novel therapeutic targets. By exploiting methods to identify mutants deficient in capsule, June Kwon-Chung and other investigators have discovered numerous genes involved in capsule biosynthesis and regulation. Successful approaches have incorporated combinations of techniques including mutagenesis and systematic gene deletion; complementation and genetic screens; morphological examination, physical separation, and antibody binding; and computational modeling based on gene expression analysis. In this review, we discuss these methods and how they have been used to identify capsule mutants.
Collapse
Affiliation(s)
| | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA;
| |
Collapse
|
22
|
Castro-Lopez N, Wormley FL. Models for Inducing Experimental Cryptococcosis in Mice. Methods Mol Biol 2024; 2775:29-46. [PMID: 38758309 DOI: 10.1007/978-1-0716-3722-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Cryptococcus neoformans and Cryptococcus gattii are the predominant etiological agents of cryptococcosis, a particularly problematic disease in immunocompromised individuals. The increased clinical use of immunosuppressive drugs, the inherent ability of Cryptococcus species to suppress and evade host immune responses, and the emergence of drug-resistant yeast support the need for model systems that facilitate the design of novel immunotherapies and antifungals to combat disease progression. The mouse model of cryptococcosis is a widely used system to study Cryptococcus pathogenesis and the efficacy of antifungal drugs in vivo. In this chapter, we describe three commonly used strategies to establish cryptococcosis in mice: intranasal, intratracheal, and intravenous inoculations. Also, we discuss the methodology for delivering drugs to mice via intraperitoneal injection.
Collapse
Affiliation(s)
| | - Floyd L Wormley
- Department of Biology, Texas Christian University, Fort Worth, TX, USA.
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
23
|
Senthil N, Pacifici N, Cruz-Acuña M, Diener A, Han H, Lewis JS. An Image Processing Algorithm for Facile and Reproducible Quantification of Vomocytosis. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:831-842. [PMID: 38155727 PMCID: PMC10751783 DOI: 10.1021/cbmi.3c00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 12/30/2023]
Abstract
Vomocytosis is a process that occurs when internalized fungal pathogens escape from phagocytes without compromising the viability of the pathogen and the host cell. Manual quantification of time-lapse microscopy videos is currently used as the standard to study pathogen behavior and vomocytosis incidence. However, human-driven quantification of vomocytosis (and the closely related phenomenon, exocytosis) is incredibly burdensome, especially when a large volume of cells and interactions needs to be analyzed. In this study, we designed a MATLAB algorithm that measures the extent of colocalization between the phagocyte and fungal cell (Cryptococcus neoformans; CN) and rapidly reports the occurrence of vomocytosis in a high throughput manner. Our code processes multichannel, time-lapse microscopy videos of cocultured CN and immune cells that have each been fluorescently stained with unique dyes and provides quantitative readouts of the spatiotemporally dynamic process that is vomocytosis. This study also explored metrics, such as the rate of change of pathogen colocalization with the host cell, that could potentially be used to predict vomocytosis occurrence based on the quantitative data collected. Ultimately, the algorithm quantifies vomocytosis events and reduces the time for video analysis from over 1 h to just 10 min, a reduction in labor of 83%, while simultaneously minimizing human error. This tool significantly minimizes the vomocytosis analysis pipeline, accelerates our ability to elucidate unstudied aspects of this phenomenon, and expedites our ability to characterize CN strains for the study of their epidemiology and virulence.
Collapse
Affiliation(s)
- Neeraj Senthil
- Department
of Biomedical Engineering, University of
California − Davis, Davis, California 95616, United States
| | - Noah Pacifici
- Department
of Biomedical Engineering, University of
California − Davis, Davis, California 95616, United States
| | - Melissa Cruz-Acuña
- Department
of Biomedical Engineering, University of
California − Davis, Davis, California 95616, United States
| | - Agustina Diener
- Department
of Biomedical Engineering, University of
California − Davis, Davis, California 95616, United States
| | - Hyunsoo Han
- Department
of Biomedical Engineering, University of
California − Davis, Davis, California 95616, United States
| | - Jamal S. Lewis
- Department
of Biomedical Engineering, University of
California − Davis, Davis, California 95616, United States
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
24
|
Caza M, Santos DA, Burden E, Brisland A, Hu G, Kronstad JW. Proteasome inhibition as a therapeutic target for the fungal pathogen Cryptococcus neoformans. Microbiol Spectr 2023; 11:e0190423. [PMID: 37750732 PMCID: PMC10580939 DOI: 10.1128/spectrum.01904-23] [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: 05/05/2023] [Accepted: 08/07/2023] [Indexed: 09/27/2023] Open
Abstract
The current therapeutic challenges for treating fungal diseases demand new approaches and new drugs. A promising strategy involves combination therapy with agents of distinct mechanisms of action to increase fungicidal activity and limit the impact of mutations leading to resistance. In this study, we evaluated the antifungal potential of bortezomib by examining the inhibition of proteasome activity, cell proliferation, and capsule production by Cryptococcus neoformans, the causative agent of fungal meningoencephalitis. Chemical genetic screens with collections of deletion mutants identified potential druggable targets for combination therapy with bortezomib. In vitro assays of combinations of bortezomib with flucytosine, chlorpromazine, bafilomycin A1, copper sulfate, or hydroxyurea revealed antifungal effects against C. neoformans. Furthermore, combination treatment with bortezomib and flucytosine in a murine inhalation model of cryptococcosis resulted in the improvement of neurological functions and reduced fungal replication and dissemination, leading to a delay in disease progression. This study therefore highlights the utility of chemical genetic screens to identify new therapeutic approaches as well as the antifungal potential of proteasome inhibition. IMPORTANCE Fungal diseases of humans are difficult to treat, and there is a clear need for additional antifungal drugs, better diagnostics, effective vaccines, and new approaches to deal with emerging drug resistance. Fungi are challenging to control because they share many common biochemical functions with their mammalian hosts and it is therefore difficult to identify fungal-specific targets for drug development. One approach is to employ existing antifungal drugs in combination with agents that target common cellular processes at levels that are (ideally) not toxic for the host. We pursued this approach in this study by examining the potential of the clinically approved proteasome inhibitor bortezomib to influence the proliferation and virulence of Cryptococcus neoformans. We found that the combination of bortezomib with the anti-cryptococcal drug flucytosine improved the survival of infected mice, thus demonstrating the potential of this strategy for antifungal therapy.
Collapse
Affiliation(s)
- Mélissa Caza
- Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Assis Santos
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elizabeth Burden
- Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Brisland
- Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guanggan Hu
- Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - James W. Kronstad
- Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Fusco-Almeida AM, de Matos Silva S, dos Santos KS, de Lima Gualque MW, Vaso CO, Carvalho AR, Medina-Alarcón KP, Pires ACMDS, Belizario JA, de Souza Fernandes L, Moroz A, Martinez LR, Ruiz OH, González Á, Mendes-Giannini MJS. Alternative Non-Mammalian Animal and Cellular Methods for the Study of Host-Fungal Interactions. J Fungi (Basel) 2023; 9:943. [PMID: 37755051 PMCID: PMC10533014 DOI: 10.3390/jof9090943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
In the study of fungal pathogenesis, alternative methods have gained prominence due to recent global legislation restricting the use of mammalian animals in research. The principle of the 3 Rs (replacement, reduction, and refinement) is integrated into regulations and guidelines governing animal experimentation in nearly all countries. This principle advocates substituting vertebrate animals with other invertebrate organisms, embryos, microorganisms, or cell cultures. This review addresses host-fungus interactions by employing three-dimensional (3D) cultures, which offer more faithful replication of the in vivo environment, and by utilizing alternative animal models to replace traditional mammals. Among these alternative models, species like Caenorhabditis elegans and Danio rerio share approximately 75% of their genes with humans. Furthermore, models such as Galleria mellonella and Tenebrio molitor demonstrate similarities in their innate immune systems as well as anatomical and physiological barriers, resembling those found in mammalian organisms.
Collapse
Affiliation(s)
- Ana Marisa Fusco-Almeida
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Samanta de Matos Silva
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
| | - Kelvin Sousa dos Santos
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Marcos William de Lima Gualque
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Carolina Orlando Vaso
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Angélica Romão Carvalho
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Kaila Petrolina Medina-Alarcón
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Ana Carolina Moreira da Silva Pires
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Jenyffie Araújo Belizario
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Lígia de Souza Fernandes
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Andrei Moroz
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Luis R. Martinez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA;
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA
- Center for Immunology and Transplantation, University of Florida, Gainesville, FL 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Orville Hernandez Ruiz
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
- Cellular and Molecular Biology Group University of Antioquia, Corporation for Biological Research, Medellin 050010, Colombia
| | - Ángel González
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
| | - Maria José Soares Mendes-Giannini
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| |
Collapse
|
26
|
Liu Y, Zhang Y, Zhao X, Lu W, Zhong Y, Fu YV. Antifungal Peptide SP1 Damages Polysaccharide Capsule of Cryptococcus neoformans and Enhances Phagocytosis of Macrophages. Microbiol Spectr 2023; 11:e0456222. [PMID: 36916981 PMCID: PMC10100895 DOI: 10.1128/spectrum.04562-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
Cryptococcus neoformans is a fungal pathogen which causes nearly half a million deaths worldwide each year. Under host-relevant conditions, it produces a characteristic polysaccharide capsule. The polysaccharide capsule is one of the main virulence factors of C. neoformans, which involves antiphagocytosis and immune responses of the host to cause a lack of an immune. Meanwhile, the polysaccharide capsule is a promising drug target because of the absence of analogs in the host. Here, we demonstrate that antifungal peptide SP1, which is derived from the N terminus of Saccharomyces cerevisiae GAPDH (glyceraldehyde-3-phosphate dehydrogenase), disrupts the polysaccharide capsule of C. neoformans H99. The mechanism is possibly due to the interaction of SP1 with glucuronoxylomannan (GXM). Disruption of the polysaccharide capsule enhances the adhesion and phagocytosis of C. neoformans H99 by macrophages and reduces the replication of C. neoformans H99 within macrophages. Additionally, SP1 exhibits antifungal activity against cryptococcal biofilms associated with the capsular polysaccharides. These findings suggest the potential of SP1 as a drug candidate for the treatment of cryptococcosis. IMPORTANCE C. neoformans is an opportunistic pathogen that causes invasive infections with a high mortality rate. Currently, the clinical drugs available for the treatment of cryptococcosis are limited to amphotericin B, azoles, and flucytosine. Amphotericin is nephrotoxic, and the widespread use of azoles and 5-flucytosine has led to a rapid development of drug resistance in C. neoformans. There is an urgent need to develop new and effective anticryptococcal drugs. Targeting virulence factors is a novel strategy for developing antifungal drugs. The antifungal peptide SP1 is capable of disrupting the polysaccharide capsule, which is a principal virulence factor of C. neoformans. Studying the mechanism by which SP1 damages the polysaccharide capsule and investigating the potential benefits of SP1 in removing C. neoformans from the host provides baseline data to develop a therapeutic strategy against refractory cryptococcal infections. This strategy would involve both inhibiting virulence factors and directly killing C. neoformans cells.
Collapse
Affiliation(s)
- Yan Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Zhang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xi Zhao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Weilai Lu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu V. Fu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Nielson JA, Davis JM. Roles for Microglia in Cryptococcal Brain Dissemination in the Zebrafish Larva. Microbiol Spectr 2023; 11:e0431522. [PMID: 36719205 PMCID: PMC10100726 DOI: 10.1128/spectrum.04315-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 02/01/2023] Open
Abstract
Cryptococcal infection begins in the lungs, but yeast cells subsequently access the bloodstream, from which they can reach the central nervous system (CNS). The resulting meningoencephalitis is the most common presentation and is very difficult to treat. How this fungus interacts with the blood-brain barrier (BBB) and establishes growth in the brain parenchyma remains a central question in fungal pathogenesis. We and others have developed the zebrafish larva as a model host for cryptococcosis and demonstrated that hematogenous CNS infection is replicated in this model. Here, we have used this model to examine the details of BBB crossing and the events immediately before and after. We have observed multiple mechanisms of BBB crossing and found that microglia, the resident phagocytes of the brain, likely have multiple roles. First, microglia either actively transfer yeast cells across the BBB or take up a significant proportion of them immediately after crossing. Second, microglia are capable of clearing individual cryptococcal cells at a developmental stage before adaptive immune cells have emerged. Third, microglia serve to maintain endothelial integrity, preventing other, phagocyte-independent forms of crossing. These proposed microglial functions during infection in the zebrafish larva generate new hypotheses concerning the establishment and control of cryptococcal meningoencephalitis. IMPORTANCE Cryptococcal meningitis is a fungal infection of the brain and a major cause of death in people with uncontrolled HIV. Infection begins in the lungs but can enter the bloodstream and disseminate to the brain. A structure called the blood-brain barrier must be crossed for the fungus to enter and cause meningitis. Learning how Cryptococcus crosses the blood-brain barrier will be crucial to understanding and possibly preventing brain infection. Using the zebrafish larva as a model host, we show that microglia, the resident phagocytes of the brain, potentially play multiple previously unappreciated roles in cryptococcal infection of the brain. These roles include reinforcing the integrity of the blood-brain barrier, clearing cryptococcal cells after they have crossed, and possibly participating directly in crossing via a previously unknown mechanism.
Collapse
Affiliation(s)
- Jacquelyn A. Nielson
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - J. Muse Davis
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, USA
| |
Collapse
|
28
|
Chalakova ZP, Johnston SA. Zebrafish Larvae as an Experimental Model of Cryptococcal Meningitis. Methods Mol Biol 2023; 2667:47-69. [PMID: 37145275 DOI: 10.1007/978-1-0716-3199-7_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
This chapter provides guidance for introducing Cryptococcus neoformans into the zebrafish larvae model system to establish a CNS infection phenotype that mimics cryptococcal meningitis as seen in humans. The method outlines techniques for visualizing different stages of pathology development, from initial to severe infection profiles. The chapter provides tips for real time visualization of the interactions between the pathogen and different aspects of the CNS anatomy and immune system.
Collapse
Affiliation(s)
- Z P Chalakova
- University of Sheffield, Firth Court, Western Bank, UK
| | - S A Johnston
- University of Sheffield, Firth Court, Western Bank, UK.
| |
Collapse
|
29
|
Denham ST, Brammer B, Chung KY, Wambaugh MA, Bednarek JM, Guo L, Moreau CT, Brown JCS. A dissemination-prone morphotype enhances extrapulmonary organ entry by Cryptococcus neoformans. Cell Host Microbe 2022; 30:1382-1400.e8. [PMID: 36099922 PMCID: PMC9588642 DOI: 10.1016/j.chom.2022.08.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 01/08/2023]
Abstract
Environmental pathogens move from ecological niches to mammalian hosts, requiring adaptation to dramatically different environments. Microbes that disseminate farther, including the fungal meningitis pathogen Cryptococcus neoformans, require additional adaptation to diverse tissues. We demonstrate that the formation of a small C. neoformans morphotype-called "seed" cells due to their colonizing ability-is critical for extrapulmonary organ entry. Seed cells exhibit changes in fungal cell size and surface expression that result in an enhanced macrophage update. Seed cell formation is triggered by environmental factors, including C. neoformans' environmental niche, and pigeon guano with phosphate plays a central role. Seed cells show the enhanced expression of phosphate acquisition genes, and mutants unable to acquire phosphate fail to adopt the seed cell morphotype. Additionally, phosphate can be released by tissue damage, potentially establishing a feed-forward loop of seed cell formation and dissemination. Thus, C. neoformans' size variation represent inducible morphotypes that change host interactions to facilitate microbe spread.
Collapse
Affiliation(s)
- Steven T Denham
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Brianna Brammer
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Krystal Y Chung
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Morgan A Wambaugh
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph M Bednarek
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Li Guo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Christian T Moreau
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Jessica C S Brown
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
30
|
Jung EH, Park YD, Dragotakes Q, Ramirez LS, Smith DQ, Reis FCG, Dziedzic A, Rodrigues ML, Baker RP, Williamson PR, Jedlicka A, Casadevall A, Coelho C. Cryptococcus neoformans releases proteins during intracellular residence that affect the outcome of the fungal-macrophage interaction. MICROLIFE 2022; 3:uqac015. [PMID: 36247839 PMCID: PMC9552768 DOI: 10.1093/femsml/uqac015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/13/2022] [Indexed: 05/26/2023]
Abstract
Cryptococcus neoformans is a facultative intracellular pathogen that can replicate and disseminate in mammalian macrophages. In this study, we analyzed fungal proteins identified in murine macrophage-like cells after infection with C. neoformans. To accomplish this, we developed a protocol to identify proteins released from cryptococcal cells inside macrophage-like cells; we identified 127 proteins of fungal origin in infected macrophage-like cells. Among the proteins identified was urease, a known virulence factor, and others such as transaldolase and phospholipase D, which have catalytic activities that could contribute to virulence. This method provides a straightforward methodology to study host-pathogen interactions. We chose to study further Yeast Oligomycin Resistance (Yor1), a relatively uncharacterized protein belonging to the large family of ATP binding cassette transporter (ABC transporters). These transporters belong to a large and ancient protein family found in all extant phyla. While ABC transporters have an enormous diversity of functions across varied species, in pathogenic fungi they are better studied as drug efflux pumps. Analysis of C. neoformans yor1Δ strains revealed defects in nonlytic exocytosis, capsule size, and dimensions of extracellular vesicles, when compared to wild-type strains. We detected no difference in growth rates and cell body size. Our results indicate that C. neoformans releases a large suite of proteins during macrophage infection, some of which can modulate fungal virulence and are likely to affect the fungal-macrophage interaction.
Collapse
Affiliation(s)
- Eric H Jung
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Yoon-Dong Park
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Memorial Drive, Bethesda, MD 20814, United States
| | - Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Lia S Ramirez
- Department of Molecular and Cell Biology, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Daniel Q Smith
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Flavia C G Reis
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Rua Professor Algacyr Munhoz Mader, 3775, Curitiba - PR, 81310-020, Brazil
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS), Fundação Oswaldo Cruz Av. Brasil 4036. Room 814, Rio de Janeiro - RJ, 21040-361, Brazil
| | - Amanda Dziedzic
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Marcio L Rodrigues
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Rua Professor Algacyr Munhoz Mader, 3775, Curitiba - PR, 81310-020, Brazil
- Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro, Rio de Janeiro Cidade Universitária da Universidade Federal do Rio de Janeiro,, Rio de Janeiro - RJ, 21941-902, Brazil
| | - Rosanna P Baker
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Memorial Drive, Bethesda, MD 20814, United States
| | - Anne Jedlicka
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Arturo Casadevall
- Corresponding author: Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 N, Wolfe Street, Room E5132, Baltimore, MD 21205, United States. E-mail:
| | - Carolina Coelho
- Corresponding author: Medical Research Council Centre for Medical Mycology at University of Exeter, College of Health and Medicine, Geoffrey Pope Building, Room 325, University of Exeter, Stocker Road, Exeter EX4 4QD, Devon, United Kingdom. E-mail:
| |
Collapse
|
31
|
Ehemann K, Mantilla MJ, Mora-Restrepo F, Rios-Navarro A, Torres M, Celis Ramírez AM. Many ways, one microorganism: Several approaches to study Malassezia in interactions with model hosts. PLoS Pathog 2022; 18:e1010784. [PMID: 36074792 PMCID: PMC9455852 DOI: 10.1371/journal.ppat.1010784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Malassezia, a lipophilic and lipid-dependent yeast, is a microorganism of current interest to mycobiologists because of its role as a commensal or pathogen in health conditions such as dermatological diseases, fungemia, and, as discovered recently, cancer and certain neurological disorders. Various novel approaches in the study of Malassezia have led to increased knowledge of the cellular and molecular mechanisms of this yeast. However, additional efforts are needed for more comprehensive understanding of the behavior of Malassezia in interactions with the host. This article reviews advances useful in the experimental field for Malassezia.
Collapse
Affiliation(s)
- Kevin Ehemann
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - María Juliana Mantilla
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Felipe Mora-Restrepo
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Andrea Rios-Navarro
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Maritza Torres
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Adriana Marcela Celis Ramírez
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
32
|
Ellis PS, Martins RR, Thompson EJ, Farhat A, Renshaw SA, Henriques CM. A subset of gut leukocytes has telomerase-dependent "hyper-long" telomeres and require telomerase for function in zebrafish. Immun Ageing 2022; 19:31. [PMID: 35820929 PMCID: PMC9277892 DOI: 10.1186/s12979-022-00287-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Telomerase, the enzyme capable of elongating telomeres, is usually restricted in human somatic cells, which contributes to progressive telomere shortening with cell-division and ageing. T and B-cells cells are somatic cells that can break this rule and can modulate telomerase expression in a homeostatic manner. Whereas it seems intuitive that an immune cell type that depends on regular proliferation outbursts for function may have evolved to modulate telomerase expression it is less obvious why others may also do so, as has been suggested for macrophages and neutrophils in some chronic inflammation disease settings. The gut has been highlighted as a key modulator of systemic ageing and is a key tissue where inflammation must be carefully controlled to prevent dysfunction. How telomerase may play a role in innate immune subtypes in the context of natural ageing in the gut, however, remains to be determined. RESULTS Using the zebrafish model, we show that subsets of gut immune cells have telomerase-dependent"hyper-long" telomeres, which we identified as being predominantly macrophages and dendritics (mpeg1.1+ and cd45+mhcII+). Notably, mpeg1.1+ macrophages have much longer telomeres in the gut than in their haematopoietic tissue of origin, suggesting that there is modulation of telomerase in these cells, in the gut. Moreover, we show that a subset of gut mpeg1.1+ cells express telomerase (tert) in young WT zebrafish, but that the relative proportion of these cells decreases with ageing. Importantly, this is accompanied by telomere shortening and DNA damage responses with ageing and a telomerase-dependent decrease in expression of autophagy and immune activation markers. Finally, these telomerase-dependent molecular alterations are accompanied by impaired phagocytosis of E. coli and increased gut permeability in vivo. CONCLUSIONS Our data show that limiting levels of telomerase lead to alterations in gut immunity, impacting on the ability to clear pathogens in vivo. These are accompanied by increased gut permeability, which, together, are likely contributors to local and systemic tissue degeneration and increased susceptibility to infection with ageing.
Collapse
Affiliation(s)
- Pam S Ellis
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Raquel R Martins
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Emily J Thompson
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Asma Farhat
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Stephen A Renshaw
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - Catarina M Henriques
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK.
| |
Collapse
|
33
|
Dragotakes Q, Jacobs E, Ramirez LS, Yoon OI, Perez-Stable C, Eden H, Pagnotta J, Vij R, Bergman A, D’Alessio F, Casadevall A. Bet-hedging antimicrobial strategies in macrophage phagosome acidification drive the dynamics of Cryptococcus neoformans intracellular escape mechanisms. PLoS Pathog 2022; 18:e1010697. [PMID: 35816543 PMCID: PMC9302974 DOI: 10.1371/journal.ppat.1010697] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/21/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022] Open
Abstract
The fungus Cryptococcus neoformans is a major human pathogen with a remarkable intracellular survival strategy that includes exiting macrophages through non-lytic exocytosis (Vomocytosis) and transferring between macrophages (Dragotcytosis) by a mechanism that involves sequential events of non-lytic exocytosis and phagocytosis. Vomocytosis and Dragotcytosis are fungal driven processes, but their triggers are not understood. We hypothesized that the dynamics of Dragotcytosis could inherit the stochasticity of phagolysosome acidification and that Dragotcytosis was triggered by fungal cell stress. Consistent with this view, fungal cells involved in Dragotcytosis reside in phagolysosomes characterized by low pH and/or high oxidative stress. Using fluorescent microscopy, qPCR, live cell video microscopy, and fungal growth assays we found that the that mitigating pH or oxidative stress reduced Dragotcytosis frequency, whereas ROS susceptible mutants of C. neoformans underwent Dragotcytosis more frequently. Dragotcytosis initiation was linked to phagolysosomal pH, oxidative stresses, and macrophage polarization state. Dragotcytosis manifested stochastic dynamics thus paralleling the dynamics of phagosomal acidification, which correlated with the inhospitality of phagolysosomes in differently polarized macrophages. Hence, randomness in phagosomal acidification randomly created a population of inhospitable phagosomes where fungal cell stress triggered stochastic C. neoformans non-lytic exocytosis dynamics to escape a non-permissive intracellular macrophage environment.
Collapse
Affiliation(s)
- Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ella Jacobs
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Lia Sanchez Ramirez
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Olivia Insun Yoon
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Caitlin Perez-Stable
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Hope Eden
- Department of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jenlu Pagnotta
- Department of Molecular and Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Raghav Vij
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Aviv Bergman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York City, New York, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | - Franco D’Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
34
|
Wirth F, Staudt KJ, Araújo BV, Ishida K. Experimental models for pharmacokinetic and pharmacodynamic studies of antifungals used in cryptococcosis treatment. Future Microbiol 2022; 17:969-982. [PMID: 35694892 DOI: 10.2217/fmb-2021-0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Studies on cryptococcosis in the mammal animal model have demonstrated the occurrence of central nervous system infection and similarities in fungal pathogenicity with clinical and immunological features of the human infection. Although there is still a lack of studies involving pharmacokinetics (PK) and pharmacodynamics (PD) in animal models of cryptococcosis in the literature, these experimental models are useful for understanding this mycosis and antifungal effectiveness in improving the therapeutic schemes. The scope of this review is to describe and discuss the main mammal animal models for PK and PD studies of antifungals used in cryptococcosis treatment. Alternative models and computational methods are also addressed. All approaches for PK/PD studies are relevant to investigating drug-infection interaction and improving cryptococcosis therapy.
Collapse
Affiliation(s)
- Fernanda Wirth
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Keli J Staudt
- Faculty of Pharmacy, Pharmaceutical Sciences Post-Graduation Program, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90610-000, Brazil
| | - Bibiana V Araújo
- Faculty of Pharmacy, Pharmaceutical Sciences Post-Graduation Program, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90610-000, Brazil
| | - Kelly Ishida
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| |
Collapse
|
35
|
Stream A, Madigan CA. Zebrafish: an underutilized tool for discovery in host-microbe interactions. Trends Immunol 2022; 43:426-437. [PMID: 35527182 PMCID: PMC11302990 DOI: 10.1016/j.it.2022.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 12/31/2022]
Abstract
Zebrafish are relatively new to the field of host-pathogen interactions, although they have been a valuable vertebrate model for decades in developmental biology and neuroscience. Transparent zebrafish larvae have most components of the human innate immune system, and adult zebrafish also produce cells of the adaptive immune system. Recent discoveries using zebrafish infection models include mechanisms of pathogen survival and host cell sensing of microbes. These discoveries were enabled by zebrafish technology, which is constantly evolving and providing new opportunities for immunobiology research. Recent tools include CRISPR/Cas9 mutagenesis, in vivo biotinylation, and genetically encoded biosensors. We argue that the zebrafish model - which remains underutilized in immunology - provides fertile ground for a new understanding of host-microbe interactions in a transparent host.
Collapse
Affiliation(s)
- Alexandra Stream
- Department of Biological Sciences, University of California San Diego (UCSD), San Diego, CA, USA
| | - Cressida A Madigan
- Department of Biological Sciences, University of California San Diego (UCSD), San Diego, CA, USA.
| |
Collapse
|
36
|
Goodwin N, Westall L. Design and Husbandry Considerations for a Containment Level 2 Aquatic Facility. Zebrafish 2022; 19:56-66. [PMID: 35439044 PMCID: PMC9070741 DOI: 10.1089/zeb.2021.0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The greatly increased use of aquatic species to study disease over the past 20 years necessitates understanding their husbandry and housing requirements to optimize research and welfare and to ensure compliance with regulations. To achieve these goals, aquatic systems have expanded from pet shop and home aquaria to research-grade systems incorporating designs and features to increase their robustness, practicality, and flexibility. Moreover, these last decades have seen the increasing use of aquatic animals for infectious disease research using containment level 2 (CL2)/biosafety level 2 pathogens. In this study, we discuss the facility design requirements and modifications, which must be considered for the planning, construction, and use of an aquatic facility for zebrafish infected with CL2 pathogens. These include decontamination of water and equipment, racking and filtration design, personal protective equipment, and husbandry procedures. This guidance is based on our experience in the design and ongoing management of such facilities.
Collapse
Affiliation(s)
- Nicola Goodwin
- University Biomedical Services, University of Cambridge, Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Lynda Westall
- Research Support Facility, Wellcome Trust Sanger Institute, Cambridge, United Kingdom of Great Britain and Northern Ireland, Retired
| |
Collapse
|
37
|
Rutherford HA, Clarke A, Chambers EV, Petts JJ, Carson EG, Isles HM, Duque-Jaramillo A, Renshaw SA, Levraud JP, Hamilton N. A zebrafish reporter line reveals immune and neuronal expression of endogenous retrovirus. Dis Model Mech 2022; 15:dmm048921. [PMID: 35142349 PMCID: PMC9016899 DOI: 10.1242/dmm.048921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/01/2022] [Indexed: 01/12/2023] Open
Abstract
Endogenous retroviruses (ERVs) are fossils left in our genome from retrovirus infections of the past. Their sequences are part of every vertebrate genome and their random integrations are thought to have contributed to evolution. Although ERVs are mainly silenced by the host genome, they have been found to be activated in multiple disease states, such as auto-inflammatory disorders and neurological diseases. However, the numerous copies in mammalian genomes and the lack of tools to study them make defining their role in health and diseases challenging. In this study, we identified eight copies of the zebrafish endogenous retrovirus zferv. We created and characterised the first in vivo ERV reporter line in any species. Using a combination of live imaging, flow cytometry and single-cell RNA sequencing, we mapped zferv expression to early T cells and neurons. Thus, this new tool identified tissues expressing ERV in zebrafish, highlighting a potential role of ERV during brain development and strengthening the hypothesis that ERV play a role in immunity and neurological diseases. This transgenic line is therefore a suitable tool to study the function of ERV in health and diseases.
Collapse
Affiliation(s)
- Holly A. Rutherford
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Amy Clarke
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Emily V. Chambers
- The Bioinformatics Core, Faculty of Medicine and Dentistry, University of Sheffield, Sheffield S10 2TN, UK
| | - Jessica J. Petts
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Euan G. Carson
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Hannah M. Isles
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Alejandra Duque-Jaramillo
- Institute of Microbiology (IMUL), Lausanne University Hospital and University of Lausanne, Rue du Bugnon 48, 1011 Lausanne, Switzerland
| | - Stephen A. Renshaw
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Jean-Pierre Levraud
- Macrophages et Développement de l'Immunité, Institut Pasteur, CNRS UMR3738, 25 Rue du Docteur Roux, 75015 Paris,France
| | - Noémie Hamilton
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
- The Institute of Neuroscience, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
38
|
Gibson JF, Bojarczuk A, Evans RJ, Kamuyango AA, Hotham R, Lagendijk AK, Hogan BM, Ingham PW, Renshaw SA, Johnston SA. Blood vessel occlusion by Cryptococcus neoformans is a mechanism for haemorrhagic dissemination of infection. PLoS Pathog 2022; 18:e1010389. [PMID: 35446924 PMCID: PMC9022829 DOI: 10.1371/journal.ppat.1010389] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/21/2022] [Indexed: 11/18/2022] Open
Abstract
Meningitis caused by infectious pathogens is associated with vessel damage and infarct formation, however the physiological cause is often unknown. Cryptococcus neoformans is a human fungal pathogen and causative agent of cryptococcal meningitis, where vascular events are observed in up to 30% of patients, predominantly in severe infection. Therefore, we aimed to investigate how infection may lead to vessel damage and associated pathogen dissemination using a zebrafish model that permitted noninvasive in vivo imaging. We find that cryptococcal cells become trapped within the vasculature (dependent on their size) and proliferate there resulting in vasodilation. Localised cryptococcal growth, originating from a small number of cryptococcal cells in the vasculature was associated with sites of dissemination and simultaneously with loss of blood vessel integrity. Using a cell-cell junction tension reporter we identified dissemination from intact blood vessels and where vessel rupture occurred. Finally, we manipulated blood vessel tension via cell junctions and found increased tension resulted in increased dissemination. Our data suggest that global vascular vasodilation occurs following infection, resulting in increased vessel tension which subsequently increases dissemination events, representing a positive feedback loop. Thus, we identify a mechanism for blood vessel damage during cryptococcal infection that may represent a cause of vascular damage and cortical infarction during cryptococcal meningitis. Meningitis is a life threatening form of infection in the brain that is difficult to treat. How infection spreads from the blood to cause meningitis is not well understood. Here we have shown how infection with the fungus Cryptococcus neoformans can be spread from the blood by blocking and bursting blood vessels. Using zebrafish larvae, we were able to follow the same infections over a period of days to understand how this infection behaves in blood vessels. We found that fungal cells become stuck within blood vessels depending on their size. These cells grow within blood vessels, resulting in the blood vessels becoming wider. We measured increased tension in blood vessels suggesting that, with the bloackage and widening of vessels, there was increased local blood pressure. We found that vessel blockage was associated with their rupture and spreading of fungus into the surround tissue. Finally, by increasing the tension in vessels we could increase the number of blood bursting events supporting our conclusion that blood vessel blockage leads to the spread of the infection outside of blood vessels.
Collapse
Affiliation(s)
- Josie F. Gibson
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-Star), Singapore
| | - Aleksandra Bojarczuk
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Robert J. Evans
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
| | - Alfred Alinafe Kamuyango
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
| | - Richard Hotham
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
| | - Anne K. Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Benjamin M. Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Philip W. Ingham
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-Star), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
| | - Simon A. Johnston
- Department of Infection, Immunity and Cardiovascular disease, Bateson Centre and Florey Institute, University of Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Lineages Derived from Cryptococcus neoformans Type Strain H99 Support a Link between the Capacity to Be Pleomorphic and Virulence. mBio 2022; 13:e0028322. [PMID: 35258331 PMCID: PMC9040854 DOI: 10.1128/mbio.00283-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The pathogenic yeast Cryptococcus neoformans causes nearly 200,000 deaths annually in immunocompromised individuals. Cryptococcus cells can undergo substantial morphological change during mammalian infection, including increased capsule and cell size, the release of shed capsule, and the production of titan (>10 μm), micro (<2 μm)-, and irregular cells. We examined phenotypic variation under conditions designed to simulate in vivo stress in a collection of nine lineages derived from the C. neoformans type strain H99. These lineages are highly genetically similar but have a range of virulence levels. Strains from hypervirulent lineages had a larger average capsule size, greater variation in cell size, and an increased production of microcells and shed capsule. We tested whether disruption of SGF29, which encodes a component of the SAGA histone acetylation complex that has previously been implicated in the hypervirulence of some lineages, also has a role in the production of morphological variants. Deletion of SGF29 in a lineage with intermediate virulence substantially increased its production of microcells and released capsule, consistent with a switch to hypervirulence. We further examined SGF29 in a set of 52 clinical isolates and found loss-of-function mutations were significantly correlated with patient death. Expansion of a TA repeat in the second intron of SGF29 was positively correlated with cell and capsule size, suggesting it also affects Sgf29 function. This study extends the evidence for a link between pleomorphism and virulence in Cryptococcus, with a likely role for epigenetic mechanisms mediated by SAGA-induced histone acetylation.
Collapse
|
40
|
Mohamed SH, Nyazika TK, Ssebambulidde K, Lionakis MS, Meya DB, Drummond RA. Fungal CNS Infections in Africa: The Neuroimmunology of Cryptococcal Meningitis. Front Immunol 2022; 13:804674. [PMID: 35432326 PMCID: PMC9010970 DOI: 10.3389/fimmu.2022.804674] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/03/2022] [Indexed: 01/13/2023] Open
Abstract
Cryptococcal meningitis (CM) is the leading cause of central nervous system (CNS) fungal infections in humans, with the majority of cases reported from the African continent. This is partly due to the high burden of HIV infection in the region and reduced access to standard-of-care including optimal sterilising antifungal drug treatments. As such, CM is responsible for 10-15% of all HIV-related mortality, with a large proportion being preventable. Immunity to the causative agent of CM, Cryptococcus neoformans, is only partially understood. IFNγ producing CD4+ T-cells are required for the activation of myeloid cells, especially macrophages, to enable fungal killing and clearance. However, macrophages may also act as a reservoir of the fungal yeast cells, shielding them from host immune detection thus promoting latent infection or persistent chronic inflammation. In this chapter, we review the epidemiology and pathogenesis of CNS fungal infections in Africa, with a major focus on CM, and the antifungal immune pathways operating to protect against C. neoformans infection. We also highlight the areas of research and policy that require prioritisation to help reduce the burden of CNS fungal diseases in Africa.
Collapse
Affiliation(s)
- Sally H Mohamed
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Tinashe K Nyazika
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kenneth Ssebambulidde
- College of Health Sciences, Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - David B Meya
- College of Health Sciences, Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Rebecca A Drummond
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Institute of Microbiology & Infection, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
41
|
Miskolci V, Tweed KE, Lasarev MR, Britt EC, Walsh AJ, Zimmerman LJ, McDougal CE, Cronan MR, Fan J, Sauer JD, Skala MC, Huttenlocher A. In vivo fluorescence lifetime imaging of macrophage intracellular metabolism during wound responses in zebrafish. eLife 2022; 11:66080. [PMID: 35200139 PMCID: PMC8871371 DOI: 10.7554/elife.66080] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
The function of macrophages in vitro is linked to their metabolic rewiring. However, macrophage metabolism remains poorly characterized in situ. Here, we used two-photon intensity and lifetime imaging of autofluorescent metabolic coenzymes, nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) and flavin adenine dinucleotide (FAD), to assess the metabolism of macrophages in the wound microenvironment. Inhibiting glycolysis reduced NAD(P)H mean lifetime and made the intracellular redox state of macrophages more oxidized, as indicated by reduced optical redox ratio. We found that TNFα+ macrophages had lower NAD(P)H mean lifetime and were more oxidized compared to TNFα- macrophages. Both infection and thermal injury induced a macrophage population with a more oxidized redox state in wounded tissues. Kinetic analysis detected temporal changes in the optical redox ratio during tissue repair, revealing a shift toward a more reduced redox state over time. Metformin reduced TNFα+ wound macrophages, made intracellular redox state more reduced and improved tissue repair. By contrast, depletion of STAT6 increased TNFα+ wound macrophages, made redox state more oxidized and impaired regeneration. Our findings suggest that autofluorescence of NAD(P)H and FAD is sensitive to dynamic changes in intracellular metabolism in tissues and can be used to probe the temporal and spatial regulation of macrophage metabolism during tissue damage and repair.
Collapse
Affiliation(s)
- Veronika Miskolci
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Kelsey E Tweed
- Morgridge Institute for ResearchMadisonUnited States,Department of Biomedical Engineering, University of Wisconsin-MadisonMadisonUnited States
| | - Michael R Lasarev
- Department of Biostatistics & Medical Informatics, University of Wisconsin-MadisonMadisonUnited States
| | - Emily C Britt
- Morgridge Institute for ResearchMadisonUnited States,Department of Nutritional Sciences, University of Wisconsin-MadisonMadisonUnited States
| | - Alex J Walsh
- Morgridge Institute for ResearchMadisonUnited States
| | - Landon J Zimmerman
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Courtney E McDougal
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
| | - Jing Fan
- Morgridge Institute for ResearchMadisonUnited States,Department of Nutritional Sciences, University of Wisconsin-MadisonMadisonUnited States
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Melissa C Skala
- Morgridge Institute for ResearchMadisonUnited States,Department of Biomedical Engineering, University of Wisconsin-MadisonMadisonUnited States
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States,Department of Pediatrics, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
42
|
The Antimicrobial Peptide MK58911-NH 2 Acts on Planktonic, Biofilm, and Intramacrophage Cells of Cryptococcus neoformans. Antimicrob Agents Chemother 2021; 65:e0090421. [PMID: 34516241 PMCID: PMC8597745 DOI: 10.1128/aac.00904-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cryptococcosis is associated with high rates of morbidity and mortality, especially in AIDS patients. Its treatment is carried out by combining amphotericin B and azoles or flucytosine, which causes unavoidable toxicity issues in the host. Thus, the urgency in obtaining new antifungals drives the search for antimicrobial peptides (AMPs). This study aimed to extend the understanding of the mechanism of action of an AMP analog from wasp peptide toxins, MK58911-NH2, on Cryptococcus neoformans. We also evaluated if MK58911-NH2 can act on cryptococcal cells in macrophages, biofilms, and an immersion zebrafish model of infection. Finally, we investigated the structure-antifungal action and the toxicity relationship of MK58911-NH2 fragments and a derivative of this peptide (MH58911-NH2). The results demonstrated that MK58911-NH2 did not alter the fluorescence intensity of the cell wall-binding dye calcofluor white or the capsule-binding dye 18b7 antibody-fluorescein isothiocyanate (FITC) in C. neoformans but rather reduced the number and size of fungal cells. This activity reduced the fungal burden of C. neoformans in both macrophages and zebrafish embryos as well as within biofilms. Three fragments of the MK58911-NH2 peptide showed no activity against Cryptococcus and not toxicity in lung cells. The derivative peptide MH58911-NH2, in which the lysine residues of MK58911-NH2 were replaced by histidines, reduced the activity against extracellular and intracellular C. neoformans. On the other hand, it was active against biofilms and showed reduced toxicity. In summary, these results showed that peptide MK58911-NH2 could be a promising agent against cryptococcosis. This work also opens a perspective for the verification of the antifungal activity of other derivatives.
Collapse
|
43
|
Gibson JF, Pidwill GR, Carnell OT, Surewaard BGJ, Shamarina D, Sutton JAF, Jeffery C, Derré-Bobillot A, Archambaud C, Siggins MK, Pollitt EJG, Johnston SA, Serror P, Sriskandan S, Renshaw SA, Foster SJ. Commensal bacteria augment Staphylococcus aureus infection by inactivation of phagocyte-derived reactive oxygen species. PLoS Pathog 2021; 17:e1009880. [PMID: 34529737 PMCID: PMC8478205 DOI: 10.1371/journal.ppat.1009880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/28/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a human commensal organism and opportunist pathogen, causing potentially fatal disease. The presence of non-pathogenic microflora or their components, at the point of infection, dramatically increases S. aureus pathogenicity, a process termed augmentation. Augmentation is associated with macrophage interaction but by a hitherto unknown mechanism. Here, we demonstrate a breadth of cross-kingdom microorganisms can augment S. aureus disease and that pathogenesis of Enterococcus faecalis can also be augmented. Co-administration of augmenting material also forms an efficacious vaccine model for S. aureus. In vitro, augmenting material protects S. aureus directly from reactive oxygen species (ROS), which correlates with in vivo studies where augmentation restores full virulence to the ROS-susceptible, attenuated mutant katA ahpC. At the cellular level, augmentation increases bacterial survival within macrophages via amelioration of ROS, leading to proliferation and escape. We have defined the molecular basis for augmentation that represents an important aspect of the initiation of infection. S. aureus is a commensal inhabitant of the human skin and nares. However, it can cause serious diseases if it is able to breach our protective barriers such as the skin, often via wounds or surgery. If infection occurs via a wound, this initial inoculum contains both the pathogen, other members of the microflora and also wider environmental microbes. We have previously described “augmentation”, whereby this other non-pathogenic material can enhance the ability of S. aureus to lead to a serious disease outcome. Here we have determined the breadth of augmenting material and elucidated the cellular and molecular basis for its activity. Augmentation occurs via shielding of S. aureus from the direct bactericidal effects of reactive oxygen species produced by macrophages. This initial protection enables the effective establishment of S. aureus infection. Understanding augmentation not only explains an important facet of the interaction of S. aureus with our innate immune system, but also provides a platform for the development of novel prophylaxis approaches.
Collapse
Affiliation(s)
- Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Oliver T. Carnell
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Bas G. J. Surewaard
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daria Shamarina
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Joshua A. F. Sutton
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Charlotte Jeffery
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | | | - Cristel Archambaud
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Matthew K. Siggins
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Eric J. G. Pollitt
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Simon A. Johnston
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Pascale Serror
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- * E-mail: (SAR); (SJF)
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- * E-mail: (SAR); (SJF)
| |
Collapse
|
44
|
|
45
|
The Environmental Effects on Virulence Factors and the Antifungal Susceptibility of Cryptococcus neoformans. Int J Mol Sci 2021; 22:ijms22126302. [PMID: 34208294 PMCID: PMC8230809 DOI: 10.3390/ijms22126302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 01/10/2023] Open
Abstract
Cryptococcus neoformans is a facultative intracellular pathogen responsible for fungal meningoencephalitis primarily in immunocompromised individuals. It has become evident the pathogenicity of C. neoformans is dependent on the fungal cell’s environment. The differential expression of virulence factors, based on the cell’s environmental conditions, is one mechanism allowing for the environmental control of the pathogenic ability of C. neoformans. Here, we discuss how these virulence factors (including melanin, the polysaccharide capsule, and Antiphagocytic protein 1) have been shown to be differentially expressed dependent on the cell’s environment. The genetics and signaling pathways leading to the environmental-dependent regulation of virulence factors will also be examined. Susceptibility to antifungal therapeutics is also regulated by the environment, and thus affects the pathogenic abilities of C. neoformans and disease outcomes. This review will also examine the role of the C. neoformans’s environment on antifungal susceptibilities, and the genetics and signaling pathways responsible for these susceptibility alterations. By examining the complex interplay between the environment and the pathogenicity of C. neoformans, we have a better understanding of the intricacies of the pathogen–environment interaction and how to exploit this interaction to develop the most effective treatment protocols.
Collapse
|
46
|
Jacobovitz MR, Rupp S, Voss PA, Maegele I, Gornik SG, Guse A. Dinoflagellate symbionts escape vomocytosis by host cell immune suppression. Nat Microbiol 2021; 6:769-782. [PMID: 33927382 PMCID: PMC7611106 DOI: 10.1038/s41564-021-00897-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/25/2021] [Indexed: 02/02/2023]
Abstract
Alveolata comprises diverse taxa of single-celled eukaryotes, many of which are renowned for their ability to live inside animal cells. Notable examples are apicomplexan parasites and dinoflagellate symbionts, the latter of which power coral reef ecosystems. Although functionally distinct, they evolved from a common, free-living ancestor and must evade their host's immune response for persistence. Both the initial cellular events that gave rise to this intracellular lifestyle and the role of host immune modulation in coral-dinoflagellate endosymbiosis are poorly understood. Here, we use a comparative approach in the cnidarian endosymbiosis model Aiptasia, which re-establishes endosymbiosis with free-living dinoflagellates every generation. We find that uptake of microalgae is largely indiscriminate, but non-symbiotic microalgae are expelled by vomocytosis, while symbionts induce host cell innate immune suppression and form a lysosomal-associated membrane protein 1-positive niche. We demonstrate that exogenous immune stimulation results in symbiont expulsion and, conversely, inhibition of canonical Toll-like receptor signalling enhances infection of host animals. Our findings indicate that symbiosis establishment is dictated by local innate immune suppression, to circumvent expulsion and promote niche formation. This work provides insight into the evolution of the cellular immune response and key steps involved in mediating endosymbiotic interactions.
Collapse
Affiliation(s)
- Marie R Jacobovitz
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Sebastian Rupp
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Philipp A Voss
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Ira Maegele
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Sebastian G Gornik
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Annika Guse
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
47
|
van Rhijn N, Bromley M. The Consequences of Our Changing Environment on Life Threatening and Debilitating Fungal Diseases in Humans. J Fungi (Basel) 2021; 7:367. [PMID: 34067211 PMCID: PMC8151111 DOI: 10.3390/jof7050367] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/16/2022] Open
Abstract
Human activities have significantly impacted the environment and are changing our climate in ways that will have major consequences for ourselves, and endanger animal, plant and microbial life on Earth. Rising global temperatures and pollution have been highlighted as potential drivers for increases in infectious diseases. Although infrequently highlighted, fungi are amongst the leading causes of infectious disease mortality, resulting in more than 1.5 million deaths every year. In this review we evaluate the evidence linking anthropomorphic impacts with changing epidemiology of fungal disease. We highlight how the geographic footprint of endemic mycosis has expanded, how populations susceptible to fungal infection and fungal allergy may increase and how climate change may select for pathogenic traits and indirectly contribute to the emergence of drug resistance.
Collapse
Affiliation(s)
| | - Michael Bromley
- Manchester Fungal Infection Group, University of Manchester, Manchester M13 9PL, UK;
| |
Collapse
|
48
|
Strickland AB, Shi M. Mechanisms of fungal dissemination. Cell Mol Life Sci 2021; 78:3219-3238. [PMID: 33449153 PMCID: PMC8044058 DOI: 10.1007/s00018-020-03736-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/23/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022]
Abstract
Fungal infections are an increasing threat to global public health. There are more than six million fungal species worldwide, but less than 1% are known to infect humans. Most of these fungal infections are superficial, affecting the hair, skin and nails, but some species are capable of causing life-threatening diseases. The most common of these include Cryptococcus neoformans, Aspergillus fumigatus and Candida albicans. These fungi are typically innocuous and even constitute a part of the human microbiome, but if these pathogens disseminate throughout the body, they can cause fatal infections which account for more than one million deaths worldwide each year. Thus, systemic dissemination of fungi is a critical step in the development of these deadly infections. In this review, we discuss our current understanding of how fungi disseminate from the initial infection sites to the bloodstream, how immune cells eliminate fungi from circulation and how fungi leave the blood and enter distant organs, highlighting some recent advances and offering some perspectives on future directions.
Collapse
Affiliation(s)
- Ashley B Strickland
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, USA.
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, USA.
| |
Collapse
|
49
|
Prajsnar TK, Serba JJ, Dekker BM, Gibson JF, Masud S, Fleming A, Johnston SA, Renshaw SA, Meijer AH. The autophagic response to Staphylococcus aureus provides an intracellular niche in neutrophils. Autophagy 2021; 17:888-902. [PMID: 32174246 PMCID: PMC8078660 DOI: 10.1080/15548627.2020.1739443] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 02/17/2020] [Accepted: 02/28/2020] [Indexed: 11/22/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen causing multiple pathologies, from cutaneous lesions to life-threatening sepsis. Although neutrophils contribute to immunity against S. aureus, multiple lines of evidence suggest that these phagocytes can provide an intracellular niche for staphylococcal dissemination. However, the mechanism of neutrophil subversion by intracellular S. aureus remains unknown. Targeting of intracellular pathogens by macroautophagy/autophagy is recognized as an important component of host innate immunity, but whether autophagy is beneficial or detrimental to S. aureus-infected hosts remains controversial. Here, using larval zebrafish, we showed that the autophagy marker Lc3 rapidly decorates S. aureus following engulfment by macrophages and neutrophils. Upon phagocytosis by neutrophils, Lc3-positive, non-acidified spacious phagosomes are formed. This response is dependent on phagocyte NADPH oxidase as both cyba/p22phox knockdown and diphenyleneiodonium (DPI) treatment inhibited Lc3 decoration of phagosomes. Importantly, NADPH oxidase inhibition diverted neutrophil S. aureus processing into tight acidified vesicles, which resulted in increased host resistance to the infection. Some intracellular bacteria within neutrophils were also tagged by Sqstm1/p62-GFP fusion protein and loss of Sqstm1 impaired host defense. Together, we have shown that intracellular handling of S. aureus by neutrophils is best explained by Lc3-associated phagocytosis (LAP), which appears to provide an intracellular niche for bacterial pathogenesis, while the selective autophagy receptor Sqstm1 is host-protective. The antagonistic roles of LAP and Sqstm1-mediated pathways in S. aureus-infected neutrophils may explain the conflicting reports relating to anti-staphylococcal autophagy and provide new insights for therapeutic strategies against antimicrobial-resistant Staphylococci.Abbreviations: ATG: autophagy related; CFU: colony-forming units; CMV: cytomegalovirus; Cyba/P22phox: cytochrome b-245, alpha polypeptide; DMSO: dimethyl sulfoxide; DPI: diphenyleneiodonium; EGFP: enhanced green fluorescent protein; GFP: green fluorescent protein; hpf: hours post-fertilization; hpi: hours post-infection; Irf8: interferon regulatory factor 8; LAP: LC3-associated phagocytosis; lyz: lysozyme; LWT: london wild type; Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase; RFP: red fluorescent protein; ROS: reactive oxygen species; RT-PCR: reverse transcriptase polymerase chain reaction; Sqstm1/p62: sequestosome 1; Tg: transgenic; TSA: tyramide signal amplification.
Collapse
Affiliation(s)
- Tomasz K. Prajsnar
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Justyna J. Serba
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Bernice M. Dekker
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Josie F. Gibson
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Krebs Institute and Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Samrah Masud
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Angeleen Fleming
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Simon A. Johnston
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Stephen A. Renshaw
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Annemarie H. Meijer
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| |
Collapse
|
50
|
Role of Extracellular Mycobacteria in Blood-Retinal Barrier Invasion in a Zebrafish Model of Ocular TB. Pathogens 2021; 10:pathogens10030333. [PMID: 33805720 PMCID: PMC7999171 DOI: 10.3390/pathogens10030333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 12/30/2022] Open
Abstract
Intraocular inflammation following mycobacterial dissemination to the eye is common in tuberculosis (TB)-endemic countries. However, the early host–pathogen interactions during ocular dissemination are unknown. In this study, we investigated the early events during mycobacterial invasion of the blood-retinal barriers (BRBs) with fluorescent-tagged Mycobacterium marinum (Mm), host macrophages, and retinal vasculature in a zebrafish model of ocular TB. We found that Mm invaded the vascular endothelium in either the extracellular or intracellular (inside phagocytes) state, typically 3–4 days post-injection (dpi). Extracellular Mm are phagocytosed in the retinal tissue and progress to form a compact granuloma around 6 dpi. Intracellular Mm crossing the BRBs are likely to be less virulent and either persist inside solitary macrophages (in most cases) or progress to loosely arranged granuloma (rarely). The early interactions between mycobacteria and host immune cells can thus determine the course of disease during mycobacterial dissemination to the eye.
Collapse
|