1
|
Baker RP, Liu AZ, Casadevall A. Cell wall melanin impedes growth of the Cryptococcus neoformans polysaccharide capsule by sequestering calcium. Proc Natl Acad Sci U S A 2024; 121:e2412534121. [PMID: 39259590 PMCID: PMC11420191 DOI: 10.1073/pnas.2412534121] [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: 06/21/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
Cryptococcus neoformans has emerged as a frontrunner among deadly fungal pathogens and is particularly life-threatening for many HIV-infected individuals with compromised immunity. Multiple virulence factors contribute to the growth and survival of C. neoformans within the human host, the two most prominent of which are the polysaccharide capsule and melanin. As both of these features are associated with the cell wall, we were interested to explore possible cooperative or competitive interactions between these two virulence factors. Whereas capsule thickness had no effect on the rate at which cells became melanized, build-up of the melanin pigment layer resulted in a concomitant loss of polysaccharide material, leaving melanized cells with significantly thinner capsules than their nonmelanized counterparts. When melanin was provided exogenously to cells in a transwell culture system we observed a similar inhibition of capsule growth and maintenance. Our results show that melanin sequesters calcium thereby limiting its availability to form divalent bridges between polysaccharide subunits required for outer capsule assembly. The decreased ability of melanized cells to incorporate exported polysaccharide into the growing capsule correlated with the amount of shed polysaccharide, which could have profound negative impacts on the host immune response.
Collapse
Affiliation(s)
- Rosanna P. Baker
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Amy Z. Liu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| |
Collapse
|
2
|
Ball B, Sukumaran A, Krieger JR, Geddes-McAlister J. Comparative Cross-Kingdom DDA- and DIA-PASEF Proteomic Profiling Reveals Novel Determinants of Fungal Virulence and a Putative Druggable Target. J Proteome Res 2024; 23:3917-3932. [PMID: 39140824 PMCID: PMC11385706 DOI: 10.1021/acs.jproteome.4c00255] [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: 08/15/2024]
Abstract
Accurate and reliable detection of fungal pathogens presents an important hurdle to manage infections, especially considering that fungal pathogens, including the globally important human pathogen, Cryptococcus neoformans, have adapted diverse mechanisms to survive the hostile host environment and moderate virulence determinant production during coinfections. These pathogen adaptations present an opportunity for improvements (e.g., technological and computational) to better understand the interplay between a host and a pathogen during disease to uncover new strategies to overcome infection. In this study, we performed comparative proteomic profiling of an in vitro coinfection model across a range of fungal and bacterial burden loads in macrophages. Comparing data-dependent acquisition and data-independent acquisition enabled with parallel accumulation serial fragmentation technology, we quantified changes in dual-perspective proteome remodeling. We report enhanced and novel detection of pathogen proteins with data-independent acquisition-parallel accumulation serial fragmentation (DIA-PASEF), especially for fungal proteins during single and dual infection of macrophages. Further characterization of a fungal protein detected only with DIA-PASEF uncovered a novel determinant of fungal virulence, including altered capsule and melanin production, thermotolerance, and macrophage infectivity, supporting proteomics advances for the discovery of a novel putative druggable target to suppress C. neoformans pathogenicity.
Collapse
Affiliation(s)
- Brianna Ball
- Department of Molecular and Cellular Biology, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | - Arjun Sukumaran
- Department of Molecular and Cellular Biology, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | | | | |
Collapse
|
3
|
Guerrini V, Prideaux B, Khan R, Subbian S, Wang Y, Sadimin E, Pawar S, Ukey R, Singer EA, Xue C, Gennaro ML. Cryptococcosis, tuberculosis, and a kidney cancer fail to fit the atherosclerosis paradigm for foam cell lipid content. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.08.542766. [PMID: 37333211 PMCID: PMC10274805 DOI: 10.1101/2023.06.08.542766] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Foam cells are dysfunctional, lipid-laden macrophages associated with chronic inflammation of diverse origin. The long-standing paradigm that foam cells are cholesterol-laden derives from atherosclerosis research. We previously showed that, in tuberculosis, foam cells surprisingly accumulate triglycerides. Here, we utilized bacterial ( Mycobacterium tuberculosis ), fungal ( Cryptococcus neoformans ), and human papillary renal cell carcinoma (pRCC) models to address the need for a new explanation of foam cell biogenesis. We applied mass spectrometry-based imaging to assess the spatial distribution of storage lipids relative to foam-cell-rich areas in lesional tissues, and we characterized lipid-laden macrophages generated under corresponding in vitro conditions. The in vivo data and the in vitro findings showed that cryptococcus-infected macrophages accumulate triglycerides, while macrophages exposed to pRCC- conditioned-medium accumulated both triglycerides and cholesterol. Moreover, cryptococcus- and mycobacterium-infected macrophages accumulated triglycerides in different ways. Collectively, the data show that the molecular events underlying foam cell formation are specific to disease and microenvironment. Since foam cells are potential therapeutic targets, recognizing that their formation is disease-specific opens new biomedical research directions.
Collapse
|
4
|
Baker RP, Liu AZ, Casadevall A. Cell wall melanin impedes growth of the Cryptococcus neoformans polysaccharide capsule by sequestering calcium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599928. [PMID: 38948764 PMCID: PMC11212976 DOI: 10.1101/2024.06.20.599928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Cryptococcus neoformans has emerged as a frontrunner among deadly fungal pathogens and is particularly life-threatening for many HIV-infected individuals with compromised immunity. Multiple virulence factors contribute to the growth and survival of C. neoformans within the human host, the two most prominent of which are the polysaccharide capsule and melanin. As both of these features are associated with the cell wall, we were interested to explore possible cooperative or competitive interactions between these two virulence factors. Whereas capsule thickness had no effect on the rate at which cells became melanized, build-up of the melanin pigment layer resulted in a concomitant loss of polysaccharide material, leaving melanized cells with significantly thinner capsules than their non-melanized counterparts. When melanin was provided exogenously to cells in a transwell culture system we observed a similar inhibition of capsule growth and maintenance. Our results show that melanin sequesters calcium thereby limiting its availability to form divalent bridges between polysaccharide subunits required for outer capsule assembly. The decreased ability of melanized cells to incorporate exported polysaccharide into the growing capsule correlated with the amount of shed polysaccharide, which could have profound negative impacts on the host immune response. Significance Statement Cryptococcus neoformans is an opportunistic fungal pathogen that presents a significant health risk for immunocompromised individuals. We report an interaction between the two major cryptococcal virulence factors, the polysaccharide capsule and melanin. Melanin impacted the growth and maintenance of the polysaccharide capsule, resulting in loss of capsular material during melanization. Our results suggest that melanin can act as a sink for calcium, thereby limiting its availability to form ionic bridges between polysaccharide chains on the growing surface of the outer capsule. As polysaccharide is continuously exported to support capsule growth, failure of melanized cells to incorporate this material results in a higher concentration of shed polysaccharide in the extracellular milieu, which is expected to interfere with host immunity.
Collapse
|
5
|
Newton HP, Higgins DP, Casteriano A, Wright BR, Krockenberger MB, Miranda LHM. The CARD9 Gene in Koalas ( Phascolarctos cinereus): Does It Play a Role in the Cryptococcus-Koala Interaction? J Fungi (Basel) 2024; 10:409. [PMID: 38921395 PMCID: PMC11205041 DOI: 10.3390/jof10060409] [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: 05/08/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/27/2024] Open
Abstract
Cryptococcus is a genus of fungal pathogens that can infect and cause disease in a range of host species and is particularly prominent in koalas (Phascolarctos cinerus). Like other host species, koalas display a range of outcomes upon exposure to environmental Cryptococcus, from external nasal colonization to asymptomatic invasive infection and, in rare cases, severe clinical disease resulting in death. Host factors contributing to these varied outcomes are poorly understood. Due to their close relationship with eucalypt trees (a key environmental niche for Cryptococcus gattii) and suspected continual exposure to the pathogen, koalas provide a unique opportunity to examine host susceptibility in natural infections. Caspase recruitment domain-containing protein 9 (CARD9) is a key intracellular signaling protein in the fungal innate immune response. Humans with mutations in CARD9 succumb to several different severe and chronic fungal infections. This study is the first to sequence and explore CARD9 variation in multiple koalas using Sanger sequencing. Four CARD9 exons were successfully sequenced in 22 koalas from a New South Wales, Australia population. We found minimal variation between koalas across all four exons, an observation that was also made when CARD9 sequences were compared between koalas and six other species, including humans and mice. Ten single-nucleotide polymorphisms (SNP) were identified in this study and explored in the context of cryptococcal exposure outcomes. While we did not find any significant association with variation in cryptococcal outcomes, we found a high degree of conservation between species at several SNP loci that requires further investigation. The findings from this study lay the groundwork for further investigations of CARD9 and Cryptococcus both in koalas and other species, and highlight several considerations for future studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Luisa H. M. Miranda
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia; (H.P.N.); (D.P.H.); (A.C.); (B.R.W.); (M.B.K.)
| |
Collapse
|
6
|
Rossi SA, García-Barbazán I, Chamorro-Herrero I, Taborda CP, Zaragoza Ó, Zambrano A. Use of 2D minilungs from human embryonic stem cells to study the interaction of Cryptococcus neoformans with the respiratory tract. Microbes Infect 2024; 26:105260. [PMID: 37981028 DOI: 10.1016/j.micinf.2023.105260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
Organoids can meet the needs between the use of cell culture and in vivo work, bringing together aspects of multicellular tissues, providing a more similar in vitro system for the study of various components, including host-interactions with pathogens and drug response. Organoids are structures that resemble organs in vivo, originating from pluripotent stem cells (PSCs) or adult stem cells (ASCs). There is great interest in deepening the understanding of the use of this technology to produce information about fungal infections and their treatments. This work aims the use 2D human lung organoid derived from human embryonic stem cells (hESCs), to investigate Cryptococcus neoformans-host interactions. C. neoformans is an opportunistic fungus acquired by inhalation that causes systemic mycosis mainly in immunocompromised individuals. Our work highlights the suitability of human minilungs for the study of C. neoformans infection (adhesion, invasion and replication), the interaction with the surfactant and induction of the host's alveolar pro-inflammatory response.
Collapse
Affiliation(s)
- Suélen Andreia Rossi
- Biotechnology of Stem Cells and Organoids, Chronic Disease Program, Carlos III Health Institute, Madrid, Spain; Department of Microbiology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo, Brazil; Mycology Reference Laboratory, National Centre for Microbiology, Carlos III Health Institute, Madrid, Spain
| | - Irene García-Barbazán
- Mycology Reference Laboratory, National Centre for Microbiology, Carlos III Health Institute, Madrid, Spain
| | - Irene Chamorro-Herrero
- Biotechnology of Stem Cells and Organoids, Chronic Disease Program, Carlos III Health Institute, Madrid, Spain
| | - Carlos Pelleschi Taborda
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo, Brazil; Tropical Medicine Institute, Department of Dermatology, Faculty of Medicine, University of São Paulo, São Paulo 05403-000, Brazil.
| | - Óscar Zaragoza
- Mycology Reference Laboratory, National Centre for Microbiology, Carlos III Health Institute, Madrid, Spain; Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC, Carlos III Health Institute, CB21/13/00105), Madrid, Spain.
| | - Alberto Zambrano
- Biotechnology of Stem Cells and Organoids, Chronic Disease Program, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
7
|
Hansakon A, Angkasekwinai P. Murine Models of Cryptococcus Infection. Curr Protoc 2024; 4:e1001. [PMID: 38456766 DOI: 10.1002/cpz1.1001] [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: 03/09/2024]
Abstract
Cryptococcus is recognized as one of the emerging fungal pathogens that have major impact on diverse populations worldwide. Because of the high mortality rate and limited antifungal therapy options, there is an urgent need to understand the impact of dynamic processes between fungal pathogens and hosts that influence cryptococcal pathogenesis and disease outcomes. With known common limitations in human studies, experimental murine cryptococcosis models that can recapitulate human disease provide a valuable tool for studying fungal virulence and the host interaction, leading to development of better treatment strategies. Infection with Cryptococcus in mice via intranasal inhalation is mostly used because it is noninvasive and considered to be the most common mode of infection, strongly correlating with cryptococcal disease in humans. The protocols described in this article provide the procedures of establishing a murine model of Cryptococcus infection by intranasal inhalation and assessing the host immune response and disease progression during Cryptococcus infection. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Murine model of pulmonary cryptococcal infection via intranasal inhalation Basic Protocol 2: Assessment of the pulmonary immune response during Cryptococcus infection Support Protocol: Evaluation of pulmonary gene expression by real-time PCR Basic Protocol 3: Enumeration of survival rate and organ fungal burden.
Collapse
Affiliation(s)
- Adithap Hansakon
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|
8
|
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
|
9
|
Mjokane N, Sabiu S, Folorunso OS, Gcilitshana OMN, Albertyn J, Pohl CH, Sebolai OM. Cryptococcal proteases exhibit the potential to activate the latent SARS-CoV-2 spike protein. J Infect Public Health 2024; 17:263-270. [PMID: 38128410 DOI: 10.1016/j.jiph.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/04/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic has affected more than 650 million people and resulted in over 6.8 million deaths. Notably, the disease could co-manifest with microbial infections, like cryptococcosis, which also presents as a primary lung infection. OBJECTIVE In this contribution, we sought to determine if cryptococcal supernatant (which contains secreted furin-like proteases) could activate the SARS-CoV-2 spike protein. METHODS Molecular docking of the crystal structures of the SARS-CoV-2 spike protein (target) and selected cryptococcal proteases (ligands) was executed using the high ambiguity driven protein-protein docking (HADDOCK) server, with the furin protease serving as a reference ligand. The furin protease is found in human cells and typically activates the SARS-CoV-2 spike protein. Importantly, in order to provide experimental evidence for enzymatic activity, we also assessed the biochemical efficiency of cryptococcal proteases to initiate viral entry into HEK-293 T cells by SARS-CoV-2 spike pseudotyped Lentivirus. RESULTS We show that the selected cryptococcal proteases could interact with the spike protein, and some had a better or comparable binding affinity for the spike protein than furin protease following an in silico comparative analysis of the molecular docking parameters. Furthermore, it was noted that the biochemical efficiency of the cryptococcal supernatant to transduce HEK-293 T cells with SARS-CoV-2 pseudovirions was comparable (p > 0.05) to that of recombinant furin. CONCLUSIONS Taken together, these data show that cryptococcal proteases could activate the SARS-CoV-2 spike protein. In practice, it may be critical to determine if patients have an underlying cryptococcal infection, as this microbe could secrete proteases that may further activate the SARS-CoV-2 viral particles, thus undermining COVID-19 intervention measures.
Collapse
Affiliation(s)
- Nozethu Mjokane
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Drive, Park West, Bloemfontein, 9301, South Africa
| | - Saheed Sabiu
- Department of Biotechnology and Food Technology, Durban University of Technology, 121 Steve Biko Road, Berea Durban 4001, South Africa
| | - Olufemi S Folorunso
- Harvard Medical School, Department of Ophthalmology, Boston, MA, United States
| | - Onele M N Gcilitshana
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Drive, Park West, Bloemfontein, 9301, South Africa
| | - Jacobus Albertyn
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Drive, Park West, Bloemfontein, 9301, South Africa
| | - Carolina H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Drive, Park West, Bloemfontein, 9301, South Africa
| | - Olihile M Sebolai
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Drive, Park West, Bloemfontein, 9301, South Africa.
| |
Collapse
|
10
|
de Castro RJA, Marina CL, Sturny-Leclère A, Hoffmann C, Bürgel PH, Wong SSW, Aimanianda V, Varet H, Agrawal R, Bocca AL, Alanio A. Kicking sleepers out of bed: Macrophages promote reactivation of dormant Cryptococcus neoformans by extracellular vesicle release and non-lytic exocytosis. PLoS Pathog 2023; 19:e1011841. [PMID: 38033163 PMCID: PMC10715671 DOI: 10.1371/journal.ppat.1011841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/12/2023] [Accepted: 11/18/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages play a key role in disseminated cryptococcosis, a deadly fungal disease caused by Cryptococcus neoformans. This opportunistic infection can arise following the reactivation of a poorly characterized latent infection attributed to dormant C. neoformans. Here, we investigated the mechanisms underlying reactivation of dormant C. neoformans using an in vitro co-culture model of viable but non-culturable (VBNC; equivalent of dormant) yeast cells with bone marrow-derived murine macrophages (BMDMs). Comparative transcriptome analysis of BMDMs incubated with log, stationary phase or VBNC cells of C. neoformans showed that VBNC cells elicited a reduced transcriptional modification of the macrophage but retaining the ability to regulate genes important for immune response, such as NLRP3 inflammasome-related genes. We further confirmed the maintenance of the low immunostimulatory capacity of VBNC cells using multiplex cytokine profiling, and analysis of cell wall composition and dectin-1 ligands exposure. In addition, we evaluated the effects of classic (M1) or alternative (M2) macrophage polarization on VBNC cells. We observed that intracellular residence sustained dormancy, regardless of the polarization state of macrophages and despite indirect detection of pantothenic acid (or its derivatives), a known reactivator for VBNC cells, in the C. neoformans-containing phagolysosome. Notably, M0 and M2, but not M1 macrophages, induced extracellular reactivation of VBNC cells by the secretion of extracellular vesicles and non-lytic exocytosis. Our results indicate that VBNC cells retain the low immunostimulatory profile required for persistence of C. neoformans in the host. We also describe a pro-pathogen role of macrophage-derived extracellular vesicles in C. neoformans infection and reinforce the impact of non-lytic exocytosis and the macrophage profile on the pathophysiology of cryptococcosis.
Collapse
Affiliation(s)
- Raffael Júnio Araújo de Castro
- Translational Mycology Research Group, National Reference Center for Invasive Mycoses and Antifungals, Mycology Department, Institut Pasteur, Université Paris Cité, Paris, France
- Laboratory of Applied Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília, Distrito Federal, Brazil
| | - Clara Luna Marina
- Laboratory of Applied Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília, Distrito Federal, Brazil
| | - Aude Sturny-Leclère
- Translational Mycology Research Group, National Reference Center for Invasive Mycoses and Antifungals, Mycology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Christian Hoffmann
- Food Research Center, Department of Food Sciences and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Pedro Henrique Bürgel
- Laboratory of Applied Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília, Distrito Federal, Brazil
| | - Sarah Sze Wah Wong
- Immunobiology of Aspergillus, Institut Pasteur, Université Paris Cité, Paris, France
| | - Vishukumar Aimanianda
- Immunobiology of Aspergillus, Institut Pasteur, Université Paris Cité, Paris, France
| | - Hugo Varet
- Plate-forme Technologique Biomics, Institut Pasteur, Université Paris Cité, Paris, France
| | - Ruchi Agrawal
- Translational Mycology Research Group, National Reference Center for Invasive Mycoses and Antifungals, Mycology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Anamélia Lorenzetti Bocca
- Laboratory of Applied Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília, Distrito Federal, Brazil
| | - Alexandre Alanio
- Translational Mycology Research Group, National Reference Center for Invasive Mycoses and Antifungals, Mycology Department, Institut Pasteur, Université Paris Cité, Paris, France
- Laboratoire de parasitologie-mycologie, AP-HP, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
11
|
Davis MJ, Martin RE, Pinheiro GM, Hoke ES, Moyer S, Ueno K, Rodriguez-Gil JL, Mallett MA, Khillan JS, Pavan WJ, Chang YC, Kwon-Chung KJ. Inbred SJL mice recapitulate human resistance to Cryptococcus infection due to differential immune activation. mBio 2023; 14:e0212323. [PMID: 37800917 PMCID: PMC10653822 DOI: 10.1128/mbio.02123-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE Cryptococcosis studies often utilize the common C57BL/6J mouse model. Unfortunately, infection in these mice fails to replicate the basic course of human disease, particularly hampering immunological studies. This work demonstrates that SJL/J mice can recapitulate human infection better than other mouse strains. The immunological response to Cryptococcus infection in SJL/J mice was markedly different from C57BL/6J and much more productive in combating this infection. Characterization of infected mice demonstrated strain-specific genetic linkage and differential regulation of multiple important immune-relevant genes in response to Cryptococcus infection. While our results validate many of the previously identified immunological features of cryptococcosis, we also demonstrate limitations from previous mouse models as they may be less translatable to human disease. We concluded that SJL/J mice more faithfully recapitulate human cryptococcosis serving as an exciting new animal model for immunological and genetic studies.
Collapse
Affiliation(s)
- M. J. Davis
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - R. E. Martin
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - G. M. Pinheiro
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - E. S. Hoke
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - S. Moyer
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - K. Ueno
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - J. L. Rodriguez-Gil
- Genomics, Development and Disease Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - M. A. Mallett
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - J. S. Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - W. J. Pavan
- Genomics, Development and Disease Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Y. C. Chang
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - K. J. Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
12
|
Gouveia-Eufrasio L, de Freitas GJC, Costa MC, Peres-Emidio EC, Carmo PHF, Rodrigues JGM, de Rezende MC, Rodrigues VF, de Brito CB, Miranda GS, de Lima PA, da Silva LMV, Oliveira JBS, da Paixão TA, da Glória de Souza D, Fagundes CT, Peres NTDA, Negrão-Correa DA, Santos DA. The Th2 Response and Alternative Activation of Macrophages Triggered by Strongyloides venezuelensis Is Linked to Increased Morbidity and Mortality Due to Cryptococcosis in Mice. J Fungi (Basel) 2023; 9:968. [PMID: 37888224 PMCID: PMC10607621 DOI: 10.3390/jof9100968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Abstract
Cryptococcosis is a systemic mycosis that causes pneumonia and meningoencephalitis. Strongyloidiasis is a chronic gastrointestinal infection caused by parasites of the genus Strongyloides. Cryptococcosis and strongyloidiasis affect the lungs and are more prevalent in the same world regions, i.e., Africa and tropical countries such as Brazil. It is undeniable that those coincidences may lead to the occurrence of coinfections. However, there are no studies focused on the interaction between Cryptococcus spp. and Strongyloides spp. In this work, we aimed to investigate the interaction between Strongyloides venezuelensis (Sv) and Cryptococcus gattii (Cg) in a murine coinfection model. Murine macrophage exposure to Sv antigens reduced their ability to engulf Cg and produce reactive oxygen species, increasing the ability of fungal growth intracellularly. We then infected mice with both pathogens. Sv infection skewed the host's response to fungal infection, increasing lethality in a murine coinfection model. In addition to increased NO levels and arginase activity, coinfected mice presented a classic Th2 anti-Sv response: eosinophilia, higher levels of alternate activated macrophages (M2), increased concentrations of CCL24 and IL-4, and lower levels of IL-1β. This milieu favored fungal growth in the lungs with prominent translocation to the brain, increasing the host's tissue damage. In conclusion, our data shows that primary Sv infection promotes Th2 bias of the pulmonary response to Cg-infection and worsens its pathological outcomes.
Collapse
Affiliation(s)
- Ludmila Gouveia-Eufrasio
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - Gustavo José Cota de Freitas
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - Marliete Carvalho Costa
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - Eluzia Castro Peres-Emidio
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - Paulo Henrique Fonseca Carmo
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - João Gustavo Mendes Rodrigues
- Departamento de Parasitologia, Laboratório de Esquistossomose, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.A.N.-C.)
| | - Michelle Carvalho de Rezende
- Departamento de Parasitologia, Laboratório de Esquistossomose, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.A.N.-C.)
| | - Vanessa Fernandes Rodrigues
- Departamento de Parasitologia, Laboratório de Esquistossomose, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.A.N.-C.)
| | - Camila Bernardo de Brito
- Departamento de Microbiologia, Laboratório de Interação Microrganismo-Hospedeiro, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.d.G.d.S.); (C.T.F.)
| | - Guilherme Silva Miranda
- Departamento de Parasitologia, Laboratório de Esquistossomose, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.A.N.-C.)
| | - Pâmela Aparecida de Lima
- Departamento de Patologia, Laboratório de Patologia Celular e Molecular, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (T.A.d.P.)
| | - Lívia Mara Vitorino da Silva
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - Jefferson Bruno Soares Oliveira
- Departamento de Patologia, Laboratório de Patologia Celular e Molecular, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (T.A.d.P.)
| | - Tatiane Alves da Paixão
- Departamento de Patologia, Laboratório de Patologia Celular e Molecular, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (T.A.d.P.)
| | - Daniele da Glória de Souza
- Departamento de Microbiologia, Laboratório de Interação Microrganismo-Hospedeiro, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.d.G.d.S.); (C.T.F.)
| | - Caio Tavares Fagundes
- Departamento de Microbiologia, Laboratório de Interação Microrganismo-Hospedeiro, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.d.G.d.S.); (C.T.F.)
| | - Nalu Teixeira de Aguiar Peres
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| | - Deborah Aparecida Negrão-Correa
- Departamento de Parasitologia, Laboratório de Esquistossomose, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil (D.A.N.-C.)
| | - Daniel Assis Santos
- Departamento de Microbiologia, Laboratório de Micologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.-E.); (N.T.d.A.P.)
| |
Collapse
|
13
|
Jadimurthy R, Jagadish S, Nayak SC, Kumar S, Mohan CD, Rangappa KS. Phytochemicals as Invaluable Sources of Potent Antimicrobial Agents to Combat Antibiotic Resistance. Life (Basel) 2023; 13:948. [PMID: 37109477 PMCID: PMC10145550 DOI: 10.3390/life13040948] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Plants have been used for therapeutic purposes against various human ailments for several centuries. Plant-derived natural compounds have been implemented in clinics against microbial diseases. Unfortunately, the emergence of antimicrobial resistance has significantly reduced the efficacy of existing standard antimicrobials. The World Health Organization (WHO) has declared antimicrobial resistance as one of the top 10 global public health threats facing humanity. Therefore, it is the need of the hour to discover new antimicrobial agents against drug-resistant pathogens. In the present article, we have discussed the importance of plant metabolites in the context of their medicinal applications and elaborated on their mechanism of antimicrobial action against human pathogens. The WHO has categorized some drug-resistant bacteria and fungi as critical and high priority based on the need to develope new drugs, and we have considered the plant metabolites that target these bacteria and fungi. We have also emphasized the role of phytochemicals that target deadly viruses such as COVID-19, Ebola, and dengue. Additionally, we have also elaborated on the synergetic effect of plant-derived compounds with standard antimicrobials against clinically important microbes. Overall, this article provides an overview of the importance of considering phytogenous compounds in the development of antimicrobial compounds as therapeutic agents against drug-resistant microbes.
Collapse
Affiliation(s)
- Ragi Jadimurthy
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | - Swamy Jagadish
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | - Siddaiah Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India;
| | - Sumana Kumar
- Department of Microbiology, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | | |
Collapse
|
14
|
Rush RE, Blackwood CB, Lemons AR, Dannemiller KC, Green BJ, Croston TL. Persisting Cryptococcus yeast species Vishniacozyma victoriae and Cryptococcus neoformans elicit unique airway inflammation in mice following repeated exposure. Front Cell Infect Microbiol 2023; 13:1067475. [PMID: 36864880 PMCID: PMC9971225 DOI: 10.3389/fcimb.2023.1067475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Background Allergic airway disease (AAD) is a growing concern in industrialized nations and can be influenced by fungal exposures. Basidiomycota yeast species such as Cryptococcus neoformans are known to exacerbate allergic airway disease; however, recent indoor assessments have identified other Basidiomycota yeasts, including Vishniacozyma victoriae (syn. Cryptococcus victoriae), to be prevalent and potentially associated with asthma. Until now, the murine pulmonary immune response to repeated V. victoriae exposure was previously unexplored. Objective This study aimed to compare the immunological impact of repeated pulmonary exposure to Cryptococcus yeasts. Methods Mice were repeatedly exposed to an immunogenic dose of C. neoformans or V. victoriae via oropharyngeal aspiration. Bronchoalveolar lavage fluid (BALF) and lungs were collected to examine airway remodeling, inflammation, mucous production, cellular influx, and cytokine responses at 1 day and 21 days post final exposure. The responses to C. neoformans and V. victoriae were analyzed and compared. Results Following repeated exposure, both C. neoformans and V. victoriae cells were still detectable in the lungs 21 days post final exposure. Repeated C. neoformans exposure initiated myeloid and lymphoid cellular infiltration into the lung that worsened over time, as well as an IL-4 and IL-5 response compared to PBS-exposed controls. In contrast, repeated V. victoriae exposure induced a strong CD4+ T cell-driven lymphoid response that started to resolve by 21 days post final exposure. Discussion C. neoformans remained in the lungs and exacerbated the pulmonary immune responses as expected following repeated exposure. The persistence of V. victoriae in the lung and strong lymphoid response following repeated exposure were unexpected given its lack of reported involvement in AAD. Given the abundance in indoor environments and industrial utilization of V. victoriae, these results highlight the importance to investigate the impact of frequently detected fungal organisms on the pulmonary response following inhalational exposure. Moreover, it is important to continue to address the knowledge gap involving Basidiomycota yeasts and their impact on AAD.
Collapse
Affiliation(s)
- Rachael E. Rush
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Catherine B. Blackwood
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Angela R. Lemons
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Karen C. Dannemiller
- Department of Civil, Environmental & Geodetic Engineering, College of Engineering, Ohio State University, Columbus, OH, United States
- Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH, United States
| | - Brett J. Green
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Tara L. Croston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| |
Collapse
|
15
|
Sato K, Kawakami K. Mouse Model of Latent Cryptococcal Infection and Reactivation. Methods Mol Biol 2023; 2667:87-98. [PMID: 37145277 DOI: 10.1007/978-1-0716-3199-7_6] [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: 05/06/2023]
Abstract
AbstractCryptococcus neoformans is an opportunistic fungal pathogen that frequently causes fatal meningoencephalitis in patients with impaired immune responses. This fungus, an intracellularly growing microbe, evades host immunity, leading to a latent infection (latent C. neoformans infection: LCNI), and cryptococcal disease is developed by its reactivation when host immunity is suppressed. Elucidation of the pathophysiology of LCNI is difficult due to the lack of mouse models. Here we show the established methods for LCNI and reactivation.
Collapse
Affiliation(s)
- Ko Sato
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
- Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
16
|
Harvey W, Hutto EH, Chilton JA, Chamanza R, Mysore JV, Parry NM, Dick E, Wojcinski ZW, Piaia A, Garcia B, Flandre TD, Pardo ID, Cramer S, Wright JA, Bradley AE. Infectious diseases of non-human primates. SPONTANEOUS PATHOLOGY OF THE LABORATORY NON-HUMAN PRIMATE 2023:15-69. [DOI: 10.1016/b978-0-12-813088-9.00020-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
17
|
Tharappel AM, Li Z, Zhu YC, Wu X, Chaturvedi S, Zhang QY, Li H. Calcimycin Inhibits Cryptococcus neoformans In Vitro and In Vivo by Targeting the Prp8 Intein Splicing. ACS Infect Dis 2022; 8:1851-1868. [PMID: 35948057 PMCID: PMC9464717 DOI: 10.1021/acsinfecdis.2c00137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Drug resistance is a significant concern in the treatment of diseases, including cryptococcosis caused by Cryptococcus neoformans (Cne) and Cryptococcus gattii (Cga). Alternative drug targets are necessary to overcome drug resistance before it attains a critical stage. Splicing of inteins from pro-protein precursors is crucial for activities of essential proteins hosting intein elements in many organisms, including human pathogens such as Cne and Cga. Through a high-throughput screening, we identified calcimycin (CMN) as a potent Prp8 intein splicing inhibitor with a minimum inhibitory concentration (MIC) of 1.5 μg/mL against the wild-type Cne-H99 (Cne-WT or Cne). In contrast, CMN inhibited the intein-less mutant strain (Cne-Mut) with a 16-fold higher MIC. Interestingly, Aspergillus fumigatus and a few Candida species were resistant to CMN. Further studies indicated that CMN reduced virulence factors such as urease activity, melanin production, and biofilm formation in Cne. CMN also inhibited Cne intracellular infection in macrophages. In a target-specific split nanoluciferase assay, the IC50 of CMN was 4.6 μg/mL. Binding of CMN to recombinant Prp8 intein was demonstrated by thermal shift assay and microscale thermophoresis. Treating Cne cells with CMN reduced intein splicing. CMN was fungistatic and showed a synergistic effect with the known antifungal drug amphotericin B. Finally, CMN treatment at 20 mg/kg body weight led to 60% reduction in lung fungal load in a cryptococcal pulmonary infection mouse model. Overall, CMN represents a potent antifungal with a novel mechanism of action to treat Cne and possibly Cga infections.
Collapse
Affiliation(s)
- Anil Mathew Tharappel
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Zhong Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Yan Chun Zhu
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Xiangmeng Wu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
| | - Sudha Chaturvedi
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Qing-Yu Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
- The BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
18
|
Kassaza K, Wasswa F, Nielsen K, Bazira J. Cryptococcus neoformans Genotypic Diversity and Disease Outcome among HIV Patients in Africa. J Fungi (Basel) 2022; 8:734. [PMID: 35887489 PMCID: PMC9325144 DOI: 10.3390/jof8070734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
Cryptococcal meningoencephalitis, a disease with poor patient outcomes, remains the most prevalent invasive fungal infection worldwide, accounting for approximately 180,000 deaths each year. In several areas of sub-Saharan Africa with the highest HIV prevalence, cryptococcal meningitis is the leading cause of community-acquired meningitis, with a high mortality among HIV-infected individuals. Recent studies show that patient disease outcomes are impacted by the genetics of the infecting isolate. Yet, there is still limited knowledge of how these genotypic variations contribute to clinical disease outcome. Further, it is unclear how the genetic heterogeneity of C. neoformans and the extensive phenotypic variation observed between and within isolates affects infection and disease. In this review, we discuss current knowledge of how various genotypes impact disease progression and patient outcome in HIV-positive populations in sub-Saharan African, a setting with a high burden of cryptococcosis.
Collapse
Affiliation(s)
- Kennedy Kassaza
- Department of Microbiology and Parasitology, Mbarara University of Science and Technology, Mbarara P.O. Box 1410, Uganda; (K.K.); (F.W.)
| | - Fredrickson Wasswa
- Department of Microbiology and Parasitology, Mbarara University of Science and Technology, Mbarara P.O. Box 1410, Uganda; (K.K.); (F.W.)
| | - Kirsten Nielsen
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joel Bazira
- Department of Microbiology and Parasitology, Mbarara University of Science and Technology, Mbarara P.O. Box 1410, Uganda; (K.K.); (F.W.)
| |
Collapse
|
19
|
Cross-Kingdom Infection of Macrophages Reveals Pathogen- and Immune-Specific Global Reprogramming and Adaptation. mBio 2022; 13:e0168722. [PMID: 35862772 PMCID: PMC9426421 DOI: 10.1128/mbio.01687-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The relationship between the human microbiota and infectious disease outcome is a rapidly expanding area of study. Understanding how the host responds to changes in its symbiotic relationship with microbes provides new insight into how disruption can promote disease.
Collapse
|
20
|
Qian W, Li X, Liu Q, Lu J, Wang T, Zhang Q. Antifungal and Antibiofilm Efficacy of Paeonol Treatment Against Biofilms Comprising Candida albicans and/or Cryptococcus neoformans. Front Cell Infect Microbiol 2022; 12:884793. [PMID: 35669114 PMCID: PMC9163411 DOI: 10.3389/fcimb.2022.884793] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
Fungal populations are commonly found in natural environments and present enormous health care challenges, due to increased resistance to antifungal agents. Paeonol exhibits antifungal activities; nevertheless, the antifungal and antibiofilm activities of paeonol against Candida albicans and Cryptococcus neoformans remain largely unexplored. Here, we aimed to evaluate the antifungal and antibiofilm activities of paeonol against C. albicans and/or C. neoformans (i.e., against mono- or dual-species). The minimum inhibitory concentrations (MICs) of paeonol for mono-species comprising C. albicans or C. neoformans were 250 μg ml−1, whereas the MIC values of paeonol for dual-species were 500 μg ml−1. Paeonol disrupted cell membrane integrity and increased the influx of gatifloxacin into cells of mono- and dual-species cells, indicating an antifungal mode of action. Moreover, paeonol at 8 times the MIC damaged mono- and dual-species cells within C. albicans and C. neoformans biofilms, as it did planktonic cells. In particular, at 4 and 8 mg ml−1, paeonol efficiently dispersed preformed 48-h biofilms formed by mono- and dual-species cells, respectively. Paeonol inhibited effectively the yeast-to-hyphal-form transition of C. albicans and impaired capsule and melanin production of C. neoformans. The addition of 10 MIC paeonol to the medium did not shorten the lifespan of C. elegans, and 2 MIC paeonol could effectively protect the growth of C. albicans and C. neoformans-infected C. elegans. Furthermore, RNA sequencing was employed to examine the transcript profiling of C. albicans and C. neoformans biofilm cells in response to 1/2 MIC paeonol. RNA sequencing data revealed that paeonol treatment impaired biofilm formation of C. albicans by presumably downregulating the expression level of initial filamentation, adhesion, and growth-related genes, as well as biofilm biosynthesis genes, whereas paeonol inhibited biofilm formation of C. neoformans by presumably upregulating the expression level of ergosterol biosynthesis-related genes. Together, the findings of this study indicate that paeonol can be explored as a candidate antifungal agent for combating serious single and mixed infections caused by C. albicans and C. neoformans.
Collapse
Affiliation(s)
- Weidong Qian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Xinchen Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Qiming Liu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Jiaxing Lu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Ting Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
- *Correspondence: Ting Wang, ; Qian Zhang,
| | - Qian Zhang
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
- *Correspondence: Ting Wang, ; Qian Zhang,
| |
Collapse
|
21
|
An Immunogenic and Slow-Growing Cryptococcal Strain Induces a Chronic Granulomatous Infection in Murine Lungs. Infect Immun 2022; 90:e0058021. [PMID: 35587201 PMCID: PMC9202370 DOI: 10.1128/iai.00580-21] [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
Many successful pathogens cause latent infections, remaining dormant within the host for years but retaining the ability to reactivate to cause symptomatic disease. The human opportunistic fungal pathogen Cryptococcus neoformans establishes latent pulmonary infections in immunocompetent individuals upon inhalation from the environment. These latent infections are frequently characterized by granulomas, or foci of chronic inflammation, that contain dormant and persistent cryptococcal cells. Immunosuppression can cause these granulomas to break down and release fungal cells that proliferate, disseminate, and eventually cause lethal cryptococcosis. This course of fungal latency and reactivation is understudied due to limited models, as chronic pulmonary granulomas do not typically form in mouse cryptococcal infections. A loss-of-function mutation in the Cryptococcus-specific MAR1 gene was previously described to alter cell surface remodeling in response to host signals. Here, we demonstrate that the mar1Δ mutant strain persists long term in a murine inhalation model of cryptococcosis, inducing a chronic pulmonary granulomatous response. We find that murine infections with the mar1Δ mutant strain are characterized by reduced fungal burden, likely due to the low growth rate of the mar1Δ mutant strain at physiological temperature, and an altered host immune response, likely due to inability of the mar1Δ mutant strain to properly employ virulence factors. We propose that this combination of features in the mar1Δ mutant strain collectively promotes the induction of a more chronic inflammatory response and enables long-term fungal persistence within these granulomatous regions.
Collapse
|
22
|
Normile TG, Del Poeta M. Three Models of Vaccination Strategies Against Cryptococcosis in Immunocompromised Hosts Using Heat-Killed Cryptococcus neoformans Δ sgl1. Front Immunol 2022; 13:868523. [PMID: 35615354 PMCID: PMC9124966 DOI: 10.3389/fimmu.2022.868523] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/11/2022] [Indexed: 12/16/2022] Open
Abstract
Vaccines are one of the greatest medical accomplishments to date, yet no fungal vaccines are currently available in humans mainly because opportunistic mycoses generally occur during immunodeficiencies necessary for vaccine protection. In previous studies, a live, attenuated Cryptococcus neoformans Δsgl1 mutant accumulating sterylglucosides was found to be avirulent and protected mice from a subsequent lethal infection even in absence of CD4+ T cells, a condition most associated with cryptococcosis (e.g., HIV). Here, we tested three strategies of vaccination against cryptococcosis. First, in our preventative model, protection was achieved even after a 3-fold increase of the vaccination window. Second, because live C. neoformans Δsgl1-vaccinated mice challenged more than once with WT strain had a significant decrease in lung fungal burden, we tested C. neoformans Δsgl1 as an immunotherapeutic. We found that therapeutic administrations of HK C. neoformans Δsgl1 post WT challenge significantly improves the lung fungal burden. Similarly, therapeutic administration of HK C. neoformans Δsgl1 post WT challenge resulted in 100% or 70% survival depending on the time of vaccine administration, suggesting that HK Δsgl1 is a robust immunotherapeutic option. Third, we investigated a novel model of vaccination in preventing reactivation from lung granuloma using C. neoformans Δgcs1. Remarkably, we show that administration of HK Δsgl1 prevents mice from reactivating Δgcs1 upon inducing immunosuppression with corticosteroids or by depleting CD4+ T cells. Our results suggest that HK Δsgl1 represents a clinically relevant, efficacious vaccine that confers robust host protection in three models of vaccination against cryptococcosis even during CD4-deficiency.
Collapse
Affiliation(s)
- Tyler G. Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States,Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY, United States,Veterans Administration Medical Center, Northport, NY, United States,*Correspondence: Maurizio Del Poeta,
| |
Collapse
|
23
|
Forrester JV, Mölzer C, Kuffova L. Immune Privilege Furnishes a Niche for Latent Infection. FRONTIERS IN OPHTHALMOLOGY 2022; 2:869046. [PMID: 38983514 PMCID: PMC11182092 DOI: 10.3389/fopht.2022.869046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 07/11/2024]
Abstract
The microenvironment of the CNS (eye and brain) is fertile ground for infection if the barriers are breached. The result of pathogen invasion is often devastating destruction of tissues. In the eye, inflammation is broadly classified either as "infectious" (i.e. caused by infection) or "non-infectious". However, increasingly, forms of intraocular inflammation (IOI), which clinically appear to be "non-infectious" turn out to be initiated by infectious agents, suggesting that pathogens have been retained in latent or persistent form within ocular tissues and have reactivated to cause overt disease. A similar pathogenesis applies to latent infections in the brain. Not all CNS tissues provide an equally protective niche while different pathogens escape detection using different strategies. This review summarises how immune privilege (IP) in the CNS may be permissive for latent infection and allow the eye and the brain to act as a reservoir of pathogens which often remain undetected for the lifetime of the host but in states of immune deficiency may be activated to cause sight- and life-threatening inflammation.
Collapse
Affiliation(s)
- John V Forrester
- Ocular Immunology Group, Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Christine Mölzer
- Ocular Immunology Group, Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Lucia Kuffova
- Ocular Immunology Group, Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| |
Collapse
|
24
|
Muselius B, Durand SL, Geddes-McAlister J. Proteomics of Cryptococcus neoformans: From the Lab to the Clinic. Int J Mol Sci 2021; 22:12390. [PMID: 34830272 PMCID: PMC8618913 DOI: 10.3390/ijms222212390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Fungal pathogens cause an array of diseases by targeting both immunocompromised and immunocompetent hosts. Fungi overcome our current arsenal of antifungals through the emergence and evolution of resistance. In particular, the human fungal pathogen, Cryptococcus neoformans is found ubiquitously within the environment and causes severe disease in immunocompromised individuals around the globe with limited treatment options available. To uncover fundamental knowledge about this fungal pathogen, as well as investigate new detection and treatment strategies, mass spectrometry-based proteomics provides a plethora of tools and applications, as well as bioinformatics platforms. In this review, we highlight proteomics approaches within the laboratory to investigate changes in the cellular proteome, secretome, and extracellular vesicles. We also explore regulation by post-translational modifications and the impact of protein-protein interactions. Further, we present the development and comprehensive assessment of murine models of cryptococcal infection, which provide valuable tools to define the dynamic relationship between the host and pathogen during disease. Finally, we explore recent quantitative proteomics studies that begin to extrapolate the findings from the bench to the clinic for improved methods of fungal detection and monitoring. Such studies support a framework for personalized medical approaches to eradicate diseases caused by C. neoformans.
Collapse
Affiliation(s)
| | | | - Jennifer Geddes-McAlister
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON N1G 2W1, Canada; (B.M.); (S.-L.D.)
| |
Collapse
|
25
|
Normile TG, Rella A, Del Poeta M. Cryptococcus neoformans Δ sgl1 Vaccination Requires Either CD4 + or CD8 + T Cells for Complete Host Protection. Front Cell Infect Microbiol 2021; 11:739027. [PMID: 34568097 PMCID: PMC8455912 DOI: 10.3389/fcimb.2021.739027] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022] Open
Abstract
Cryptococcus neoformans is a fungal pathogen causing life-threatening meningoencephalitis in susceptible individuals. Fungal vaccine development has been hampered by the fact that cryptococcosis occurs during immunodeficiency. We previously reported that a C. neoformans mutant (Δsgl1) accumulating sterylglucosides (SGs) is avirulent and provides complete protection to WT challenge, even under CD4+ T cell depletion, an immunodeficient condition commonly associated with cryptococcosis. We found high levels of SGs in the lungs post-immunization with Δsgl1 that decreased upon fungal clearance. Th1 cytokines increased whereas Th2 cytokines concurrently decreased, coinciding with a large recruitment of leukocytes to the lungs. Depletion of B or CD8+ T cells did not affect either Δsgl1 clearance or protection from WT challenge. Although CD4+ T cell depletion affected clearance, mice were still protected indicating that clearance of the mutant was not necessary for host protection. Protection was lost only when both CD4+ and CD8+ T cells were depleted, highlighting a previously unexplored role of fungal-derived SGs as an immunoadjuvant for host protection against cryptococcosis.
Collapse
Affiliation(s)
- Tyler G. Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
| | - Antonella Rella
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY, United States
- Veterans Administration Medical Center, Northport, NY, United States
| |
Collapse
|
26
|
Ma Y, Wang X, Li R. Cutaneous and subcutaneous fungal infections: recent developments on host-fungus interactions. Curr Opin Microbiol 2021; 62:93-102. [PMID: 34098513 DOI: 10.1016/j.mib.2021.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
The incidence of skin fungal infections is increasing at an alarming rate worldwide, presenting a major challenge to health professionals. Cutaneous and subcutaneous fungal infections are caused by pathogenic or opportunistic organisms varying from mold, yeasts, to dimorphic fungi. Recently, skin fungal have been increasingly reported and studied, giving rise to crucial breakthroughs in etiology and pathogenesis. This review aims to summarize recent insights into the clinical and etiological characteristics of common skin fungal infections according to different fungal species, as well as remarkable advances in the immune mechanisms. We hope it will be helpful to understand these diverse skin fungal infections, and bring about the latest developments that may facilitate novel diagnostic and therapeutic approaches to improve the outcomes in these patients.
Collapse
Affiliation(s)
- Yubo Ma
- Department of Dermatology and Venerology, Peking University First Hospital, China; Research Center for Medical Mycology, Peking University, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, China; National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xiaowen Wang
- Department of Dermatology and Venerology, Peking University First Hospital, China; Research Center for Medical Mycology, Peking University, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, China; National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China.
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, China; Research Center for Medical Mycology, Peking University, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, China; National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China.
| |
Collapse
|
27
|
Santos ALS, Braga-Silva LA, Gonçalves DS, Ramos LS, Oliveira SSC, Souza LOP, Oliveira VS, Lins RD, Pinto MR, Muñoz JE, Taborda CP, Branquinha MH. Repositioning Lopinavir, an HIV Protease Inhibitor, as a Promising Antifungal Drug: Lessons Learned from Candida albicans-In Silico, In Vitro and In Vivo Approaches. J Fungi (Basel) 2021; 7:jof7060424. [PMID: 34071195 PMCID: PMC8229492 DOI: 10.3390/jof7060424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
The repurposing strategy was applied herein to evaluate the effects of lopinavir, an aspartic protease inhibitor currently used in the treatment of HIV-infected individuals, on the globally widespread opportunistic human fungal pathogen Candida albicans by using in silico, in vitro and in vivo approaches in order to decipher its targets on fungal cells and its antifungal mechanisms of action. Secreted aspartic proteases (Saps) are the obviously main target of lopinavir. To confirm this hypothesis, molecular docking assays revealed that lopinavir bound to the Sap2 catalytic site of C. albicans as well as inhibited the Sap hydrolytic activity in a typically dose-dependent manner. The inhibition of Saps culminated in the inability of C. albicans yeasts to assimilate the unique nitrogen source (albumin) available in the culture medium, culminating with fungal growth inhibition (IC50 = 39.8 µM). The antifungal action of lopinavir was corroborated by distinct microscopy analyses, which evidenced drastic and irreversible changes in the morphology that justified the fungal death. Furthermore, our results revealed that lopinavir was able to (i) arrest the yeasts-into-hyphae transformation, (ii) disturb the synthesis of neutral lipids, including ergosterol, (iii) modulate the surface-located molecules, such as Saps and mannose-, sialic acid- and N-acetylglucosamine-containing glycoconjugates, (iv) diminish the secretion of hydrolytic enzymes, such as Saps and esterase, (v) negatively influence the biofilm formation on polystyrene surface, (vi) block the in vitro adhesion to epithelial cells, (vii) contain the in vivo infection in both immunocompetent and immunosuppressed mice and (viii) reduce the Sap production by yeasts recovered from kidneys of infected animals. Conclusively, the exposed results highlight that lopinavir may be used as a promising repurposing drug against C. albicans infection as well as may be used as a lead compound for the development of novel antifungal drugs.
Collapse
Affiliation(s)
- André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
- Correspondence: (A.L.S.S.); (M.H.B.); Tel.: +55-21-3938-0366 (A.L.S.S.)
| | - Lys A. Braga-Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Diego S. Gonçalves
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
| | - Simone S. C. Oliveira
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
| | - Lucieri O. P. Souza
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
| | - Vanessa S. Oliveira
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife 50740-465, Brazil; (V.S.O.); (R.D.L.)
| | - Roberto D. Lins
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife 50740-465, Brazil; (V.S.O.); (R.D.L.)
| | - Marcia R. Pinto
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense (UFF), Niterói 24210-130, Brazil;
| | - Julian E. Muñoz
- MICROS Group, Medicine Traslacional Institute, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia;
| | - Carlos P. Taborda
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo 05508-060, Brazil;
- Laboratório de Micologia Médica—LIM53/IMTSP, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Correspondence: (A.L.S.S.); (M.H.B.); Tel.: +55-21-3938-0366 (A.L.S.S.)
| |
Collapse
|