1
|
Takemura K, Kolasinski V, Del Poeta M, Vieira de Sa NF, Garg A, Ojima I, Del Poeta M, Pereira de Sa N. Iron acquisition strategies in pathogenic fungi. mBio 2025:e0121125. [PMID: 40391928 DOI: 10.1128/mbio.01211-25] [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: 05/22/2025] Open
Abstract
Iron plays a crucial role in various biological processes, including enzyme function, DNA replication, energy production, oxygen transport, lipid, and carbon metabolism. Although it is abundant in the Earth's crust, its bioavailability is restricted by the insolubility of ferric iron (Fe³+) and the auto-oxidation of ferrous iron (Fe²+) in oxygen-rich environments. This limitation poses significant challenges for all organisms, including fungi, which have developed intricate mechanisms for iron acquisition and utilization. These mechanisms include reductive iron uptake, siderophore production/transport, and heme utilization. Fungi employ a variety of enzymes-such as ferric reductases, ferroxidases, permeases, and transporters-to regulate intracellular iron levels effectively. The challenge is heightened for pathogenic fungi during infection, as they must compete with the host's iron-binding proteins like transferrin and lactoferrin, which sequester iron to restrict pathogen growth. This review delves into the iron acquisition strategies of medically important fungi, emphasizing the roles of reductive iron uptake and siderophore pathways. Understanding these mechanisms is vital for enhancing our knowledge of fungal pathogenesis and developing effective treatments. By targeting these iron acquisition processes, new antifungal therapies can be formulated more effectively to combat fungal infections.
Collapse
Affiliation(s)
- Kathryn Takemura
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Vanessa Kolasinski
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Matteo Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | | | - Ashna Garg
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Maurizio Del Poeta
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Veterans Affairs Medical Center, Northport, New York, USA
| | - Nivea Pereira de Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
2
|
Sen M, Priyanka BM, Anusha D, Puneetha S, Setlur AS, Karunakaran C, Tandur A, Prashant CS, Niranjan V. Computational targeting of iron uptake proteins in Covid-19 induced mucormycosis to identify inhibitors via molecular dynamics, molecular mechanics and density function theory studies. In Silico Pharmacol 2024; 12:90. [PMID: 39355758 PMCID: PMC11439861 DOI: 10.1007/s40203-024-00264-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 10/03/2024] Open
Abstract
Mucormycosis is a concerning invasive fungal infection with difficult diagnosis, high mortality rates, and limited treatment options. Iron availability is crucial for fungal growth that causes this disease. This study aimed to computationally target iron uptake proteins in Rhizopus arrhizus, Lichtheimia corymbifera, and Mucor circinelloides to identify inhibitors, thereby halting fungal growth and intervening in mucormycosis pathogenesis. Seven important iron uptake proteins were identified, modeled, and validated using Ramachandran plots. An in-house antifungal library of ~ 15,401 compounds was screened in molecular docking studies with these proteins. The best small molecule-protein complexes were simulated at 100 ns using Maestro, Schrodinger. Toxicity predictions suggested all six molecules, identified as the best binding compounds to seven proteins, belonged to lower toxicity levels per GHS classification. A molecular mechanics GBSA study for all seven complexes indicated low standard deviations after calculating free binding energies every 10 ns of the 100 ns trajectory. Density functional theory via quantum mechanics approaches highlighted the HOMO, LUMO, and other properties of the six best-bound molecules, revealing their binding capabilities and behaviour. This study sheds light on the molecular mechanisms and protein-ligand interactions, providing a multi-dimensional view towards the use of FDBD01920, FDBD01923, and FDBD01848 as stable antifungal ligands. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00264-7.
Collapse
Affiliation(s)
- Manjima Sen
- Department of Public Health Dentistry, DAPM RV Dental College, Bangalore, 560078 India
| | - B M Priyanka
- Department of Oral Medicine and Diagnostic Radiology, DAPM RV Dental College, Bangalore, 560078 India
| | - D Anusha
- Department of Periodontia, DAPM RV Dental College, Bangalore, 560078 India
| | - S Puneetha
- Department of Oral Pathology and Microbiology, DAPM RV Dental College, Bangalore, 560078 India
| | - Anagha S Setlur
- Department of Biotechnology, RV College of Engineering, Bangalore, 560059 India
| | | | - Amulya Tandur
- Department of Biotechnology, RV College of Engineering, Bangalore, 560059 India
| | - C S Prashant
- Department of Orthodontics, DAPM RV Dental College, Bangalore, 560078 India
| | - Vidya Niranjan
- Department of Biotechnology, RV College of Engineering, Bangalore, 560059 India
| |
Collapse
|
3
|
Nayak A, Khedri A, Chavarria A, Sanders KN, Ghalei H, Khoshnevis S. Sinefungin, a natural nucleoside analog of S-adenosyl methionine, impairs the pathogenicity of Candida albicans. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:23. [PMID: 39268078 PMCID: PMC11391927 DOI: 10.1038/s44259-024-00040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/19/2024] [Indexed: 09/15/2024]
Abstract
Candida albicans, an opportunistic fungal pathogen, causes life-threatening infections in immunocompromised patients. Current antifungals are limited by toxicity, drug-drug interactions, and emerging resistance, underscoring the importance of identifying novel treatment approaches. Here, we elucidate the impact of sinefungin, an analog of S-adenosyl methionine, on the virulence of C. albicans strain SC5314 and clinical isolates. Our data indicate that sinefungin impairs pathogenic traits of C. albicans including hyphal morphogenesis, biofilm formation, adhesion to epithelial cells, and virulence towards Galleria mellonella, highlighting sinefungin as an avenue for therapeutic intervention. We determine that sinefungin particularly disturbs N6-methyladenosine (m6A) formation. Transcriptome analysis of C. albicans hyphae upon sinefungin treatment reveals an increase in transcripts related to the yeast form and decrease in those associated with hyphae formation and virulence. Collectively, our data propose sinefungin as a potent molecule against C. albicans and emphasize further exploration of post-transcriptional control mechanisms of pathogenicity for antifungal design.
Collapse
Affiliation(s)
- Anushka Nayak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Azam Khedri
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Alejandro Chavarria
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Kyla N. Sanders
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Sohail Khoshnevis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Van Genechten W, Vergauwen R, Van Dijck P. The intricate link between iron, mitochondria and azoles in Candida species. FEBS J 2024; 291:3568-3580. [PMID: 37846606 DOI: 10.1111/febs.16977] [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: 05/21/2023] [Revised: 09/19/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Invasive fungal infections are rapidly increasing, and the opportunistic pathogenic Candida species are the fourth most common cause of nosocomial systemic infections. The current antifungal classes, of which azoles are the most widely used, all have shortcomings. Azoles are generally considered fungistatic rather than fungicidal, they do not actively kill fungal cells and therefore resistance against azoles can be rapidly acquired. Combination therapies with azoles provide an interesting therapeutic outlook and agents limiting iron are excellent candidates. We summarize how iron is acquired by the host and transported towards both storage and iron-utilizing organelles. We indicate whether these pathways alter azole susceptibility and/or tolerance, to finally link these transport mechanisms to mitochondrial iron availability. In this review, we highlight putative novel intracellular iron shuffling mechanisms and indicate that mitochondrial iron dynamics in relation to azole treatment and iron limitation is a significant knowledge gap.
Collapse
Affiliation(s)
- Wouter Van Genechten
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| | - Rudy Vergauwen
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| |
Collapse
|
5
|
Xiong L, Goerlich K, Do E, Mitchell AP. Strain variation in the Candida albicans iron limitation response. mSphere 2024; 9:e0037224. [PMID: 38980069 PMCID: PMC11288005 DOI: 10.1128/msphere.00372-24] [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: 05/03/2024] [Accepted: 06/16/2024] [Indexed: 07/10/2024] Open
Abstract
Iron acquisition is critical for pathogens to proliferate during invasive infection, and the human fungal pathogen Candida albicans is no exception. The iron regulatory network, established in reference strain SC5314 and derivatives, includes the central player Sef1, a transcription factor that activates iron acquisition genes in response to iron limitation. Here, we explored potential variation in this network among five diverse C. albicans strains through mutant analysis, Nanostring gene expression profiling, and, for two strains, RNA-Seq. Our findings highlight four features that may inform future studies of natural variation and iron acquisition in this species. (i) Conformity: In all strains, major iron acquisition genes are upregulated during iron limitation, and a sef1Δ/Δ mutation impairs that response and growth during iron limitation. (ii) Response variation: Some aspects of the iron limitation response vary among strains, notably the activation of hypha-associated genes. As this gene set is tied to tissue damage and virulence, variation may impact the progression of infection. (iii) Genotype-phenotype variation: The impact of a sef1Δ/Δ mutation on cell wall integrity varies, and for the two strains examined the phenotype correlated with sef1Δ/Δ impact on several cell wall integrity genes. (iv) Phenotype discovery: DNA repair genes were induced modestly by iron limitation in sef1Δ/Δ mutants, with fold changes we would usually ignore. However, the response occurred in both strains tested and was reminiscent of a much stronger response described in Cryptococcus neoformans, a suggestion that it may have biological meaning. In fact, we observed that the iron limitation of a sef1Δ/Δ mutant caused recessive phenotypes to emerge at two heterozygous loci. Overall, our results show that a network that is critical for pathogen proliferation presents variation outside of its core functions.IMPORTANCEA key virulence factor of Candida albicans is the ability to maintain iron homeostasis in the host where iron is scarce. We focused on a central iron regulator, SEF1. We found that iron regulator Sef1 is required for growth, cell wall integrity, and genome integrity during iron limitation. The novel aspect of this work is the characterization of strain variation in a circuit that is required for survival in the host and the connection of iron acquisition to genome integrity in C. albicans.
Collapse
Affiliation(s)
- Liping Xiong
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | | | - Eunsoo Do
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
6
|
Tanwar M, Singh A, Singh TP, Sharma S, Sharma P. Comprehensive Review on the Virulence Factors and Therapeutic Strategies with the Aid of Artificial Intelligence against Mucormycosis. ACS Infect Dis 2024; 10:1431-1457. [PMID: 38682683 DOI: 10.1021/acsinfecdis.4c00082] [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: 05/01/2024]
Abstract
Mucormycosis, a rare but deadly fungal infection, was an epidemic during the COVID-19 pandemic. The rise in cases (COVID-19-associated mucormycosis, CAM) is attributed to excessive steroid and antibiotic use, poor hospital hygiene, and crowded settings. Major contributing factors include diabetes and weakened immune systems. The main manifesting forms of CAM─cutaneous, pulmonary, and the deadliest, rhinocerebral─and disseminated infections elevated mortality rates to 85%. Recent focus lies on small-molecule inhibitors due to their advantages over standard treatments like surgery and liposomal amphotericin B (which carry several long-term adverse effects), offering potential central nervous system penetration, diverse targets, and simpler dosing owing to their small size, rendering the ability to traverse the blood-brain barrier via passive diffusion facilitated by the phospholipid membrane. Adaptation and versatility in mucormycosis are facilitated by a multitude of virulence factors, enabling the pathogen to dynamically respond to various environmental stressors. A comprehensive understanding of these virulence mechanisms is imperative for devising effective therapeutic interventions against this highly opportunistic pathogen that thrives in immunocompromised individuals through its angio-invasive nature. Hence, this Review delineates the principal virulence factors of mucormycosis, the mechanisms it employs to persist in challenging host environments, and the current progress in developing small-molecule inhibitors against them.
Collapse
Affiliation(s)
- Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Tej Pal Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
7
|
Pinsky M, Kornitzer D. Genetic Analysis of Candida albicans Filamentation by the Iron Chelator BPS Reveals a Role for a Conserved Kinase-WD40 Protein Pair. J Fungi (Basel) 2024; 10:83. [PMID: 38276029 PMCID: PMC10820326 DOI: 10.3390/jof10010083] [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: 12/17/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Candida albicans is a major human pathogenic fungus that is distinguished by its capability to switch from a yeast to a hyphal morphology under different conditions. Here, we analyze the cellular effects of high concentrations of the iron chelator bathophenanthroline disulfonate (BPS). BPS inhibits cellular growth by withholding iron, but when iron chelation is overcome by the addition of hemoglobin as an iron source, the cells resume growth as hyphae. The BPS hyphal induction pathway was characterized by identifying the hyphal-specific transcription factors that it requires and by a forward genetic screen for mutants that fail to form hyphae in BPS using a transposon library generated in a haploid strain. Among the mutants identified are the DYRK1-like kinase Yak1 and Orf19.384, a homolog of the DYRK1-associated protein WDR68/DCAF7. Orf19.384 nuclear localization depends on Yak1, similar to their mammalian counterparts. We identified the hyphal suppressor transcription factor Sfl1 as a candidate target of Yak1-Orf19.384 and show that Sfl1 modification is similarly affected in the yak1 and orf19.384 mutant strains. These results suggest that DYRK1/Yak1 and WDR68/Orf19.384 represent a conserved protein pair that regulates cell differentiation from fungi to animals.
Collapse
Affiliation(s)
| | - Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion—I.I.T., Haifa 31096, Israel;
| |
Collapse
|
8
|
Velumani K, Arasu A, Issac PK, Kishore Kumar MS, Guru A, Arockiaraj J. Advancements of fish-derived peptides for mucormycosis: a novel strategy to treat diabetic compilation. Mol Biol Rep 2023; 50:10485-10507. [PMID: 37917415 DOI: 10.1007/s11033-023-08882-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023]
Abstract
Mucormycosis, an extremely fatal fungal infection, is a major hurdle in the treatment of diabetes consequences. The increasing prevalence and restricted treatment choices urge the investigation of novel therapeutic techniques. Because of their effective antimicrobial characteristics and varied modes of action, fish-derived peptides have lately emerged as viable options in the fight against mucormycosis. This review examines the potential further application of fish-derived peptides in diagnosing and managing mucormycosis in relation to diabetic complications. First, we examine the pathophysiology of mucormycosis and the difficulties in treating it in diabetics. We emphasize the critical need for alternative therapeutic methods for tackling the limitations of currently available antifungal medicines. The possibility of fish-derived peptides as an innovative approach to combat mucormycosis is then investigated. These peptides, derived from several fish species, provide wide antimicrobial properties against a variety of diseases. They also have distinct modes of action, such as rupture of cell membranes, suppression of development, and modification of the host immunological response. Furthermore, we investigate the problems and prospects connected with the clinical application of fish-derived peptides. Ultimately, future advances in fish-derived peptides, offer interesting avenues for the management of mucormycosis in the context of diabetic comorbidities. More research and clinical trials are needed to properly investigate these peptide's therapeutic potential and pave the way for their adoption into future antifungal therapies.
Collapse
Affiliation(s)
- Kadhirmathiyan Velumani
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, Tamil Nadu, 602 105, India
| | - Abirami Arasu
- Department of Microbiology, SRM Arts and Science College, Kattankulathur, Chennai, Tamil Nadu, 603 203, India
| | - Praveen Kumar Issac
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, Tamil Nadu, 602 105, India.
| | - Meenakshi Sundaram Kishore Kumar
- Biomedical Research Unit and Laboratory Animal Centre (BRULAC), Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
9
|
Delaney C, Alapati S, Alshehri M, Kubalova D, Veena CLR, Abusrewil S, Short B, Bradshaw D, Brown JL. Investigating the role of Candida albicans as a universal substrate for oral bacteria using a transcriptomic approach: implications for interkingdom biofilm control? APMIS 2023; 131:601-612. [PMID: 37170476 DOI: 10.1111/apm.13327] [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: 03/03/2023] [Accepted: 04/21/2023] [Indexed: 05/13/2023]
Abstract
Candida albicans is frequently identified as a colonizer of the oral cavity in health and has recently been termed a "keystone" commensal due to its role on the bacterial communities. However, the role that C. albicans plays in such interactions is not fully understood. Therefore, this study aimed to identify the relationship between C. albicans and bacteria associated with oral symbiosis and dysbiosis. To do this, we evaluated the ability of C. albicans to support the growth of the aerobic commensal Streptococcus gordonii and the anaerobic pathogens Fusobacterium nucleatum and Porphyromonas gingivalis in the biofilm environment. RNA-Sequencing with the Illumina platform was then utilized to identify C. albicans gene expression and functional pathways involved during such interactions in dual-species and a 4-species biofilm model. Results indicated that C. albicans was capable of supporting growth of all three bacteria, with a significant increase in colony counts of each bacteria in the dual-species biofilm (p < 0.05). We identified specific functional enrichment of pathways in our 4-species community as well as transcriptional profiles unique to the F. nucleatum and S. gordonii dual-species biofilms, indicating a species-specific effect on C. albicans. Candida-related hemin acquisition and heat shock protein mediated processes were unique to the organism following co-culture with anaerobic and aerobic bacteria, respectively, suggestive that such pathways may be feasible options for therapeutic targeting to interfere with these fungal-bacterial interactions. Targeted antifungal therapy may be considered as an option for biofilm destabilization and treatment of complex communities. Moving forward, we propose that further studies must continue to investigate the role of this fungal organism in the context of the interkingdom nature of oral diseases.
Collapse
Affiliation(s)
- Christopher Delaney
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | - Susanth Alapati
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | - Muhanna Alshehri
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | - Dominika Kubalova
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | - Chandra Lekha Ramalingham Veena
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | - Sumaya Abusrewil
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | - Bryn Short
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| | | | - Jason L Brown
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Biofilm Research Network (www.glasgowbiofilms.ac.uk), Glasgow, UK
| |
Collapse
|
10
|
Pijuan J, Moreno DF, Yahya G, Moisa M, Ul Haq I, Krukiewicz K, Mosbah R, Metwally K, Cavalu S. Regulatory and pathogenic mechanisms in response to iron deficiency and excess in fungi. Microb Biotechnol 2023; 16:2053-2071. [PMID: 37804207 PMCID: PMC10616654 DOI: 10.1111/1751-7915.14346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023] Open
Abstract
Iron is an essential element for all eukaryote organisms because of its redox properties, which are important for many biological processes such as DNA synthesis, mitochondrial respiration, oxygen transport, lipid, and carbon metabolism. For this reason, living organisms have developed different strategies and mechanisms to optimally regulate iron acquisition, transport, storage, and uptake in different environmental responses. Moreover, iron plays an essential role during microbial infections. Saccharomyces cerevisiae has been of key importance for decrypting iron homeostasis and regulation mechanisms in eukaryotes. Specifically, the transcription factors Aft1/Aft2 and Yap5 regulate the expression of genes to control iron metabolism in response to its deficiency or excess, adapting to the cell's iron requirements and its availability in the environment. We also review which iron-related virulence factors have the most common fungal human pathogens (Aspergillus fumigatus, Cryptococcus neoformans, and Candida albicans). These factors are essential for adaptation in different host niches during pathogenesis, including different fungal-specific iron-uptake mechanisms. While being necessary for virulence, they provide hope for developing novel antifungal treatments, which are currently scarce and usually toxic for patients. In this review, we provide a compilation of the current knowledge about the metabolic response to iron deficiency and excess in fungi.
Collapse
Affiliation(s)
- Jordi Pijuan
- Laboratory of Neurogenetics and Molecular MedicineInstitut de Recerca Sant Joan de DéuBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadridSpain
| | - David F. Moreno
- Department of Molecular Cellular and Developmental BiologyYale UniversityNew HavenConnecticutUSA
- Systems Biology InstituteYale UniversityWest HavenConnecticutUSA
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of PharmacyZagazig UniversityAl SharqiaEgypt
| | - Mihaela Moisa
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| | - Ihtisham Ul Haq
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Programa de Pós‐graduação em Inovação TecnológicaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Centre for Organic and Nanohybrid ElectronicsSilesian University of TechnologyGliwicePoland
| | - Rasha Mosbah
- Infection Control UnitHospitals of Zagazig UniversityZagazigEgypt
| | - Kamel Metwally
- Department of Medicinal Chemistry, Faculty of PharmacyUniversity of TabukTabukSaudi Arabia
- Department of Pharmaceutical Medicinal Chemistry, Faculty of PharmacyZagazig UniversityZagazigEgypt
| | - Simona Cavalu
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| |
Collapse
|
11
|
Alqarihi A, Kontoyiannis DP, Ibrahim AS. Mucormycosis in 2023: an update on pathogenesis and management. Front Cell Infect Microbiol 2023; 13:1254919. [PMID: 37808914 PMCID: PMC10552646 DOI: 10.3389/fcimb.2023.1254919] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus, Mucor, and Lichtheimia, accounting for > 90% of all cases. MCR is seen in patients with severe immunosuppression such as those with hematologic malignancy or transplantation, Diabetes Mellitus (DM) and diabetic ketoacidosis (DKA) and immunocompetent patients with severe wounds. The recent SARS COV2 epidemy in India has resulted in a tremendous increase in MCR cases, typically seen in the setting of uncontrolled DM and corticosteroid use. In addition to the diversity of affected hosts, MCR has pleiotropic clinical presentations, with rhino-orbital/rhino-cerebral, sino-pulmonary and necrotizing cutaneous forms being the predominant manifestations. Major insights in MCR pathogenesis have brought into focus the host receptors (GRP78) and signaling pathways (EGFR activation cascade) as well as the adhesins used by Mucorales for invasion. Furthermore, studies have expanded on the importance of iron availability and the complex regulation of iron homeostasis, as well as the pivotal role of mycotoxins as key factors for tissue invasion. The molecular toolbox to study Mucorales pathogenesis remains underdeveloped, but promise is brought by RNAi and CRISPR/Cas9 approaches. Important recent advancements have been made in early, culture-independent molecular diagnosis of MCR. However, development of new potent antifungals against Mucorales remains an unmet need. Therapy of MCR is multidisciplinary and requires a high index of suspicion for initiation of early Mucorales-active antifungals. Reversal of underlying immunosuppression, if feasible, rapid DKA correction and in selected patients, surgical debulking are crucial for improved outcomes.
Collapse
Affiliation(s)
- Abdullah Alqarihi
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Ashraf S Ibrahim
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
12
|
Shah NN, Khan Z, Ahad H, Elderdery AY, Alomary MN, Atwah B, Alhindi Z, Alsugoor MH, Elkhalifa AME, Nabi S, Bashir SM, Yaqub T, Rather GA, Ansari MA. Mucormycosis an added burden to Covid-19 Patients: An in-depth systematic review. J Infect Public Health 2022; 15:1299-1314. [PMID: 36279686 PMCID: PMC9562622 DOI: 10.1016/j.jiph.2022.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/01/2022] [Accepted: 10/10/2022] [Indexed: 01/08/2023] Open
Abstract
As of 25th July, 2022, global Disease burden of 575,430,244 confirmed cases and over 6,403,511 deaths have been attributed to coronavirus disease 2019 (COVID-19). Co-infections/secondary infections continue to plague patients around the world as result of the co-morbidities like diabetes mellitus, biochemical changes caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) especially significant elevation in free iron levels, immune suppression caused by SARS-CoV-2, and indiscriminate use of systemic corticosteroids for the treatment of severe COVID-19 disease. In such circumstances, opportunistic fungal infections pose significant challenge for COVID-19 disease therapy in patients with other co-morbidities. Although COVID-19-associated Mucormycosis (CAM) has been widely recognized, currently extensive research is being conducted on mucormycosis. It has been widely agreed that patients undergoing corticosteroid therapy are highly susceptible for CAM, henceforth high index of screening and intensive care and management is need of an hour in order to have favorable outcomes in these patients. Diagnosis in such cases is often delayed and eventually the disease progresses quickly which poses added burden to clinician and increases patient load in critical care units of hospitals. A vast perusal of literature indicated that patients with diabetes mellitus and those with other co-morbidities might be highly vulnerable to develop mucormycosis. In the present work, the case series of three patients presented at Chest Disease Hospital Srinagar, Jammu and Kashmir infected with CAM has been described with their epidemiological data in supplementary section. All these cases were found to be affected with co-morbidity of Diabetes Mellitus (DM) and were under corticosteroid therapy. Furthermore, given the significant death rate linked with mucormycosis and the growing understanding of the diseases significance, systematic review of the literature on CAM has been discussed and we have attempted to discuss emerging CAM and related aspects of the disease.
Collapse
Affiliation(s)
- Naveed Nazir Shah
- Department of Chest Medicine, Govt. Medical College Srinagar, Jammu & Kashmir, India
| | - Zaid Khan
- Department of Chest Medicine, Govt. Medical College Srinagar, Jammu & Kashmir, India
| | - Hashim Ahad
- Government Dental College, Srinagar, Jammu & Kashmir, India
| | - Abozer Y Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Saudi Arabia
| | - Mohammad N Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Banan Atwah
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Zain Alhindi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mahdi H Alsugoor
- Department of Emergency Medical Services, faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia
| | - Ahmed M E Elkhalifa
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia & Department of Haematology, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti 1158, Sudan
| | - Showket Nabi
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar, Jammu & Kashmir 190006, India
| | - Showkeen Muzamil Bashir
- Molecular biology Laboratory, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar, Jammu & Kashmir 190006, India.
| | - Tahir Yaqub
- Institute of Microbiology University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Gulzar Ahmed Rather
- Department of Biomedical Engineering, Sathyabama Institute of Science & Technology, Deemed to be University, Chennai, Tamil Nadu, India
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| |
Collapse
|
13
|
Role of Iron and Iron Overload in the Pathogenesis of Invasive Fungal Infections in Patients with Hematological Malignancies. J Clin Med 2022; 11:jcm11154457. [PMID: 35956074 PMCID: PMC9369168 DOI: 10.3390/jcm11154457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Iron is an essential trace metal necessary for the reproduction and survival of fungal pathogens. The latter have developed various mechanisms to acquire iron from their mammalian hosts, with whom they participate in a continuous struggle for dominance over iron. Invasive fungal infections are an important problem in the treatment of patients with hematological malignancies, and they are associated with significant morbidity and mortality. The diagnosis of invasive clinical infections in these patients is complex, and the treatment, which must occur as early as possible, is difficult. There are several studies that have shown a possible link between iron overload and an increased susceptibility to infections. This link is also relevant for patients with hematological malignancies and for those treated with allogeneic hematopoietic stem cell transplantation. The role of iron and its metabolism in the virulence and pathogenesis of various invasive fungal infections is intriguing, and so far, there is some evidence linking invasive fungal infections to iron or iron overload. Clarifying the possible association of iron and iron overload with susceptibility to invasive fungal infections could be important for a better prevention and treatment of these infections in patients with hematological malignancies.
Collapse
|
14
|
Simm C, Weerasinghe H, Thomas DR, Harrison PF, Newton HJ, Beilharz TH, Traven A. Disruption of Iron Homeostasis and Mitochondrial Metabolism Are Promising Targets to Inhibit Candida auris. Microbiol Spectr 2022; 10:e0010022. [PMID: 35412372 PMCID: PMC9045333 DOI: 10.1128/spectrum.00100-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/21/2022] [Indexed: 11/20/2022] Open
Abstract
Fungal infections are a global threat, but treatments are limited due to a paucity in antifungal drug targets and the emergence of drug-resistant fungi such as Candida auris. Metabolic adaptations enable microbial growth in nutrient-scarce host niches, and they further control immune responses to pathogens, thereby offering opportunities for therapeutic targeting. Because it is a relatively new pathogen, little is known about the metabolic requirements for C. auris growth and its adaptations to counter host defenses. Here, we establish that triggering metabolic dysfunction is a promising strategy against C. auris. Treatment with pyrvinium pamoate (PP) induced metabolic reprogramming and mitochondrial dysfunction evident in disrupted mitochondrial morphology and reduced tricarboxylic acid (TCA) cycle enzyme activity. PP also induced changes consistent with disrupted iron homeostasis. Nutrient supplementation experiments support the proposition that PP-induced metabolic dysfunction is driven by disrupted iron homeostasis, which compromises carbon and lipid metabolism and mitochondria. PP inhibited C. auris replication in macrophages, which is a relevant host niche for this yeast pathogen. We propose that PP causes a multipronged metabolic hit to C. auris: it restricts the micronutrient iron to potentiate nutritional immunity imposed by immune cells, and it further causes metabolic dysfunction that compromises the utilization of macronutrients, thereby curbing the metabolic plasticity needed for growth in host environments. Our study offers a new avenue for therapeutic development against drug-resistant C. auris, shows how complex metabolic dysfunction can be caused by a single compound triggering antifungal inhibition, and provides insights into the metabolic needs of C. auris in immune cell environments. IMPORTANCE Over the last decade, Candida auris has emerged as a human pathogen around the world causing life-threatening infections with wide-spread antifungal drug resistance, including pandrug resistance in some cases. In this study, we addressed the mechanism of action of the antiparasitic drug pyrvinium pamoate against C. auris and show how metabolism could be inhibited to curb C. auris proliferation. We show that pyrvinium pamoate triggers sweeping metabolic and mitochondrial changes and disrupts iron homeostasis. PP-induced metabolic dysfunction compromises the utilization of both micro- and macronutrients by C. auris and reduces its growth in vitro and in immune phagocytes. Our findings provide insights into the metabolic requirements for C. auris growth and define the mechanisms of action of pyrvinium pamoate against C. auris, demonstrating how this compound works by inhibiting the metabolic flexibility of the pathogen. As such, our study characterizes credible avenues for new antifungal approaches against C. auris.
Collapse
Affiliation(s)
- Claudia Simm
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Centre to Impact AMR, Monash University, Victoria, Australia
| | - Harshini Weerasinghe
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Centre to Impact AMR, Monash University, Victoria, Australia
| | - David R. Thomas
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - Hayley J. Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Traude H. Beilharz
- Development and Stem Cells Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
| | - Ana Traven
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Centre to Impact AMR, Monash University, Victoria, Australia
| |
Collapse
|
15
|
Anand C B, Senthilkumar S, P N, Ibrahim C M, Afroze M KH, M R. Estimation of Serum Ferritin in Mucormycosis Patients and Prognostication Based on the Ferritin Value. Cureus 2022; 14:e24013. [PMID: 35547446 PMCID: PMC9090233 DOI: 10.7759/cureus.24013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2022] [Indexed: 12/15/2022] Open
|
16
|
Pushparaj K, Kuchi Bhotla H, Arumugam VA, Pappusamy M, Easwaran M, Liu WC, Issara U, Rengasamy KRR, Meyyazhagan A, Balasubramanian B. Mucormycosis (black fungus) ensuing COVID-19 and comorbidity meets - Magnifying global pandemic grieve and catastrophe begins. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 805:150355. [PMID: 34818767 PMCID: PMC8443313 DOI: 10.1016/j.scitotenv.2021.150355] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/25/2021] [Accepted: 09/11/2021] [Indexed: 05/02/2023]
Abstract
Post COVID-19, mucormycosis occurred after the SARS-CoV-2 has rampaged the human population and is a scorching problem among the pandemic globally, particularly among Asian countries. Invasive mucormycosis has been extensively reported from mild to severe COVID-19 survivors. The robust predisposing factor seems to be uncontrolled diabetes mellitus, comorbidity and immunosuppression acquired through steroid therapy. The prime susceptive reason for the increase of mucormycosis cases is elevated iron levels in the serum of the COVID survivors. A panoramic understanding of the infection has been elucidated based on clinical manifestation, genetic and non- genetic mechanisms of steroid drug administration, biochemical pathways and immune modulated receptor associations. This review lime-lights and addresses the "What", "Why", "How" and "When" about the COVID-19 associated mucormycosis (CAM) in a comprehensive manner with a pure intention to bring about awareness to the common public as the cases are inevitably and exponentially increasing in India and global countries as well. The article also unearthed the pathogenesis of mucormycosis and its association with the COVID-19 sequela, the plausible routes of entry, diagnosis and counter remedies to keep the infection at bay. Cohorts of case reports were analysed to spotlight the link between the pandemic COVID-19 and the nightmare-mucormycosis.
Collapse
Affiliation(s)
- Karthika Pushparaj
- Department of Zoology, School of Biosciences, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641 043, Tamil Nadu, India
| | - Haripriya Kuchi Bhotla
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru, Karnataka 560076, India
| | - Vijaya Anand Arumugam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Manikantan Pappusamy
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru, Karnataka 560076, India
| | - Murugesh Easwaran
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Wen-Chao Liu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Utthapon Issara
- Division of Food Science and Technology Management, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi, Pathum Thani 12110, Thailand
| | - Kannan R R Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Private Bag X1106, Polokwane, Sovenga 0727, South Africa
| | - Arun Meyyazhagan
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru, Karnataka 560076, India.
| | | |
Collapse
|
17
|
Stanford FA, Matthies N, Cseresnyés Z, Figge MT, Hassan MIA, Voigt K. Expression Patterns in Reductive Iron Assimilation and Functional Consequences during Phagocytosis of Lichtheimia corymbifera, an Emerging Cause of Mucormycosis. J Fungi (Basel) 2021; 7:jof7040272. [PMID: 33916756 PMCID: PMC8065604 DOI: 10.3390/jof7040272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022] Open
Abstract
Iron is an essential micronutrient for most organisms and fungi are no exception. Iron uptake by fungi is facilitated by receptor-mediated internalization of siderophores, heme and reductive iron assimilation (RIA). The RIA employs three protein groups: (i) the ferric reductases (Fre5 proteins), (ii) the multicopper ferroxidases (Fet3) and (iii) the high-affinity iron permeases (Ftr1). Phenotyping under different iron concentrations revealed detrimental effects on spore swelling and hyphal formation under iron depletion, but yeast-like morphology under iron excess. Since access to iron is limited during pathogenesis, pathogens are placed under stress due to nutrient limitations. To combat this, gene duplication and differential gene expression of key iron uptake genes are utilized to acquire iron against the deleterious effects of iron depletion. In the genome of the human pathogenic fungus L. corymbifera, three, four and three copies were identified for FRE5, FTR1 and FET3 genes, respectively. As in other fungi, FET3 and FTR1 are syntenic and co-expressed in L. corymbifera. Expression of FRE5, FTR1 and FET3 genes is highly up-regulated during iron limitation (Fe-), but lower during iron excess (Fe+). Fe- dependent upregulation of gene expression takes place in LcFRE5 II and III, LcFTR1 I and II, as well as LcFET3 I and II suggesting a functional role in pathogenesis. The syntenic LcFTR1 I–LcFET3 I gene pair is co-expressed during germination, whereas LcFTR1 II- LcFET3 II is co-expressed during hyphal proliferation. LcFTR1 I, II and IV were overexpressed in Saccharomyces cerevisiae to represent high and moderate expression of intracellular transport of Fe3+, respectively. Challenge of macrophages with the yeast mutants revealed no obvious role for LcFTR1 I, but possible functions of LcFTR1 II and IVs in recognition by macrophages. RIA expression pattern was used for a new model of interaction between L. corymbifera and macrophages.
Collapse
Affiliation(s)
- Felicia Adelina Stanford
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Nina Matthies
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute, 12622 Jena, Germany;
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- Applied Systems Biology, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute, 12622 Jena, Germany;
| | - Mohamed I. Abdelwahab Hassan
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- National Research Centre, Pests & Plant Protection Department, 33rd El Buhouth St., Dokki, Giza 12622, Egypt
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- Correspondence: or ; Tel.: +49-3641-532-1395
| |
Collapse
|
18
|
Robinson JR, Isikhuemhen OS, Anike FN. Fungal-Metal Interactions: A Review of Toxicity and Homeostasis. J Fungi (Basel) 2021; 7:225. [PMID: 33803838 PMCID: PMC8003315 DOI: 10.3390/jof7030225] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
Metal nanoparticles used as antifungals have increased the occurrence of fungal-metal interactions. However, there is a lack of knowledge about how these interactions cause genomic and physiological changes, which can produce fungal superbugs. Despite interest in these interactions, there is limited understanding of resistance mechanisms in most fungi studied until now. We highlight the current knowledge of fungal homeostasis of zinc, copper, iron, manganese, and silver to comprehensively examine associated mechanisms of resistance. Such mechanisms have been widely studied in Saccharomyces cerevisiae, but limited reports exist in filamentous fungi, though they are frequently the subject of nanoparticle biosynthesis and targets of antifungal metals. In most cases, microarray analyses uncovered resistance mechanisms as a response to metal exposure. In yeast, metal resistance is mainly due to the down-regulation of metal ion importers, utilization of metallothionein and metallothionein-like structures, and ion sequestration to the vacuole. In contrast, metal resistance in filamentous fungi heavily relies upon cellular ion export. However, there are instances of resistance that utilized vacuole sequestration, ion metallothionein, and chelator binding, deleting a metal ion importer, and ion storage in hyphal cell walls. In general, resistance to zinc, copper, iron, and manganese is extensively reported in yeast and partially known in filamentous fungi; and silver resistance lacks comprehensive understanding in both.
Collapse
Affiliation(s)
| | - Omoanghe S. Isikhuemhen
- Department of Natural Resources and Environmental Design, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, NC 27411, USA; (J.R.R.); (F.N.A.)
| | | |
Collapse
|
19
|
Shanmugam A, Chithiravel V, Gunasekar A, Venkattappan A. Siderophores in Antifungal Drug Discovery: A Computational Approach. Fungal Biol 2021. [DOI: 10.1007/978-3-030-53077-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
20
|
Weissman Z, Pinsky M, Donegan RK, Reddi AR, Kornitzer D. Using genetically encoded heme sensors to probe the mechanisms of heme uptake and homeostasis in Candida albicans. Cell Microbiol 2020; 23:e13282. [PMID: 33104284 DOI: 10.1111/cmi.13282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 11/30/2022]
Abstract
Candida albicans is a major fungal pathogen that can utilise hemin and haemoglobin as iron sources in the iron-scarce host environment. While C. albicans is a heme prototroph, we show here that it can also efficiently utilise external heme as a cellular heme source. Using genetically encoded ratiometric fluorescent heme sensors, we show that heme extracted from haemoglobin and free hemin enter the cells with different kinetics. Heme supplied as haemoglobin is taken up via the Common in Fungal Extracellular Membrane (CFEM) hemophore cascade, and reaches the cytoplasm over several hours, whereas entry of free hemin via CFEM-dependent and independent pathways is much faster, less than an hour. To prevent an influx of extracellular heme from reaching toxic levels in the cytoplasm, the cells deploy Hmx1, a heme oxygenase. Hmx1 was previously suggested to be involved in utilisation of haemoglobin and hemin as iron sources, but we find that it is primarily required to prevent heme toxicity. Taken together, the combination of novel heme sensors with genetic analysis revealed new details of the fungal mechanisms of heme import and homeostasis, necessary to balance the uses of heme as essential cofactor and potential iron source against its toxicity.
Collapse
Affiliation(s)
- Ziva Weissman
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion-I.I.T., Haifa, Israel
| | - Mariel Pinsky
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion-I.I.T., Haifa, Israel
| | - Rebecca K Donegan
- School of Chemistry and Biochemistry and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Amit R Reddi
- School of Chemistry and Biochemistry and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion-I.I.T., Haifa, Israel
| |
Collapse
|
21
|
Stanford FA, Voigt K. Iron Assimilation during Emerging Infections Caused by Opportunistic Fungi with emphasis on Mucorales and the Development of Antifungal Resistance. Genes (Basel) 2020; 11:genes11111296. [PMID: 33143139 PMCID: PMC7693903 DOI: 10.3390/genes11111296] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Iron is a key transition metal required by most microorganisms and is prominently utilised in the transfer of electrons during metabolic reactions. The acquisition of iron is essential and becomes a crucial pathogenic event for opportunistic fungi. Iron is not readily available in the natural environment as it exists in its insoluble ferric form, i.e., in oxides and hydroxides. During infection, the host iron is bound to proteins such as transferrin, ferritin, and haemoglobin. As such, access to iron is one of the major hurdles that fungal pathogens must overcome in an immunocompromised host. Thus, these opportunistic fungi utilise three major iron acquisition systems to overcome this limiting factor for growth and proliferation. To date, numerous iron acquisition pathways have been fully characterised, with key components of these systems having major roles in virulence. Most recently, proteins involved in these pathways have been linked to the development of antifungal resistance. Here, we provide a detailed review of our current knowledge of iron acquisition in opportunistic fungi, and the role iron may have on the development of resistance to antifungals with emphasis on species of the fungal basal lineage order Mucorales, the causative agents of mucormycosis.
Collapse
Affiliation(s)
- Felicia Adelina Stanford
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology–Hans Knöll Institute, Jena, Adolf-Reichwein-Straße 23, 07745 Jena, Germany;
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller University Jena, Neugasse 25, 07743 Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology–Hans Knöll Institute, Jena, Adolf-Reichwein-Straße 23, 07745 Jena, Germany;
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller University Jena, Neugasse 25, 07743 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena Microbial Resource Collection Adolf-Reichwein-Straße 23, 07745 Jena, Germany
- Correspondence: ; Tel.: +49-3641-532-1395; Fax: +49-3641-532-2395
| |
Collapse
|
22
|
Kornitzer D, Roy U. Pathways of heme utilization in fungi. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118817. [PMID: 32777371 DOI: 10.1016/j.bbamcr.2020.118817] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Iron acquisition is challenging in most environments. As an alternative to elemental iron, organisms can take up iron-protoporphyrin IX, or heme. Heme can be found in decaying organic matter and is particularly prevalent in animal hosts. Fungi have evolved at least three distinct endocytosis-mediated heme uptake systems, which have been studied in detail in the organisms Candida albicans, Cryptococcus neoformans and Schizosaccharomyces pombe. Here we summarize the known molecular details of these three uptake systems that enable parasitic and saprophytic fungi to take advantage of external heme as either cellular iron or heme sources.
Collapse
Affiliation(s)
- Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| | - Udita Roy
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
23
|
Martínez-Pastor MT, Puig S. Adaptation to iron deficiency in human pathogenic fungi. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118797. [PMID: 32663505 DOI: 10.1016/j.bbamcr.2020.118797] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/13/2020] [Accepted: 07/05/2020] [Indexed: 02/08/2023]
Abstract
Iron is an essential micronutrient for virtually all eukaryotic organisms and plays a central role during microbial infections. Invasive fungal diseases are associated with strikingly high rates of mortality, but their impact on human health is usually underestimated. Upon a fungal infection, hosts restrict iron availability in order to limit the growth and virulence of the pathogen. Here, we use two model yeasts, Saccharomyces cerevisiae and Schizosaccharomyces pombe, to delve into the response to iron deficiency of human fungal pathogens, such as Candida glabrata, Candida albicans, Aspergillus fumigatus and Cryptococcus neoformans. Fungi possess common and species-specific mechanisms to acquire iron and to control the response to iron limitation. Upon iron scarcity, fungi activate a wide range of elegant strategies to capture and import exogenous iron, mobilize iron from intracellular stores, and modulate their metabolism to economize and prioritize iron utilization. Hence, iron homeostasis genes represent remarkable virulence factors that can be used as targets for the development of novel antifungal treatments.
Collapse
Affiliation(s)
| | - Sergi Puig
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Paterna, Valencia, Spain.
| |
Collapse
|
24
|
Duval C, Macabiou C, Garcia C, Lesuisse E, Camadro J, Auchère F. The adaptive response to iron involves changes in energetic strategies in the pathogen Candida albicans. Microbiologyopen 2020; 9:e970. [PMID: 31788966 PMCID: PMC7002100 DOI: 10.1002/mbo3.970] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/22/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
Candida albicans is an opportunist pathogen responsible for a large spectrum of infections, from superficial mycosis to systemic diseases known as candidiasis. Its ability to grow in different morphological forms, such as yeasts or filamentous hyphae, contributes to its survival in diverse microenvironments. Iron uptake has been associated with virulence, and C. albicans has developed elaborate strategies for acquiring iron from its host. In this work, we analyze the metabolic changes in response to changes in iron content in the growth medium and compare C. albicans adaptation to the presence or absence of iron. Functional and morphological studies, correlated to a quantitative proteomic analysis, were performed to assess the specific pathways underlying the response to iron, both in the yeast and filamentous forms. Overall, the results show that the adaptive response to iron is associated with a metabolic remodeling affecting the energetic pathways of the pathogen. This includes changes in the thiol-dependent redox status, the activity of key mitochondrial enzymes and the respiratory chain. Iron deficiency stimulates bioenergetic pathways, whereas iron-rich condition is associated with greater biosynthetic needs, particularly in filamentous forms. Moreover, we found that C. albicans yeast cells have an extraordinary capability to adapt to changes in environmental conditions.
Collapse
Affiliation(s)
- Celia Duval
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Carole Macabiou
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Camille Garcia
- Plateforme Protéomique structurale et fonctionnelle/Spectrométrie de masseInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Emmanuel Lesuisse
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Jean‐Michel Camadro
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Françoise Auchère
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| |
Collapse
|
25
|
Heme-iron acquisition in fungi. Curr Opin Microbiol 2019; 52:77-83. [DOI: 10.1016/j.mib.2019.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 01/09/2023]
|
26
|
Fourie R, Kuloyo OO, Mochochoko BM, Albertyn J, Pohl CH. Iron at the Centre of Candida albicans Interactions. Front Cell Infect Microbiol 2018; 8:185. [PMID: 29922600 PMCID: PMC5996042 DOI: 10.3389/fcimb.2018.00185] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/14/2018] [Indexed: 12/27/2022] Open
Abstract
Iron is an absolute requirement for both the host and most pathogens alike and is needed for normal cellular growth. The acquisition of iron by biological systems is regulated to circumvent toxicity of iron overload, as well as the growth deficits imposed by iron deficiency. In addition, hosts, such as humans, need to limit the availability of iron to pathogens. However, opportunistic pathogens such as Candida albicans are able to adapt to extremes of iron availability, such as the iron replete environment of the gastrointestinal tract and iron deficiency during systemic infection. C. albicans has developed a complex and effective regulatory circuit for iron acquisition and storage to circumvent iron limitation within the human host. As C. albicans can form complex interactions with both commensal and pathogenic co-inhabitants, it can be speculated that iron may play an important role in these interactions. In this review, we highlight host iron regulation as well as regulation of iron homeostasis in C. albicans. In addition, the review argues for the need for further research into the role of iron in polymicrobial interactions. Lastly, the role of iron in treatment of C. albicans infection is discussed.
Collapse
Affiliation(s)
- Ruan Fourie
- Pathogenic Yeast Research Group, Department of Microbial, Biochemical and Food Biotechnology, University of the Free State, Bloemfontein, South Africa
| | - Oluwasegun O Kuloyo
- Pathogenic Yeast Research Group, Department of Microbial, Biochemical and Food Biotechnology, University of the Free State, Bloemfontein, South Africa
| | - Bonang M Mochochoko
- Pathogenic Yeast Research Group, Department of Microbial, Biochemical and Food Biotechnology, University of the Free State, Bloemfontein, South Africa
| | - Jacobus Albertyn
- Pathogenic Yeast Research Group, Department of Microbial, Biochemical and Food Biotechnology, University of the Free State, Bloemfontein, South Africa
| | - Carolina H Pohl
- Pathogenic Yeast Research Group, Department of Microbial, Biochemical and Food Biotechnology, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
27
|
Gerwien F, Skrahina V, Kasper L, Hube B, Brunke S. Metals in fungal virulence. FEMS Microbiol Rev 2018; 42:4562650. [PMID: 29069482 PMCID: PMC5812535 DOI: 10.1093/femsre/fux050] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/19/2017] [Indexed: 12/25/2022] Open
Abstract
Metals are essential for life, and they play a central role in the struggle between infecting microbes and their hosts. In fact, an important aspect of microbial pathogenesis is the 'nutritional immunity', in which metals are actively restricted (or, in an extended definition of the term, locally enriched) by the host to hinder microbial growth and virulence. Consequently, fungi have evolved often complex regulatory networks, uptake and detoxification systems for essential metals such as iron, zinc, copper, nickel and manganese. These systems often differ fundamentally from their bacterial counterparts, but even within the fungal pathogens we can find common and unique solutions to maintain metal homeostasis. Thus, we here compare the common and species-specific mechanisms used for different metals among different fungal species-focusing on important human pathogens such as Candida albicans, Aspergillus fumigatus or Cryptococcus neoformans, but also looking at model fungi such as Saccharomyces cerevisiae or A. nidulans as well-studied examples for the underlying principles. These direct comparisons of our current knowledge reveal that we have a good understanding how model fungal pathogens take up iron or zinc, but that much is still to learn about other metals and specific adaptations of individual species-not the least to exploit this knowledge for new antifungal strategies.
Collapse
Affiliation(s)
- Franziska Gerwien
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Volha Skrahina
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Lydia Kasper
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Bernhard Hube
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Sascha Brunke
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| |
Collapse
|
28
|
Sherrington SL, Kumwenda P, Kousser C, Hall RA. Host Sensing by Pathogenic Fungi. ADVANCES IN APPLIED MICROBIOLOGY 2017; 102:159-221. [PMID: 29680125 DOI: 10.1016/bs.aambs.2017.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability to cause disease extends from the ability to grow within the host environment. The human host provides a dynamic environment to which fungal pathogens must adapt to in order to survive. The ability to grow under a particular condition (i.e., the ability to grow at mammalian body temperature) is considered a fitness attribute and is essential for growth within the human host. On the other hand, some environmental conditions activate signaling mechanisms resulting in the expression of virulence factors, which aid pathogenicity. Therefore, pathogenic fungi have evolved fitness and virulence attributes to enable them to colonize and infect humans. This review highlights how some of the major pathogenic fungi respond and adapt to key environmental signals within the human host.
Collapse
Affiliation(s)
- Sarah L Sherrington
- Institute for Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Pizga Kumwenda
- Institute for Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Courtney Kousser
- Institute for Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Rebecca A Hall
- Institute for Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
29
|
Skrahina V, Brock M, Hube B, Brunke S. Candida albicans Hap43 Domains Are Required under Iron Starvation but Not Excess. Front Microbiol 2017; 8:2388. [PMID: 29250054 PMCID: PMC5717023 DOI: 10.3389/fmicb.2017.02388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022] Open
Abstract
Iron availability is a central factor in infections, since iron is a critical micronutrient for all living organisms. The host employs both iron limitation and toxicity strategies to control microbial growth, and successful pathogens are able to tightly coordinate iron homeostasis in response to changing iron levels. As a commensal and opportunistic pathogen, Candida albicans copes with both iron deficiency and excess via the precise regulation of iron acquisition, consumption and storage. The C. albicans transcription factor Hap43 is known to be required for the iron starvation response, while specific domains of its ortholog, HapX, in Aspergillus fumigatus, were recently shown to regulate iron uptake and consumptions genes under both low and high iron levels. Therefore, we investigated the contribution of C. albicans Hap43 domains in response to changing iron levels. We found the C-terminus of Hap43 to be essential for the activation of iron uptake genes during iron starvation, whereas, in contrast to A. fumigatus, Hap43 was not required in mediating adaptation to iron resistance. These data indicate that the generally conserved metal acquisition systems in fungal pathogens can show individual adaptations to the host environment.
Collapse
Affiliation(s)
- Volha Skrahina
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Matthias Brock
- Fungal Genetics and Biology Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Friedrich Schiller University, Jena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| |
Collapse
|
30
|
Abstract
The devastating infections that fungal pathogens cause in humans are underappreciated relative to viral, bacterial and parasitic diseases. In recent years, the contributions to virulence of reductive iron uptake, siderophore-mediated uptake and heme acquisition have been identified in the best studied and most life-threatening fungal pathogens: Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus. In particular, exciting new work illustrates the importance of iron acquisition from heme and hemoglobin in the virulence of pathogenic yeasts. However, the challenge of establishing how these fungi gain access to hemoglobin in blood and to other sources of heme remains to be fully addressed. Recent studies are also expanding our knowledge of iron uptake in less-well studied fungal pathogens, including dimorphic fungi where new information reveals an integration of iron acquisition with morphogenesis and cell-surface properties for adhesion to host cells. Overall, the accumulating information provides opportunities to exploit iron acquisition for antifungal therapy, and new work highlights the development of specific inhibitors of siderophore biosynthesis and metal chelators for therapeutic use alone or in conjunction with existing antifungal drugs. It is clear that iron-related therapies will need to be customized for specific diseases because the emerging view is that fungal pathogens use different combinations of strategies for iron acquisition in the varied niches of vertebrate hosts.
Collapse
Affiliation(s)
- Gaurav Bairwa
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | - Won Hee Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 456-756, Republic of Korea
| | - James W Kronstad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
31
|
Hernández-Chávez MJ, Pérez-García LA, Niño-Vega GA, Mora-Montes HM. Fungal Strategies to Evade the Host Immune Recognition. J Fungi (Basel) 2017; 3:jof3040051. [PMID: 29371567 PMCID: PMC5753153 DOI: 10.3390/jof3040051] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 12/23/2022] Open
Abstract
The recognition of fungal cells by the host immune system is key during the establishment of a protective anti-fungal response. Even though the immune system has evolved a vast number of processes to control these organisms, they have developed strategies to fight back, avoiding the proper recognition by immune components and thus interfering with the host protective mechanisms. Therefore, the strategies to evade the immune system are as important as the virulence factors and attributes that damage the host tissues and cells. Here, we performed a thorough revision of the main fungal tactics to escape from the host immunosurveillance processes. These include the composition and organization of the cell wall, the fungal capsule, the formation of titan cells, biofilms, and asteroid bodies; the ability to undergo dimorphism; and the escape from nutritional immunity, extracellular traps, phagocytosis, and the action of humoral immune effectors.
Collapse
Affiliation(s)
- Marco J Hernández-Chávez
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P., Guanajuato Gto. 36050, México.
| | - Luis A Pérez-García
- Unidad Académica Multidisciplinaria Zona Huasteca, Universidad Autónoma de San Luis Potosí, Romualdo del Campo 501, Fracc. Rafael Curiel, C.P., Cd. Valle SLP. 79060, México.
| | - Gustavo A Niño-Vega
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P., Guanajuato Gto. 36050, México.
| | - Héctor M Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P., Guanajuato Gto. 36050, México.
| |
Collapse
|
32
|
Gerwien F, Safyan A, Wisgott S, Brunke S, Kasper L, Hube B. The Fungal Pathogen Candida glabrata Does Not Depend on Surface Ferric Reductases for Iron Acquisition. Front Microbiol 2017. [PMID: 28642757 PMCID: PMC5463049 DOI: 10.3389/fmicb.2017.01055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Iron acquisition is a crucial virulence determinant for many bacteria and fungi, including the opportunistic fungal pathogens Candida albicans and C. glabrata. While the diverse strategies used by C. albicans for obtaining iron from the host are well-described, much less is known about the acquisition of this micronutrient from host sources by C. glabrata – a distant relative of C. albicans with closer evolutionary ties to Saccharomyces cerevisiae, which nonetheless causes severe clinical symptoms in humans. Here we show that C. glabrata is much more restricted than C. albicans in using host iron sources, lacking, for example, the ability to grow on transferrin and hemin/hemoglobin. Instead, C. glabrata is able to use ferritin and non-protein-bound iron (FeCl3) as iron sources in a pH-dependent manner. As in other fungal pathogens, iron-dependent growth requires the reductive high affinity (HA) iron uptake system. Typically highly conserved, this uptake mechanism normally relies on initial ferric reduction by cell-surface ferric reductases. The C. glabrata genome contains only three such putative ferric reductases, which were found to be dispensable for iron-dependent growth. In addition and in contrast to C. albicans and S. cerevisiae, we also detected no surface ferric reductase activity in C. glabrata. Instead, extracellular ferric reduction was found in this and the two other fungal species, which was largely dependent on an excreted low-molecular weight, non-protein ferric reductant. We therefore propose an iron acquisition strategy of C. glabrata which differs from other pathogenic fungi, such as C. albicans, in that it depends on a limited set of host iron sources and that it lacks the need for surface ferric reductases. Extracellular ferric reduction by a secreted molecule possibly compensates for the loss of surface ferric reductase activity in the HA iron uptake system.
Collapse
Affiliation(s)
- Franziska Gerwien
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Abu Safyan
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Stephanie Wisgott
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany.,Department of Microbial Pathogenicity Mechanisms, Friedrich Schiller UniversityJena, Germany.,Center for Sepsis Control and Care, University HospitalJena, Germany
| |
Collapse
|
33
|
Mourer T, Normant V, Labbé S. Heme Assimilation in Schizosaccharomyces pombe Requires Cell-surface-anchored Protein Shu1 and Vacuolar Transporter Abc3. J Biol Chem 2017; 292:4898-4912. [PMID: 28193844 PMCID: PMC5377804 DOI: 10.1074/jbc.m117.776807] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/08/2017] [Indexed: 12/12/2022] Open
Abstract
The Schizosaccharomyces pombe shu1+ gene encodes a cell-surface protein required for assimilation of exogenous heme. In this study, shaving experiments showed that Shu1 is released from membrane preparations when spheroplast lysates are incubated with phosphoinositide-specific phospholipase C (PI-PLC). Shu1 cleavability by PI-PLC and its predicted hydropathy profile strongly suggested that Shu1 is a glycosylphosphatidylinositol-anchored protein. When heme biosynthesis is selectively blocked in hem1Δ mutant cells, the heme analog zinc mesoporphyrin IX (ZnMP) first accumulates into vacuoles and then subsequently, within the cytoplasm in a rapid and Shu1-dependent manner. An HA4-tagged shu1+ allele that retained wild-type function localizes to the cell surface in response to low hemin concentrations, but under high hemin concentrations, Shu1-HA4 re-localizes to the vacuolar membrane. Inactivation of abc3+, encoding a vacuolar membrane transporter, results in hem1Δ abc3Δ mutant cells being unable to grow in the presence of hemin as the sole iron source. In hem1Δ abc3Δ cells, ZnMP accumulates primarily in vacuoles and does not sequentially accumulate in the cytosol. Consistent with a role for Abc3 as vacuolar hemin exporter, results with hemin-agarose pulldown assays showed that Abc3 binds to hemin. In contrast, an Abc3 mutant in which an inverted Cys-Pro motif had been replaced with Ala residues fails to bind hemin with high affinity. Taken together, these results show that Shu1 undergoes rapid hemin-induced internalization from the cell surface to the vacuolar membrane and that the transporter Abc3 participates in the mobilization of stored heme from the vacuole to the cytosol.
Collapse
Affiliation(s)
- Thierry Mourer
- From the Département de Biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | - Vincent Normant
- From the Département de Biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | - Simon Labbé
- From the Département de Biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| |
Collapse
|
34
|
Hemolytic capability and expression of a putative haem oxygenase-encoding gene by blood isolates of Candida tropicalis are influenced by iron deprivation and the presence of hemoglobin and erythrocytes. Microb Pathog 2017; 105:235-239. [PMID: 28254443 DOI: 10.1016/j.micpath.2017.02.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/31/2017] [Accepted: 02/26/2017] [Indexed: 11/22/2022]
Abstract
Although hemolytic activity is known to be a putative virulence factor contributing to candidal pathogenesis, its production by Candida tropicalis, a species closely related to Candida albicans, is poor understood. The present study was undertaken to evaluate the hemolytic activity and the expression level of a putative haem oxygenase encoding gene by blood isolates of C. tropicalis following growth in iron deprivation, and in the presence of hemoglobin and erythrocytes. The lowest values of hemolytic activity were observed in cell-free culture supernatants of isolates growing in iron-restricted medium (RPMI medium and RPMI medium supplemented with iron chelator bathophenanthrolindisulphonic acid). Hemolysis was increased in the presence of either hemoglobin or erythrocytes. Reverse transcriptase PCR analysis showed that the putative haem oxygenase encoding gene (CtHMX1), potentially related with iron uptake, was up-regulated (p < 0.001) following growth in iron deprivation and in the presence of hemoglobin; CtHMX1 was repressed in the presence of human erythrocytes (p < 0.001). Our data suggest that hemoglobin had positive effect in the production of hemolytic factor and gene expression related to iron uptake in C. tropicalis.
Collapse
|
35
|
Malavia D, Crawford A, Wilson D. Nutritional Immunity and Fungal Pathogenesis: The Struggle for Micronutrients at the Host-Pathogen Interface. Adv Microb Physiol 2017; 70:85-103. [PMID: 28528652 DOI: 10.1016/bs.ampbs.2017.01.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
All living organisms require certain micronutrients such as iron, zinc, manganese and copper for cellular function and growth. For human pathogens however, the maintenance of metal ion homeostasis is particularly challenging. This is because the mammalian host actively enforces extremes of micronutrient availability on potential microbial invaders-processes collectively termed nutritional immunity. The role of iron sequestration in controlling microbial infections is well established and, more recently, the importance of other metals including zinc, manganese and copper has been recognised. In this chapter, we explore the nutritional immune mechanisms that defend the human body against fungal infections and the strategies that these important pathogens exploit to counteract nutritional immunity and thrive in the infected host.
Collapse
Affiliation(s)
- Dhara Malavia
- Aberdeen Fungal Group, MRC Centre for Medical Mycology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, United Kingdom
| | - Aaron Crawford
- Aberdeen Fungal Group, MRC Centre for Medical Mycology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, United Kingdom
| | - Duncan Wilson
- Aberdeen Fungal Group, MRC Centre for Medical Mycology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, United Kingdom.
| |
Collapse
|
36
|
Disruptions of the genes involved in lysine biosynthesis, iron acquisition, and secondary metabolisms affect virulence and fitness in Metarhizium robertsii. Fungal Genet Biol 2016; 98:23-34. [PMID: 27876630 DOI: 10.1016/j.fgb.2016.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 11/14/2016] [Accepted: 11/18/2016] [Indexed: 01/07/2023]
Abstract
Based on genomic analysis, polyketide synthase (PKS) and nonribosomal peptide synthetase (NRPS) pathways account for biosynthesis of the majority of the secondary metabolites produced by the entomopathogenic fungus Metarhizium robertsii. To evaluate the contribution of these pathways to M. robertsii fitness and/or virulence, mutants deleted for mrpptA, the Sfp-type 4' phosphopantetheinyl transferase gene required for their activation were generated. ΔmrpptA strains were deficient in PKS and NRPS activity resulting in colonies that lacked the typical green pigment and failed to produce the nonribosomal peptides (destruxins, serinocylins, and the siderophores ferricrocin and metachelins) as well as the hybrid polyketide-peptides (NG-39x) that are all produced by the wild type (WT) M. robertsii. The ΔmrpptA colonies were also auxotrophic for lysine. Two other mutant strains were generated: ΔmraarA, in which the α-aminoadipate reductase gene critical for lysine biosynthesis was disrupted, and ΔmrsidA, in which the L-ornithine N5-oxygenase gene that is critical for hydroxamate siderophore biosynthesis was disrupted. The phenotypes of these mutants were compared to those of ΔmrpptA to separate effects of the loss of lysine or siderophore production from the overall effect of losing all polyketide and non-ribosomal peptide production. Loss of lysine biosynthesis marginally increased resistance to H2O2 while it had little effect on the sensitivity to the cell wall disruptor sodium dodecyl sulfate (SDS) and no effect on sensitivity to iron deprivation. In contrast, combined loss of metachelin and ferricrocin through the inactivation of mrsidA resulted in mutants that were as hypersensitive or slightly more sensitive to H2O2, iron deprivation, and SDS, and were either identical or marginally higher in ΔmrpptA strains. In contrast to ΔmrpptA, loss of mrsidA did not completely abolish siderophore activity, which suggests the production of one or more non-hydroxamate iron-chelating compounds. Deletion of mrpptA, mrsidA, and mraarA reduced conidium production and conidia of a GFP-tagged ΔmrpptA strain displayed a longer germination delay than WT on insect cuticles, a deficiency that was rescued by lysine supplementation. Compared with WT, ΔmrpptA strains displayed ∼19-fold reduction in virulence against Drosophila suzukii. In contrast, lysine auxotrophy and loss of siderophores accounted for ∼2 and ∼6-fold decreases in virulence, respectively. Deletion of mrpptA had no significant effect on growth inhibition of Bacillus cereus. Our results suggest that PKS and NRPS metabolism plays a significant role in M. robertsii virulence, depresses conidium production, and contributes marginally to resistance to oxidative stress and iron homeostasis, but has no significant antibacterial effect.
Collapse
|
37
|
Balhara M, Chaudhary R, Ruhil S, Singh B, Dahiya N, Parmar VS, Jaiwal PK, Chhillar AK. Siderophores; iron scavengers: the novel & promising targets for pathogen specific antifungal therapy. Expert Opin Ther Targets 2016; 20:1477-1489. [PMID: 27797604 DOI: 10.1080/14728222.2016.1254196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The recent emergence of resistance, toxicity paradigm and limited efficacy of conventional antifungal drugs necessitate the identification of de novo targets in fungal metabolism. One of the most critical physiological processes during in vivo pathogenesis is maintenance of iron homeostasis. The most life threatening opportunistic human fungal pathogens like Aspergillus, Candida and Cryptococcus exploit the siderophore mediated iron uptake mechanism either for survival, virulence, propagation or resistance to oxidative stress envisaged in vivo during infection. Areas covered: In this review, we will highlight the metabolic pathways; specifically siderophore biosynthesis, uptake and utilisation, triggered in the fungal pathogens in iron starving conditions and the various putative targets viable in these pathways to be recruited as novel therapeutic antidotes either via biosynthetic enzymes catalytic site inhibitors or as drug conjugates through trojan horse approach and further role in the development of fungal specific reliable diagnostic markers. Expert opinion: Siderophores are the weapons released by a pathogen to conquer the battle for iron acquisition. Hence, the fungal siderophore biosynthetic pathways along with their uptake and utilisation mechanisms represent an ideal target for pathogen specific, host friendly therapeutic strategy which would block the proliferation of parasite without causing any harm to the mammalian host.
Collapse
Affiliation(s)
- Meenakshi Balhara
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Renu Chaudhary
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Sonam Ruhil
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Bharat Singh
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Nisha Dahiya
- b Division of Epidemiology and Communicable Diseases , Indian Council of Medical Research , Delhi , India
| | - Virinder S Parmar
- c Bioorganic Laboratory, Department of Chemistry , University of Delhi , Delhi , India
| | - Pawan K Jaiwal
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Anil K Chhillar
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| |
Collapse
|
38
|
Polvi EJ, Li X, O’Meara TR, Leach MD, Cowen LE. Opportunistic yeast pathogens: reservoirs, virulence mechanisms, and therapeutic strategies. Cell Mol Life Sci 2015; 72:2261-87. [PMID: 25700837 PMCID: PMC11113693 DOI: 10.1007/s00018-015-1860-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 12/21/2022]
Abstract
Life-threatening invasive fungal infections are becoming increasingly common, at least in part due to the prevalence of medical interventions resulting in immunosuppression. Opportunistic fungal pathogens of humans exploit hosts that are immunocompromised, whether by immunosuppression or genetic predisposition, with infections originating from either commensal or environmental sources. Fungal pathogens are armed with an arsenal of traits that promote pathogenesis, including the ability to survive host physiological conditions and to switch between different morphological states. Despite the profound impact of fungal pathogens on human health worldwide, diagnostic strategies remain crude and treatment options are limited, with resistance to antifungal drugs on the rise. This review will focus on the global burden of fungal infections, the reservoirs of these pathogens, the traits of opportunistic yeast that lead to pathogenesis, host genetic susceptibilities, and the challenges that must be overcome to combat antifungal drug resistance and improve clinical outcome.
Collapse
Affiliation(s)
- Elizabeth J. Polvi
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 4368, Toronto, ON M5S 1A8 Canada
| | - Xinliu Li
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 4368, Toronto, ON M5S 1A8 Canada
| | - Teresa R. O’Meara
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 4368, Toronto, ON M5S 1A8 Canada
| | - Michelle D. Leach
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 4368, Toronto, ON M5S 1A8 Canada
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 4368, Toronto, ON M5S 1A8 Canada
| |
Collapse
|
39
|
Mourer T, Jacques JF, Brault A, Bisaillon M, Labbé S. Shu1 is a cell-surface protein involved in iron acquisition from heme in Schizosaccharomyces pombe. J Biol Chem 2015; 290:10176-10190. [PMID: 25733668 PMCID: PMC4400333 DOI: 10.1074/jbc.m115.642058] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/26/2015] [Indexed: 02/03/2023] Open
Abstract
Iron is an essential metal cofactor that is required for many biological processes. Eukaryotic cells have consequently developed different strategies for its acquisition. Until now, Schizosaccharomyces pombe was known to use reductive iron uptake and siderophore-bound iron transport to scavenge iron from the environment. Here, we report the identification of a gene designated shu1(+) that encodes a protein that enables S. pombe to take up extracellular heme for cell growth. When iron levels are low, the transcription of shu1(+) is induced, although its expression is repressed when iron levels rise. The iron-dependent down-regulation of shu1(+) requires the GATA-type transcriptional repressor Fep1, which strongly associates with a proximal promoter region of shu1(+) in vivo in response to iron repletion. HA4-tagged Shu1 localizes to the plasma membrane in cells expressing a functional shu1(+)-HA4 allele. When heme biosynthesis is selectively blocked in mutated S. pombe cells, their ability to acquire exogenous hemin or the fluorescent heme analog zinc mesoporphyrin IX is dependent on the expression of Shu1. Further analysis by absorbance spectroscopy and hemin-agarose pulldown assays showed that Shu1 interacts with hemin, with a KD of ∼2.2 μm. Taken together, results reported here revealed that S. pombe possesses an unexpected pathway for heme assimilation, which may also serve as a source of iron for cell growth.
Collapse
Affiliation(s)
- Thierry Mourer
- From the Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | - Jean-François Jacques
- From the Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | - Ariane Brault
- From the Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | - Martin Bisaillon
- From the Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | - Simon Labbé
- From the Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| |
Collapse
|
40
|
Hu G, Caza M, Cadieux B, Bakkeren E, Do E, Jung WH, Kronstad JW. The endosomal sorting complex required for transport machinery influences haem uptake and capsule elaboration in Cryptococcus neoformans. Mol Microbiol 2015; 96:973-92. [PMID: 25732100 DOI: 10.1111/mmi.12985] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2015] [Indexed: 11/29/2022]
Abstract
Iron availability is a key determinant of virulence in the pathogenic fungus Cryptococcus neoformans. Previous work revealed that the ESCRT (endosomal sorting complex required for transport) protein Vps23 functions in iron acquisition, capsule formation and virulence. Here, we further characterized the ESCRT machinery to demonstrate that defects in the ESCRT-II and III complexes caused reduced capsule attachment, impaired growth on haem and resistance to non-iron metalloprotoporphyrins. The ESCRT mutants shared several phenotypes with a mutant lacking the pH-response regulator Rim101, and in other fungi, the ESCRT machinery is known to activate Rim101 via proteolytic cleavage. We therefore expressed a truncated and activated version of Rim101 in the ESCRT mutants and found that this allele restored capsule formation but not growth on haem, thus suggesting a Rim101-independent contribution to haem uptake. We also demonstrated that the ESCRT machinery acts downstream of the cAMP/protein kinase A pathway to influence capsule elaboration. Defects in the ESCRT components also attenuated virulence in macrophage survival assays and a mouse model of cryptococcosis to a greater extent than reported for loss of Rim101. Overall, these results indicate that the ESCRT complexes function in capsule elaboration, haem uptake and virulence via Rim101-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Guanggan Hu
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Mélissa Caza
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Brigitte Cadieux
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Erik Bakkeren
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Eunsoo Do
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 456-756, Republic of Korea
| | - Won Hee Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 456-756, Republic of Korea
| | - James W Kronstad
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
41
|
Lin C, Lin CN, Wang YC, Liu FY, Chien YW, Chuang YJ, Lan CY, Hsieh WP, Chen BS. Robustness analysis on interspecies interaction network for iron and glucose competition between Candida albicans and zebrafish during infection. BMC SYSTEMS BIOLOGY 2014; 8 Suppl 5:S6. [PMID: 25603810 PMCID: PMC4305985 DOI: 10.1186/1752-0509-8-s5-s6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Candida albicans has emerged as an important model organism for the study of infectious disease. Using high-throughput simultaneously quantified time-course transcriptomics, this study constructed host-pathogen interspecies interaction networks between C. albicans and zebrafish during the adhesion, invasion, and damage stages. Given that iron and glucose have been identified as crucial resources required during the infection process between C. albicans and zebrafish, we focused on the construction of the interspecies networks associated with them. Furthermore, a randomization technique was proposed to identify differentially regulated proteins that are statistically eminent for the three infection stages. The behaviors of the highly connected or differentially regulated proteins identified from the resulting networks were further investigated. "Robustness" is an important system property that measures the ability of the system tolerating the intrinsic perturbations in a dynamic network. This characteristic provides a systematic and quantitative view to elucidate the dynamics of iron and glucose competition in terms of the interspecies interaction networks. Here, we further estimated the robustness of our constructed interspecies interaction networks for the three infection stages. The constructed networks and robustness analysis provided significant insight into dynamic interactions related to iron and glucose competition during infection and enabled us to quantify the system's intrinsic perturbation tolerance ability during iron and glucose competition throughout the three infection stages. Moreover, the networks also assist in elucidating the offensive and defensive mechanisms of C. albicans and zebrafish during their competition for iron and glucose. Our proposed method can be easily extended to identify other such networks involved in the competition for essential resources during infection.
Collapse
|
42
|
Kuznets G, Vigonsky E, Weissman Z, Lalli D, Gildor T, Kauffman SJ, Turano P, Becker J, Lewinson O, Kornitzer D. A relay network of extracellular heme-binding proteins drives C. albicans iron acquisition from hemoglobin. PLoS Pathog 2014; 10:e1004407. [PMID: 25275454 PMCID: PMC4183699 DOI: 10.1371/journal.ppat.1004407] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/18/2014] [Indexed: 12/14/2022] Open
Abstract
Iron scavenging constitutes a crucial challenge for survival of pathogenic microorganisms in the iron-poor host environment. Candida albicans, like many microbial pathogens, is able to utilize iron from hemoglobin, the largest iron pool in the host's body. Rbt5 is an extracellular glycosylphosphatidylinositol (GPI)-anchored heme-binding protein of the CFEM family that facilitates heme-iron uptake by an unknown mechanism. Here, we characterize an additional C. albicans CFEM protein gene, PGA7, deletion of which elicits a more severe heme-iron utilization phenotype than deletion of RBT5. The virulence of the pga7−/− mutant is reduced in a mouse model of systemic infection, consistent with a requirement for heme-iron utilization for C. albicans pathogenicity. The Pga7 and Rbt5 proteins exhibit distinct cell wall attachment, and discrete localization within the cell envelope, with Rbt5 being more exposed than Pga7. Both proteins are shown here to efficiently extract heme from hemoglobin. Surprisingly, while Pga7 has a higher affinity for heme in vitro, we find that heme transfer can occur bi-directionally between Pga7 and Rbt5, supporting a model in which they cooperate in a heme-acquisition relay. Together, our data delineate the roles of Pga7 and Rbt5 in a cell surface protein network that transfers heme from extracellular hemoglobin to the endocytic pathway, and provide a paradigm for how receptors embedded in the cell wall matrix can mediate nutrient uptake across the fungal cell envelope. Candida albicans, a commensal fungus of human mucosal surfaces in healthy individuals, is a common cause of superficial infections, as well as of life-threatening systemic infections in individuals suffering from a reduced immune function. As a systemic pathogen, it has to cope with a scarcity of specific nutrients in the host environment, chief among them iron. To overcome this iron limitation, C. albicans is able to extract iron from heme and hemoglobin, the largest iron pools in the human body, via a pathway that involves endocytosis into the cell. Here we show that efficient heme uptake relies on a family of extracellularly-anchored proteins that serve as heme receptors, two of which, at least, are required for efficient heme utilization. Our data suggest the existence of a relay system that transfers heme from one protein to the next across the cell envelope, explaining the requirement for multiple heme receptors for efficient heme-iron utilization. This study extends our understanding of the pathway of host heme utilization by fungal pathogens, and provides new insights into the question of how nutrients such as heme cross the fungal cell wall.
Collapse
Affiliation(s)
- Galit Kuznets
- B. Rappaport Faculty of Medicine, Technion – I.I.T. and the Rappaport Institute for Research in the Medical Sciences, Haifa, Israel
| | - Elena Vigonsky
- B. Rappaport Faculty of Medicine, Technion – I.I.T. and the Rappaport Institute for Research in the Medical Sciences, Haifa, Israel
| | - Ziva Weissman
- B. Rappaport Faculty of Medicine, Technion – I.I.T. and the Rappaport Institute for Research in the Medical Sciences, Haifa, Israel
| | - Daniela Lalli
- CERM and Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Tsvia Gildor
- B. Rappaport Faculty of Medicine, Technion – I.I.T. and the Rappaport Institute for Research in the Medical Sciences, Haifa, Israel
| | - Sarah J. Kauffman
- Microbiology Department, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Paola Turano
- CERM and Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Jeffrey Becker
- Microbiology Department, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Oded Lewinson
- B. Rappaport Faculty of Medicine, Technion – I.I.T. and the Rappaport Institute for Research in the Medical Sciences, Haifa, Israel
| | - Daniel Kornitzer
- B. Rappaport Faculty of Medicine, Technion – I.I.T. and the Rappaport Institute for Research in the Medical Sciences, Haifa, Israel
- * E-mail:
| |
Collapse
|
43
|
Lippert R, Vojnovic S, Mitrovic A, Jux N, Ivanović-Burmazović I, Vasiljevic B, Stankovic N. Effect of ferrocene-substituted porphyrin RL-91 on Candida albicans biofilm formation. Bioorg Med Chem Lett 2014; 24:3506-11. [DOI: 10.1016/j.bmcl.2014.05.061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/11/2022]
|
44
|
Hemoglobin uptake by Paracoccidioides spp. is receptor-mediated. PLoS Negl Trop Dis 2014; 8:e2856. [PMID: 24831516 PMCID: PMC4022528 DOI: 10.1371/journal.pntd.0002856] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/31/2014] [Indexed: 12/14/2022] Open
Abstract
Iron is essential for the proliferation of fungal pathogens during infection. The availability of iron is limited due to its association with host proteins. Fungal pathogens have evolved different mechanisms to acquire iron from host; however, little is known regarding how Paracoccidioides species incorporate and metabolize this ion. In this work, host iron sources that are used by Paracoccidioides spp. were investigated. Robust fungal growth in the presence of the iron-containing molecules hemin and hemoglobin was observed. Paracoccidioides spp. present hemolytic activity and have the ability to internalize a protoporphyrin ring. Using real-time PCR and nanoUPLC-MSE proteomic approaches, fungal growth in the presence of hemoglobin was shown to result in the positive regulation of transcripts that encode putative hemoglobin receptors, in addition to the induction of proteins that are required for amino acid metabolism and vacuolar protein degradation. In fact, one hemoglobin receptor ortholog, Rbt5, was identified as a surface GPI-anchored protein that recognized hemin, protoporphyrin and hemoglobin in vitro. Antisense RNA technology and Agrobacterium tumefaciens-mediated transformation were used to generate mitotically stable Pbrbt5 mutants. The knockdown strain had a lower survival inside macrophages and in mouse spleen when compared with the parental strain, which suggested that Rbt5 could act as a virulence factor. In summary, our data indicate that Paracoccidioides spp. can use hemoglobin as an iron source most likely through receptor-mediated pathways that might be relevant for pathogenic mechanisms. Fungal infections contribute substantially to human morbidity and mortality. During infectious processes, fungi have evolved mechanisms to obtain iron from high-affinity iron-binding proteins. In the current study, we demonstrated that hemoglobin is the preferential host iron source for the thermodimorphic fungus Paracoccidioides spp. To acquire hemoglobin, the fungus presents hemolytic activity and the ability to internalize protoporphyrin rings. A putative hemoglobin receptor, Rbt5, was demonstrated to be GPI-anchored at the yeast cell surface. Rbt5 was able to bind to hemin, protoporphyrin and hemoglobin in vitro. When rbt5 expression was inhibited, the survival of Paracoccidioides sp. inside macrophages and the fungal burden in mouse spleen diminished, which indicated that Rbt5 could participate in the establishment of the fungus inside the host. Drugs or vaccines could be developed against Paracoccidioides spp. Rbt5 to disturb iron uptake of this micronutrient and, thus, the proliferation of the fungus. Moreover, this protein could be used in routes to introduce antifungal agents into fungal cells.
Collapse
|
45
|
Role of ferric reductases in iron acquisition and virulence in the fungal pathogen Cryptococcus neoformans. Infect Immun 2013; 82:839-50. [PMID: 24478097 DOI: 10.1128/iai.01357-13] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Iron acquisition is critical for the ability of the pathogenic yeast Cryptococcus neoformans to cause disease in vertebrate hosts. In particular, iron overload exacerbates cryptococcal disease in an animal model, defects in iron acquisition attenuate virulence, and iron availability influences the expression of major virulence factors. C. neoformans acquires iron by multiple mechanisms, including a ferroxidase-permease high-affinity system, siderophore uptake, and utilization of both heme and transferrin. In this study, we examined the expression of eight candidate ferric reductase genes and their contributions to iron acquisition as well as to ferric and cupric reductase activities. We found that loss of the FRE4 gene resulted in a defect in production of the virulence factor melanin and increased susceptibility to azole antifungal drugs. In addition, the FRE2 gene was important for growth on the iron sources heme and transferrin, which are relevant for proliferation in the host. Fre2 may participate with the ferroxidase Cfo1 of the high-affinity uptake system for growth on heme, because a mutant lacking both genes showed a more pronounced growth defect than the fre2 single mutant. A role for Fre2 in iron acquisition is consistent with the attenuation of virulence observed for the fre2 mutant. This mutant also was defective in accumulation in the brains of infected mice, a phenotype previously observed for mutants with defects in high-affinity iron uptake (e.g., the cfo1 mutant). Overall, this study provides a more detailed view of the iron acquisition components required for C. neoformans to cause cryptococcosis.
Collapse
|
46
|
Fungal iron availability during deep seated candidiasis is defined by a complex interplay involving systemic and local events. PLoS Pathog 2013; 9:e1003676. [PMID: 24146619 PMCID: PMC3798425 DOI: 10.1371/journal.ppat.1003676] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 08/16/2013] [Indexed: 11/19/2022] Open
Abstract
Nutritional immunity – the withholding of nutrients by the host – has long been recognised as an important factor that shapes bacterial-host interactions. However, the dynamics of nutrient availability within local host niches during fungal infection are poorly defined. We have combined laser ablation-inductively coupled plasma mass spectrometry (LA-ICP MS), MALDI imaging and immunohistochemistry with microtranscriptomics to examine iron homeostasis in the host and pathogen in the murine model of systemic candidiasis. Dramatic changes in the renal iron landscape occur during disease progression. The infection perturbs global iron homeostasis in the host leading to iron accumulation in the renal medulla. Paradoxically, this is accompanied by nutritional immunity in the renal cortex as iron exclusion zones emerge locally around fungal lesions. These exclusion zones correlate with immune infiltrates and haem oxygenase 1-expressing host cells. This local nutritional immunity decreases iron availability, leading to a switch in iron acquisition mechanisms within mature fungal lesions, as revealed by laser capture microdissection and qRT-PCR analyses. Therefore, a complex interplay of systemic and local events influences iron homeostasis and pathogen-host dynamics during disease progression. Microbial pathogens must assimilate essential micronutrients to establish infections. During bacterial infection, mammals limit the availability of micronutrients to inhibit the growth of the pathogen – a phenomenon termed ‘nutrient immunity.’ Nutrient immunity has not been examined during disseminated candidiasis. Yet micronutrient assimilation, and iron assimilation in particular, is required for fungal virulence, and life-threatening disseminated fungal infections are recognised as a major medical threat for patients with compromised immune systems. We show that nutrient immunity operates during disseminated Candida albicans infections in mice. Over time immune cells congregate around the fungal lesions in the kidney cortex, driving nutrient immunity and reducing iron availability for the pathogen. The fungus responds by tuning its iron assimilation strategies to the reduced iron levels. Paradoxically, iron levels increase in other parts of the kidney as Candida infections progress. We show that the fungal infection disturbs global iron homeostasis in the host by perturbing red blood cell recycling in the spleen and this is associated with increased iron storage in the kidney medulla. Therefore, fungal infection exerts system-wide effects upon iron homeostasis in the mammalian host, whilst triggering local nutrient immunity to limit the infection.
Collapse
|
47
|
Noble SM. Candida albicans specializations for iron homeostasis: from commensalism to virulence. Curr Opin Microbiol 2013; 16:708-15. [PMID: 24121029 DOI: 10.1016/j.mib.2013.09.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/06/2013] [Accepted: 09/13/2013] [Indexed: 01/23/2023]
Abstract
Candida albicans is a fungal commensal-pathogen that persistently associates with its mammalian hosts. Between the commensal and pathogenic lifestyles, this microorganism inhabits host niches that differ markedly in the levels of bioavailable iron. A number of recent studies have exposed C. albicans specializations for acquiring iron from specific host molecules in regions where iron is scarce, while also defending against iron-related toxicity in regions where iron occurs in surfeit. Together, these results point to a central role for iron homeostasis in the evolution of this important human pathogen.
Collapse
Affiliation(s)
- Suzanne M Noble
- Department of Microbiology & Immunology, 513 Parnassus Avenue, Box 0414, San Francisco, CA 94143-0414, United States; Division of Infectious Diseases, Department of Medicine, 513 Parnassus Avenue, Box 0414, San Francisco, CA 94143-0414, United States.
| |
Collapse
|
48
|
Álvarez F, Fernández-Ruiz M, Aguado JM. [Iron and invasive fungal infection]. Rev Iberoam Micol 2013; 30:217-25. [PMID: 23684655 DOI: 10.1016/j.riam.2013.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 04/08/2013] [Accepted: 04/30/2013] [Indexed: 01/19/2023] Open
Abstract
Iron is an essential factor for both the growth and virulence of most of microorganisms. As a part of the innate (or nutritional) immune system, mammals have developed different mechanisms to store and transport this element in order to limit free iron bioavailability. To survive in this hostile environment, pathogenic fungi have specific uptake systems for host iron sources, one of the most important of which is based on the synthesis of siderophores-soluble, low-molecular-mass, high-affinity iron chelators. The increase in free iron that results from iron-overload conditions is a well-established risk factor for invasive fungal infection (IFI) such as mucormycosis or aspergillosis. Therefore, iron chelation may be an appealing therapeutic option for these infections. Nevertheless, deferoxamine -the first approved iron chelator- paradoxically increases the incidence of IFI, as it serves as a xeno-siderophore to Mucorales. On the contrary, the new oral iron chelators (deferiprone and deferasirox) have shown to exert a deleterious effect on fungal growth both in vitro and in animal models. The present review focuses on the role of iron metabolism in the pathogenesis of IFI and summarises the preclinical data, as well as the limited clinical experience so far, in the use of new iron chelators as treatment for mucormycosis and invasive aspergillosis.
Collapse
Affiliation(s)
- Florencio Álvarez
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (i+12), Universidad Complutense, Madrid, España
| | | | | |
Collapse
|
49
|
Cadieux B, Lian T, Hu G, Wang J, Biondo C, Teti G, Liu V, Murphy MEP, Creagh AL, Kronstad JW. The Mannoprotein Cig1 supports iron acquisition from heme and virulence in the pathogenic fungus Cryptococcus neoformans. J Infect Dis 2013; 207:1339-47. [PMID: 23322859 DOI: 10.1093/infdis/jit029] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Iron acquisition is critical for virulence of the human pathogenic fungus Cryptococcus neoformans. The cryptococcal transcript for the extracellular mannoprotein Cig1 is highly regulated by iron and abundant in iron-starved cells, suggesting a role in iron acquisition. Indeed, loss of Cig1 resulted in delayed growth on heme at physiological pH. Expression of CIG1 is regulated by the pH-responsive transcription factor Rim101, and loss of Rim101 also impaired growth on heme. A cig1Δ mutant was less susceptible than the wild-type strain to noniron metalloporphyrins, further indicating a role for Cig1 in heme uptake. Recombinant Cig1 exhibited the absorbance spectrum of a heme-binding protein upon heme titration, and Cig1 may therefore function as a hemophore at the cell surface. Cig1 contributed to virulence in a mouse model of cryptococcosis but only in a mutant that also lacked the high-affinity iron uptake system. Overall, Cig1-mediated heme uptake is a potential therapeutic target in C. neoformans.
Collapse
Affiliation(s)
- Brigitte Cadieux
- Michael Smith Laboratories and Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cryptococcus neoformans requires the ESCRT protein Vps23 for iron acquisition from heme, for capsule formation, and for virulence. Infect Immun 2012; 81:292-302. [PMID: 23132495 DOI: 10.1128/iai.01037-12] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Iron availability is a key regulator of virulence factor elaboration in Cryptococcus neoformans, the causative agent of fungal meningoencephalitis in HIV/AIDS patients. In addition, iron is an essential nutrient for pathogen proliferation in mammalian hosts but little is known about the mechanisms of iron sensing and uptake in fungal pathogens that attack humans. In this study, we mutagenized C. neoformans by Agrobacterium-mediated T-DNA insertion and screened for mutants with reduced growth on heme as the sole iron source. Among 34 mutants, we identified a subset with insertions in the gene for the ESCRT-I (endosomal sorting complex required for transport) protein Vps23 that resulted in a growth defect on heme, presumably due to a defect in uptake via endocytosis or misregulation of iron acquisition from heme. Remarkably, vps23 mutants were also defective in the elaboration of the cell-associated capsular polysaccharide that is a major virulence factor, while overexpression of Vps23 resulted in cells with a slightly enlarged capsule. These phenotypes were mirrored by a virulence defect in the vps23 mutant in a mouse model of cryptococcosis and by hypervirulence of the overexpression strain. Overall, these results reveal an important role for trafficking via ESCRT functions in both heme uptake and capsule formation, and they further reinforce the connection between iron and virulence factor deployment in C. neoformans.
Collapse
|