1
|
Zheng D, Zhang X, Ding J, Yue D, Yang F, Li Y. Mechanisms and regulation of iron uptake and the role of iron in pathogenesis of Candida albicans. Crit Rev Microbiol 2025:1-18. [PMID: 40411301 DOI: 10.1080/1040841x.2025.2510256] [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: 11/17/2024] [Revised: 03/25/2025] [Accepted: 05/19/2025] [Indexed: 05/26/2025]
Abstract
Candida albicans is a primary pathogen implicated in invasive fungal infections. Through its intricate iron uptake and regulatory systems, C. albicans adeptly adapts to various iron-rich environments, circumventing the growth and virulence restrictions imposed by the host's nutritional immunity and intensifying infection severity. This fungus activates the Sef1-Sfu1-Hap43 iron homeostasis regulatory circuit via iron bioavailability sensors (iron-sulfur cluster assembly system). This activation precisely regulates multiple iron uptake pathways, including the high-affinity iron reduction system, heme-iron uptake pathway, and siderophore uptake system, as well as genes involved in iron utilization and storage, thus ensuring effective iron acquisition and maintaining iron homeostasis across diverse environmental conditions and developmental stages. Conversely, disruptions in iron metabolism markedly diminish C. albicans's pathogenic potential by impairing mitochondrial function, suppressing hyphal formation, limiting fungal colonization, and reversing antifungal drug resistance. This review presents a comprehensive analysis of the mechanisms governing iron uptake and regulation in C. albicans and examines the consequences of impaired iron homeostasis on mitochondrial function, hyphal formation, infection progression, and drug resistance. Our goal is to provide a theoretical framework to better understand the pathogenesis of C. albicans and to support the development of targeted therapeutic strategies against this resilient pathogen.
Collapse
Affiliation(s)
- Dongming Zheng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Nuclear Medicine, Ya'an People's Hospital, Ya'an, China
| | - Xiaoli Zhang
- Department of Cardiovascular Medicine, Ya'an Second People's Hospital, Ya'an, China
| | - Junping Ding
- Department of Cardiovascular Medicine, Ya'an Second People's Hospital, Ya'an, China
| | - Daifan Yue
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fuzhou Yang
- Department of Nuclear Medicine, Ya'an People's Hospital, Ya'an, China
| | - Yan Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
2
|
Choi G, Bessman NJ. Iron at the crossroads of host-microbiome interactions in health and disease. Nat Microbiol 2025:10.1038/s41564-025-02001-y. [PMID: 40399686 DOI: 10.1038/s41564-025-02001-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/31/2025] [Indexed: 05/23/2025]
Abstract
Iron is an essential dietary micronutrient for both humans and microorganisms. Disruption of iron homeostasis is closely linked, as both a cause and an effect, to the development and progression of gut microbiota dysbiosis and multiple diseases. Iron absorption in humans is impacted by diverse environmental factors, including diet, medication and microbiota-derived molecules. Accordingly, treatment outcomes for iron-associated diseases may depend on an individual patient's microbiome. Here we describe various iron acquisition strategies used by the host, commensal microorganisms and pathogens to benefit or outcompete each other in the complex gut environment. We further explore recently discovered microbial species and metabolites modulating host iron absorption, which represent potential effectors of disease and therapeutic targets. Finally, we discuss the need for mechanistic studies on iron-host-microbiome interactions that can affect disease and treatment outcomes, with the ultimate aim of supporting the development of microbiome-based personalized medicine.
Collapse
Affiliation(s)
- Garam Choi
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Nicholas J Bessman
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, USA.
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
3
|
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
|
4
|
Perfect JR, Kronstad JW. Cryptococcal nutrient acquisition and pathogenesis: dining on the host. Microbiol Mol Biol Rev 2025; 89:e0001523. [PMID: 39927764 PMCID: PMC11948494 DOI: 10.1128/mmbr.00015-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025] Open
Abstract
SUMMARYPathogens must acquire essential nutrients to successfully colonize and proliferate in host tissue. Additionally, nutrients provide signals that condition pathogen deployment of factors that promote disease. A series of transcriptomics experiments over the last 20 years, primarily with Cryptococcus neoformans and to a lesser extent with Cryptococcus gattii, provide insights into the nutritional requirements for proliferation in host tissues. Notably, the identified functions include a number of transporters for key nutrients including sugars, amino acids, metals, and phosphate. Here, we first summarize the in vivo gene expression studies and then discuss the follow-up analyses that specifically test the relevance of the identified transporters for the ability of the pathogens to cause disease. The conclusion is that predictions based on transcriptional profiling of cryptococcal cells in infected tissue are well supported by subsequent investigations using targeted mutations. Overall, the combination of transcriptomic and genetic approaches provides substantial insights into the nutritional requirements that underpin proliferation in the host.
Collapse
Affiliation(s)
- John R. Perfect
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
5
|
Yang X, Li J, Yang Y, Zhang L, Dan X, Cai D, Zhou Z, Li H, Wang X, Zhong S. Early prediction of invasive fungal infection risk in acute-on-chronic liver failure: a prediction model based on admission indicators. BMC Microbiol 2025; 25:131. [PMID: 40069589 PMCID: PMC11900632 DOI: 10.1186/s12866-025-03819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Acute-on-chronic liver failure (ACLF) is a severe clinical syndrome, and the incidence of invasive fungal infection (IFI) among hospitalized patients with ACLF is steadily increasing. The aim of this study is to develop a diagnostic nomogram to assist in the identification of IFI in these patients. METHODS A retrospective study included 705 patients from January 1, 2019, to October 31, 2023, randomly divided into training (n = 493) and validation (n = 212) cohorts. The diagnosis of IFI includes proven diagnosis and probable diagnosis. Kaplan analysis was performed to analyze the survival prognosis of ACLF patients with and without IFI. A nomogram was developed based on a logistic regression model derived through least absolute shrinkage and selection operator (LASSO) regression. The discrimination, accuracy, and clinical utility of the model were assessed using receiver operating characteristic curves, Hosmer-Lemeshow tests, calibration plots, and decision curve analysis. RESULTS Kaplan-Meier survival analysis confirmed that the median survival time of ACLF patients with IFI was significantly lower (by 68 days) than that of ACLF patients without IFI, and there were significant differences in the 90-day, 180-day, and 360-day survival rates between the two groups (P < 0.05). Based on LASSO regression, the following factors were identified as significant risk factors for predicting IFI: aminotransferase levels, prothrombin activity, hemoglobin, neutrophil-to-lymphocyte ratio, and serum total bilirubin. A nomogram was constructed incorporating these variables. The nomogram demonstrated good discriminative ability, with an area under the receiver operating characteristic curve (AUC) of 0.78 (95% confidence interval [CI]: 0.72-0.84) in the training cohort and 0.79 (95% CI: 0.70-0.87) in the validation cohort. Decision curve analysis further validated the clinical applicability of the nomogram. CONCLUSION ACLF patients with IFI have lower survival time than those without IFI. A nomogram was developed and validated to assist clinicians in the early prediction of IFI in hospitalized patients with ACLF. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xu Yang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Jie Li
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Yanli Yang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Li Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Xuelian Dan
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Dachuan Cai
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Zhi Zhou
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Hu Li
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China
| | - Xiaohao Wang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China.
| | - Shan Zhong
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, No.288, Tianwen Avenue, Chayuan, Nan'an District, Chongqing, 401336, China.
| |
Collapse
|
6
|
Ma Y, Zhou Y, Jia T, Zhuang Z, Xue P, Yang L. Deciphering the role of mitochondria in human fungal drug resistance. Mycology 2025:1-14. [DOI: 10.1080/21501203.2025.2473507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 05/04/2025] Open
Affiliation(s)
| | | | | | | | | | - Liang Yang
- Southern University of Science and Technology
| |
Collapse
|
7
|
Patel NK, David MS, Yang S, Garg R, Zhao H, Cormack BP, Culotta VC. Converging Roles of the Metal Transporter SMF11 and the Ferric Reductase FRE1 in Iron Homeostasis of Candida albicans. Mol Microbiol 2024; 122:879-895. [PMID: 39529282 DOI: 10.1111/mmi.15326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Pathogenic fungi must appropriately sense the host availability of essential metals such as Fe. In Candida albicans and other yeasts, sensing of Fe involves mitochondrial Fe-S clusters. Yeast mutants for Fe-S cluster assembly sense Fe limitation even when Fe is abundant and hyperaccumulate Fe. We observe this same disrupted Fe sensing with C. albicans mutants of SMF11, a NRAMP transporter of divalent metals. Mutants of smf11 hyperaccumulate both Mn and Fe and the elevated Mn is secondary to Fe overload. As with Fe-S biogenesis mutants, smf11∆/∆ mutants show upregulation of ferric reductases that are normally repressed under high Fe, and Fe import is activated. However, unlike Fe-S biogenesis mutants, smf11∆/∆ mutants show no defects in mitochondrial Fe-S enzymes. Intriguingly, this exact condition of disrupted Fe sensing without inhibiting Fe-S clusters occurs with C. albicans fre1∆/∆ mutants encoding a ferric reductase. Mutants of fre1 and smf11 display similar perturbations in the cell wall, in filamentation and in the ROS burst of morphogenesis, a Fe-dependent process. As with FRE1, SMF11 is important for virulence in a mouse model for disseminated candidiasis. We propose a model in which FRE1 and SMF11 operate outside the mitochondrial Fe-S pathway to donate ferrous Fe for Fe sensing.
Collapse
Affiliation(s)
- Naisargi K Patel
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Marika S David
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Shuyi Yang
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ritu Garg
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Hongyu Zhao
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Brendan P Cormack
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Valeria C Culotta
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Peppe S, Farrokhi M, Waite EA, Muhi M, Matthaiou EI. Nanoparticle-Mediated Delivery of Deferasirox: A Promising Strategy Against Invasive Aspergillosis. Bioengineering (Basel) 2024; 11:1115. [PMID: 39593775 PMCID: PMC11591955 DOI: 10.3390/bioengineering11111115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Invasive aspergillosis (IA) is a deadly fungal lung infection. Antifungal resistance and treatment side effects are major concerns. Iron chelators are vital for IA management, but systemic use can cause side effects. We developed nanoparticles (NPs) to selectively deliver the iron chelator deferasirox (DFX) for IA treatment. METHODS DFX was encapsulated in poly(lactic-co-glycolic acid) (PLGA) NPs using a single emulsion solvent evaporation method. The NPs were characterized by light scattering and electron microscopy. DFX loading efficiency and release were assessed spectrophotometrically. Toxicity was evaluated using SRB, luciferase, and XTT assays. Therapeutic efficacy was tested in an IA mouse model, assessing fungal burden by qPCR and biodistribution via imaging. RESULTS DFX-NPs had a size of ~50 nm and a charge of ~-30 mV, with a loading efficiency of ~80%. Release kinetics showed DFX release via diffusion and bioerosion. The EC50 of DFX-NPs was significantly lower (p < 0.001) than the free drug, and they were significantly less toxic (p < 0.0001) in mammalian cell cultures. In vivo, NP treatment significantly reduced Af burden (p < 0.05). CONCLUSION The designed DFX-NPs effectively target and kill Af with minimal toxicity to mammalian cells. The significant in vivo therapeutic efficacy suggests these NPs could be a safe and effective treatment for IA.
Collapse
Affiliation(s)
- Sydney Peppe
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.P.); (M.F.); (M.M.)
- Albany Medical College, Washington and Lee University, Lexington, VA 24450, USA
| | - Moloud Farrokhi
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.P.); (M.F.); (M.M.)
| | - Evan A. Waite
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.P.); (M.F.); (M.M.)
- Albany Medical College, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Mustafa Muhi
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.P.); (M.F.); (M.M.)
| | - Efthymia Iliana Matthaiou
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.P.); (M.F.); (M.M.)
| |
Collapse
|
9
|
Houšt’ J, Palyzová A, Pluháček T, Novák J, Marešová H, Hubáček P, Dobiáš R, Stevens DA, Guegan H, Gangneux JP, Havlíček V. Exploring the Siderophore Portfolio for Mass Spectrometry-Based Diagnosis of Scedosporiosis and Lomentosporiosis. ACS OMEGA 2024; 9:44815-44824. [PMID: 39524635 PMCID: PMC11541790 DOI: 10.1021/acsomega.4c08257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Scedosporium apiospermum and Lomentospora prolificans secrete siderophores (iron scavengers) during hyphal proliferation. Siderophores are virulence factors and potential clinical biomarkers of invasive scedosporiosis and lomentosporiosis. Both strains secreted a uniform spectrum of siderophores, including coprogen B (CopB), N α-methyl-coprogen B, dimethyl-coprogen, and ferricrocin, with N α-methyl-coprogen B being the fastest secreted and most abundant coprogen. Under iron and zinc restriction, reflecting a nutrient-limited host environment, L. prolificans secreted 45 times more CopB than did S. apiospermum, presumably contributing to its higher virulence. This robust mobilization of CopB was further enhanced by zinc surplus. Additionally, two novel cyclic peptides, Scedocyclin A and B, were characterized inScedosporium boydii using the de novo sequencing tool CycloBranch. Utilizing matrix-assisted laser desorption/ionization, the portfolio of coprogens detected had limits of detection and quantitation of 4.9 and 14.6 fmol/spot in complex matrices, respectively, making them strong candidates for the next-generation, routine diagnosis of invasive scedosporiosis and lomentosporiosis through the Biotyper siderotyping.
Collapse
Affiliation(s)
- Jiří Houšt’
- Laboratory
of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czechia
- Department
of Analytical Chemistry, Faculty of Science, Palacký University in Olomouc, 17. listopadu 1192/12, 779 00 Olomouc, Czechia
| | - Andrea Palyzová
- Laboratory
of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czechia
| | - Tomáš Pluháček
- Laboratory
of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czechia
- Department
of Analytical Chemistry, Faculty of Science, Palacký University in Olomouc, 17. listopadu 1192/12, 779 00 Olomouc, Czechia
| | - Jiří Novák
- Laboratory
of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czechia
- Department
of Software Engineering, Faculty of Information Technology, Czech Technical University in Prague, Thákurova 9, 160 00 Prague, Czechia
| | - Helena Marešová
- Laboratory
of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czechia
| | - Petr Hubáček
- Department
of the Medical Microbiology, second Faculty of Medicine, Charles University and Motol University Hospital, V Úvalu 84, 150 06 Prague, Czechia
| | - Radim Dobiáš
- Department
of Bacteriology and Mycology, National Reference Laboratory for Mycological
Diagnostics, Public Health Institute in
Ostrava, Partyzánské
náměstí 2633/7, 702 00 Ostrava, Czechia
- Institute
of Laboratory Medicine, Faculty of Medicine, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czechia
| | - David A. Stevens
- Division
of Infectious Diseases and Geographic Medicine, Stanford University
School of Medicine, Foundation for Research
in Infectious Diseases, P.O. Box 2734, Saratoga, California 95070, United States
| | - Hélène Guegan
- Division
of Parasitology and Mycology, European Excellence Center in Medical
Mycology (ECMM EC), National Reference Center on Chronic Aspergillosis, Rennes University Hospital, Inserm UMR_S 1085 Irset, 2 Rue Henri le Guilloux, 35033 Rennes, France
| | - Jean-Pierre Gangneux
- Division
of Parasitology and Mycology, European Excellence Center in Medical
Mycology (ECMM EC), National Reference Center on Chronic Aspergillosis, Rennes University Hospital, Inserm UMR_S 1085 Irset, 2 Rue Henri le Guilloux, 35033 Rennes, France
| | - Vladimír Havlíček
- Laboratory
of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00 Prague, Czechia
| |
Collapse
|
10
|
Garg R, David MS, Yang S, Culotta VC. Metals at the Host-Fungal Pathogen Battleground. Annu Rev Microbiol 2024; 78:23-38. [PMID: 38781605 PMCID: PMC12044431 DOI: 10.1146/annurev-micro-041222-023745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Fungal infections continue to represent a major threat to public health, particularly with the emergence of multidrug-resistant fungal pathogens. As part of the innate immune response, the host modulates the availability of metals as armament against pathogenic microbes, including fungi. The transition metals Fe, Cu, Zn, and Mn are essential micronutrients for all life forms, but when present in excess, these same metals are potent toxins. The host exploits the double-edged sword of these metals, and will either withhold metal micronutrients from pathogenic fungi or attack them with toxic doses. In response to these attacks, fungal pathogens cleverly adapt by modulating metal transport, metal storage, and usage of metals as cofactors for enzymes. Here we review the current state of understanding on Fe, Cu, Zn, and Mn at the host-fungal pathogen battleground and provide perspectives for future research, including a hope for new antifungals based on metals.
Collapse
Affiliation(s)
- Ritu Garg
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - Marika S David
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - Shuyi Yang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - Valeria C Culotta
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| |
Collapse
|
11
|
Rai MN, Lan Q, Parsania C, Rai R, Shirgaonkar N, Chen R, Shen L, Tan K, Wong KH. Temporal transcriptional response of Candida glabrata during macrophage infection reveals a multifaceted transcriptional regulator CgXbp1 important for macrophage response and fluconazole resistance. eLife 2024; 13:e73832. [PMID: 39356739 PMCID: PMC11554308 DOI: 10.7554/elife.73832] [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: 09/13/2021] [Accepted: 10/01/2024] [Indexed: 10/04/2024] Open
Abstract
Candida glabrata can thrive inside macrophages and tolerate high levels of azole antifungals. These innate abilities render infections by this human pathogen a clinical challenge. How C. glabrata reacts inside macrophages and what is the molecular basis of its drug tolerance are not well understood. Here, we mapped genome-wide RNA polymerase II (RNAPII) occupancy in C. glabrata to delineate its transcriptional responses during macrophage infection in high temporal resolution. RNAPII profiles revealed dynamic C. glabrata responses to macrophages with genes of specialized pathways activated chronologically at different times of infection. We identified an uncharacterized transcription factor (CgXbp1) important for the chronological macrophage response, survival in macrophages, and virulence. Genome-wide mapping of CgXbp1 direct targets further revealed its multi-faceted functions, regulating not only virulence-related genes but also genes associated with drug resistance. Finally, we showed that CgXbp1 indeed also affects fluconazole resistance. Overall, this work presents a powerful approach for examining host-pathogen interaction and uncovers a novel transcription factor important for C. glabrata's survival in macrophages and drug tolerance.
Collapse
Affiliation(s)
| | - Qing Lan
- Faculty of Health Sciences, University of MacauTaipaChina
| | | | - Rikky Rai
- Faculty of Health Sciences, University of MacauTaipaChina
| | | | - Ruiwen Chen
- Faculty of Health Sciences, University of MacauTaipaChina
| | - Li Shen
- Faculty of Health Sciences, University of MacauTaipaChina
- Gene Expression, Genomics and Bioinformatics Core, Faculty of Health Sciences, University of MacauTaipaChina
| | - Kaeling Tan
- Faculty of Health Sciences, University of MacauTaipaChina
- Gene Expression, Genomics and Bioinformatics Core, Faculty of Health Sciences, University of MacauTaipaChina
| | - Koon Ho Wong
- Faculty of Health Sciences, University of MacauTaipaChina
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau,Avenida da UniversidadeTaipaChina
- MoE Frontiers Science Center for Precision Oncology, University of MacauTaipaChina
| |
Collapse
|
12
|
Bellavita R, Braccia S, Imbò LE, Grieco P, Galdiero S, D'Auria G, Falanga A, Falcigno L. Exploring Fe(III) coordination and membrane interaction of a siderophore-peptide conjugate: Enhancing synergistically the antimicrobial activity. J Inorg Biochem 2024; 259:112658. [PMID: 38964199 DOI: 10.1016/j.jinorgbio.2024.112658] [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: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Many microbes produce siderophores, which are extremely potent weapons capable of stealing iron ions from human tissues, fluids and cells and transferring them into bacteria through their appropriate porins. We have recently designed a multi-block molecule, each block having a dedicated role. The first component is an antimicrobial peptide, whose good effectiveness against some bacterial strains was gradually improved through interactive sequence modifications. Connected to this block is a flexible bio-band, also optimized in length, which terminates in a hydroxyamide unit, a strong metal binder. Thus, the whole molecule brings together two pieces that work synergistically to fight infection. To understand if the peptide unit, although modified with a long tail, preserves the structure and therefore the antimicrobial activity, and to characterize the mechanism of interaction with bio-membrane models mimicking Gram-negative membranes, we performed a set of fluorescence-based experiments and circular dichroism studies, which further supported our design of a combination of two different entities working synergistically. The chelating activity and iron(III) binding of the peptide was confirmed by iron(III) paramagnetic NMR analyses, and through a competitive assay with ethylenediamine-tetra acetic acid by ultraviolet-visible spectroscopy. The complexation parameters, the Michaelis constant K, and the number of sites n, evaluated with spectrophotometric techniques are confirmed by Fe(III) paramagnetic NMR analyses here reported. In conclusion, we showed that the coupling of antimicrobial capabilities with iron-trapping capabilities works well in the treatment of infectious diseases caused by Gram-negative pathogens.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Simone Braccia
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Lorenzo Emiliano Imbò
- Department of Agricultural Science, University of Naples 'Federico II', Via Università 100, Portici, 80055 Portici, Italy
| | - Paolo Grieco
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Gabriella D'Auria
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples 'Federico II', Via Università 100, Portici, 80055 Portici, Italy.
| | - Lucia Falcigno
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| |
Collapse
|
13
|
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
|
14
|
Weerasinghe H, Stölting H, Rose AJ, Traven A. Metabolic homeostasis in fungal infections from the perspective of pathogens, immune cells, and whole-body systems. Microbiol Mol Biol Rev 2024; 88:e0017122. [PMID: 39230301 PMCID: PMC11426019 DOI: 10.1128/mmbr.00171-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
SUMMARYThe ability to overcome metabolic stress is a major determinant of outcomes during infections. Pathogens face nutrient and oxygen deprivation in host niches and during their encounter with immune cells. Immune cells require metabolic adaptations for producing antimicrobial compounds and mounting antifungal inflammation. Infection also triggers systemic changes in organ metabolism and energy expenditure that range from an enhanced metabolism to produce energy for a robust immune response to reduced metabolism as infection progresses, which coincides with immune and organ dysfunction. Competition for energy and nutrients between hosts and pathogens means that successful survival and recovery from an infection require a balance between elimination of the pathogen by the immune systems (resistance), and doing so with minimal damage to host tissues and organs (tolerance). Here, we discuss our current knowledge of pathogen, immune cell and systemic metabolism in fungal infections, and the impact of metabolic disorders, such as obesity and diabetes. We put forward the idea that, while our knowledge of the use of metabolic regulation for fungal proliferation and antifungal immune responses (i.e., resistance) has been growing over the years, we also need to study the metabolic mechanisms that control tolerance of fungal pathogens. A comprehensive understanding of how to balance resistance and tolerance by metabolic interventions may provide insights into therapeutic strategies that could be used adjunctly with antifungal drugs to improve patient outcomes.
Collapse
Affiliation(s)
- Harshini Weerasinghe
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| | - Helen Stölting
- Department of Biochemistry and Molecular Biology and the Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Adam J Rose
- Department of Biochemistry and Molecular Biology and the Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
15
|
Jeong GJ, Khan F, Tabassum N, Jo DM, Jung WK, Kim YM. Roles of Pseudomonas aeruginosa siderophores in interaction with prokaryotic and eukaryotic organisms. Res Microbiol 2024; 175:104211. [PMID: 38734157 DOI: 10.1016/j.resmic.2024.104211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that produces two types of siderophores, pyoverdine and pyochelin, that play pivotal roles in iron scavenging from the environment and host cells. P. aeruginosa siderophores can serve as virulence factors and perform various functions. Several bacterial and fungal species are likely to interact with P. aeruginosa due to its ubiquity in soil and water as well as its potential to cause infections in plants, animals, and humans. Siderophores produced by P. aeruginosa play critical roles in iron scavenging for prokaryotic species (bacteria) and eukaryotic hosts (fungi, animals, insects, invertebrates, and plants) as well. This review provides a comprehensive discussion of the role of P. aeruginosa siderophores in interaction with prokaryotes and eukaryotes as well as their underlying mechanisms of action. The evolutionary relationship between P. aeruginosa siderophore recognition receptors, such as FpvA, FpvB, and FptA, and those of other bacterial species has also been investigated.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Institute of Fisheries Science, Pukyong National University. Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Du-Min Jo
- National Marine Biodiversity Institute of Korea, Seochun, Chungcheongnam-do, 33662, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
16
|
Ding JL, Feng MG, Ying SH. Two ferrous iron transporter-like proteins independently participate in asexual development under iron limitation and virulence in Beauveria bassiana. Fungal Genet Biol 2024; 173:103908. [PMID: 38857848 DOI: 10.1016/j.fgb.2024.103908] [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: 05/16/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Reductive assimilation pathway involves ferric reductase and ferrous iron transporter, which is integral for fungal iron acquisition. A family of ferric reductase-like proteins has been functionally characterized in the filamentous entomopathogenic fungus Beauveria bassiana. In this investigation, two ferrous iron transporter-like proteins (Ftr) were functionally annotated in B. bassiana. BbFtr1 and BbFtr2 displayed high similarity in structure and were associated with the plasma and nuclear membrane. Their losses had no negatively influence on fungal growth on various nutrients and development under the iron-replete condition. Single mutants of BbFTR1 and BbFTR2 displayed the iron-availability dependent developmental defects, and double mutant exhibited the significantly impaired developmental potential under the iron-limited conditions. In insect bioassay, the double mutant also showed the weaker virulence than either of two single disruption mutants. These results suggested that two ferrous iron transporter-like proteins function independently in fungal physiologies under the iron-deficient condition. Intriguingly, a bZIP transcription factor BbHapX was required for expression of BbFTR1 and BbFTR2 under iron-depleted conditions. This study enhances our understanding of the iron uptake system in the filamentous entomopathogenic fungi.
Collapse
Affiliation(s)
- Jin-Li Ding
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ming-Guang Feng
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng-Hua Ying
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
17
|
Ding JL, Lu M, Liu XL, Feng MG, Ying SH. Essential roles of ferric reductase-like proteins in growth, development, stress response, and virulence of the filamentous entomopathogenic fungus Beauveria bassiana. Microbiol Res 2024; 282:127661. [PMID: 38432016 DOI: 10.1016/j.micres.2024.127661] [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: 12/13/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
In yeasts, ferric reductase catalyzes reduction of ferric ion to ferrous form, which is essential for the reductive iron assimilation system. However, the physiological roles of ferric reductases remain largely unknown in the filamentous fungi. In this study, genome-wide annotation revealed thirteen ferric reductase-like (Fre) proteins in the filamentous insect pathogenic fungus Beauveria bassiana, and all their functions were genetically characterized. Ferric reductase family proteins exhibit different sub-cellular distributions (e.g., cell periphery and vacuole), which was due to divergent domain architectures. Fre proteins had a synergistic effect on fungal virulence, which was ascribed to their distinct functions in different physiologies. Ten Fre proteins were not involved in reduction of ferric ion in submerged mycelia, but most proteins contributed to blastospore development. Only two Fre proteins significantly contributed to B. bassiana vegetative growth under the chemical-induced iron starvation, but most Fre proteins were involved in resistance to osmotic and oxidative stresses. Notably, a bZIP-type transcription factor HapX bound to the promoter regions of all FRE genes in B. bassiana, and displayed varying roles in the transcription activation of these genes. This study reveals the important role of BbFre family proteins in development, stress response, and insect pathogenicity, as well as their distinctive role in the absorption of ferric iron from the environment.
Collapse
Affiliation(s)
- Jin-Li Ding
- Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China; Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Min Lu
- Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Xiao-Long Liu
- Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Ming-Guang Feng
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng-Hua Ying
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
18
|
Henry M, Khemiri I, Tebbji F, Abu-Helu R, Vincent AT, Sellam A. Manganese homeostasis modulates fungal virulence and stress tolerance in Candida albicans. mSphere 2024; 9:e0080423. [PMID: 38380913 PMCID: PMC10964418 DOI: 10.1128/msphere.00804-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024] Open
Abstract
Due to the scarcity of transition metals within the human host, fungal pathogens have evolved sophisticated mechanisms to uptake and utilize these micronutrients at the infection interface. While considerable attention was turned to iron and copper acquisition mechanisms and their importance in fungal fitness, less was done regarding either the role of manganese (Mn) in infectious processes or the cellular mechanism by which fungal cells achieve their Mn-homeostasis. Here, we undertook transcriptional profiling in the pathogenic fungus Candida albicans experiencing both Mn starvation and excess to capture biological processes that are modulated by this metal. We uncovered that Mn scarcity influences diverse processes associated with fungal fitness including invasion of host cells and antifungal sensitivity. We show that Mn levels influence the abundance of iron and zinc emphasizing the complex crosstalk between metals. The deletion of SMF12, a member of Mn Nramp transporters, confirmed its contribution to Mn uptake. smf12 was unable to form hyphae and damage host cells and exhibited sensitivity to azoles. We found that the unfolded protein response (UPR), likely activated by decreased glycosylation under Mn limitation, was required to recover growth when cells were shifted from an Mn-starved to an Mn-repleted medium. RNA-seq profiling of cells exposed to Mn excess revealed that UPR was also activated. Furthermore, the UPR signaling axis Ire1-Hac1 was required to bypass Mn toxicity. Collectively, this study underscores the importance of Mn homeostasis in fungal virulence and comprehensively provides a portrait of biological functions that are modulated by Mn in a fungal pathogen. IMPORTANCE Transition metals such as manganese provide considerable functionality across biological systems as they are used as cofactors for many catalytic enzymes. The availability of manganese is very limited inside the human body. Consequently, pathogenic microbes have evolved sophisticated mechanisms to uptake this micronutrient inside the human host to sustain their growth and cause infections. Here, we undertook a comprehensive approach to understand how manganese availability impacts the biology of the prevalent fungal pathogen, Candida albicans. We uncovered that manganese homeostasis in this pathogen modulates different biological processes that are essential for host infection which underscores the value of targeting fungal manganese homeostasis for potential antifungal therapeutics development.
Collapse
Affiliation(s)
- Manon Henry
- Montreal Heart Institute/Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Inès Khemiri
- Montreal Heart Institute/Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Faiza Tebbji
- Montreal Heart Institute/Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Rasmi Abu-Helu
- Department of Medical Laboratory Sciences, Faculty of Health Professions, Al-Quds University, Jerusalem, Palestine
| | - Antony T. Vincent
- Department of Animal Sciences, Université Laval, Quebec City, Québec, Canada
| | - Adnane Sellam
- Montreal Heart Institute/Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
19
|
Wu PC, Choo YL, Wei SY, Yago JI, Chung KR. Contribution of Autophagy to Cellular Iron Homeostasis and Stress Adaptation in Alternaria alternata. Int J Mol Sci 2024; 25:1123. [PMID: 38256200 PMCID: PMC10816921 DOI: 10.3390/ijms25021123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
The tangerine pathotype of Alternaria alternata produces the Alternaria citri toxin (ACT), which elicits a host immune response characterized by the increase in harmful reactive oxygen species (ROS) production. ROS detoxification in A. alternata relies on the degradation of peroxisomes through autophagy and iron acquisition using siderophores. In this study, we investigated the role of autophagy in regulating siderophore and iron homeostasis in A. alternata. Our results showed that autophagy positively influences siderophore production and iron uptake. The A. alternata strains deficient in autophagy-related genes 1 and 8 (ΔAaatg1 and ΔAaatg8) could not thrive without iron, and their adaptability to high-iron environments was also reduced. Furthermore, the ability of autophagy-deficient strains to withstand ROS was compromised. Notably, autophagy deficiency significantly reduced the production of dimerumic acid (DMA), a siderophore in A. alternata, which may contribute to ROS detoxification. Compared to the wild-type strain, ΔAaatg8 was defective in cellular iron balances. We also observed iron-induced autophagy and lipid peroxidation in A. alternata. To summarize, our study indicates that autophagy and maintaining iron homeostasis are interconnected and contribute to the stress resistance and the virulence of A. alternata. These results provide new insights into the complex interplay connecting autophagy, iron metabolism, and fungal pathogenesis in A. alternata.
Collapse
Affiliation(s)
- Pei-Ching Wu
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| | - Yen-Ling Choo
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| | - Sian-Yong Wei
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| | - Jonar I. Yago
- Plant Science Department, College of Agriculture, Nueva Vizcaya State University, Bayombong 3700, Philippines;
| | - Kuang-Ren Chung
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| |
Collapse
|
20
|
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
|
21
|
Wiesmann CL, Wang NR, Zhang Y, Liu Z, Haney CH. Origins of symbiosis: shared mechanisms underlying microbial pathogenesis, commensalism and mutualism of plants and animals. FEMS Microbiol Rev 2023; 47:fuac048. [PMID: 36521845 PMCID: PMC10719066 DOI: 10.1093/femsre/fuac048] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/17/2023] Open
Abstract
Regardless of the outcome of symbiosis, whether it is pathogenic, mutualistic or commensal, bacteria must first colonize their hosts. Intriguingly, closely related bacteria that colonize diverse hosts with diverse outcomes of symbiosis have conserved host-association and virulence factors. This review describes commonalities in the process of becoming host associated amongst bacteria with diverse lifestyles. Whether a pathogen, commensal or mutualist, bacteria must sense the presence of and migrate towards a host, compete for space and nutrients with other microbes, evade the host immune system, and change their physiology to enable long-term host association. We primarily focus on well-studied taxa, such as Pseudomonas, that associate with diverse model plant and animal hosts, with far-ranging symbiotic outcomes. Given the importance of opportunistic pathogens and chronic infections in both human health and agriculture, understanding the mechanisms that facilitate symbiotic relationships between bacteria and their hosts will help inform the development of disease treatments for both humans, and the plants we eat.
Collapse
Affiliation(s)
- Christina L Wiesmann
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Nicole R Wang
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Yue Zhang
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Zhexian Liu
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cara H Haney
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
22
|
Zheng D, Yue D, Shen J, Li D, Song Z, Huang Y, Yong J, Li Y. Berberine inhibits Candida albicans growth by disrupting mitochondrial function through the reduction of iron absorption. J Appl Microbiol 2023; 134:lxad276. [PMID: 37994672 DOI: 10.1093/jambio/lxad276] [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: 08/28/2023] [Revised: 11/04/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
AIMS This study aimed to investigate whether berberine (BBR) can inhibit the iron reduction mechanism of Candida albicans, lowering the iron uptake of the yeast and perhaps having antimicrobial effects. METHODS AND RESULTS We determined that BBR may cause extensive transcriptional remodeling in C. albicans and that iron permease Ftr1 played a crucial role in this process through eukaryotic transcriptome sequencing. Mechanistic research showed that BBR might selectively inhibit the iron reduction pathway to lower the uptake of exogenous iron ions, inhibiting C. albicans from growing and metabolizing. Subsequent research revealed that BBR caused significant mitochondrial dysfunction, which triggered the process of mitochondrial autophagy. Moreover, we discovered that C. albicans redox homeostasis, susceptibility to antifungal drugs, and hyphal growth are all impacted by the suppression of this mechanism by BBR. CONCLUSIONS The iron reduction mechanism in C. albicans is disrupted by BBR, which disrupts mitochondrial function and inhibits fungal growth. These findings highlight the potential promise of BBR in antifungal applications.
Collapse
Affiliation(s)
- Dongming Zheng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Daifan Yue
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Jinyang Shen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Dongmei Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Zhen Song
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Yifu Huang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Jiangyan Yong
- Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan 610075, China
| | - Yan Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| |
Collapse
|
23
|
Faiyazuddin M, Sophia A, Ashique S, Gholap AD, Gowri S, Mohanto S, Karthikeyan C, Nag S, Hussain A, Akhtar MS, Bakht MA, Ahmed MG, Rustagi S, Rodriguez-Morales AJ, Salas-Matta LA, Mohanty A, Bonilla-Aldana DK, Sah R. Virulence traits and novel drug delivery strategies for mucormycosis post-COVID-19: a comprehensive review. Front Immunol 2023; 14:1264502. [PMID: 37818370 PMCID: PMC10561264 DOI: 10.3389/fimmu.2023.1264502] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
The outbreak of a fatal black fungus infection after the resurgence of the cadaverous COVID-19 has exhorted scientists worldwide to develop a nutshell by repurposing or designing new formulations to address the crisis. Patients expressing COVID-19 are more susceptible to Mucormycosis (MCR) and thus fall easy prey to decease accounting for this global threat. Their mortality rates range around 32-70% depending on the organs affected and grow even higher despite the treatment. The many contemporary recommendations strongly advise using liposomal amphotericin B and surgery as first-line therapy whenever practicable. MCR is a dangerous infection that requires an antifungal drug administration on appropriate prescription, typically one of the following: Amphotericin B, Posaconazole, or Isavuconazole since the fungi that cause MCR are resistant to other medications like fluconazole, voriconazole, and echinocandins. Amphotericin B and Posaconazole are administered through veins (intravenously), and isavuconazole by mouth (orally). From last several years so many compounds are developed against invasive fungal disease but only few of them are able to induce effective treatment against the micorals. Adjuvant medicines, more particularly, are difficult to assess without prospective randomized controlled investigations, which are challenging to conduct given the lower incidence and higher mortality from Mucormycosis. The present analysis provides insight into pathogenesis, epidemiology, clinical manifestations, underlying fungal virulence, and growth mechanisms. In addition, current therapy for MCR in Post Covid-19 individuals includes conventional and novel nano-based advanced management systems for procuring against deadly fungal infection. The study urges involving nanomedicine to prevent fungal growth at the commencement of infection, delay the progression, and mitigate fatality risk.
Collapse
Affiliation(s)
- Md. Faiyazuddin
- School of Pharmacy, Al – Karim University, Katihar, Bihar, India
- Nano Drug Delivery®, Raleigh-Durham, NC, United States
| | - A. Sophia
- PG & Research Department of Physics, Cauvery College for Women (Autonomous), Tiruchirappalli, Tamil Nadu, India
| | - Sumel Ashique
- Department of Pharmaceutics, Pandaveswar School of Pharmacy, Pandaveswar, West Bengal, India
| | - Amol D. Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, India
| | - S. Gowri
- PG & Research Department of Physics, Cauvery College for Women (Autonomous), Tiruchirappalli, Tamil Nadu, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - C. Karthikeyan
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology (SBST), Vellore Institute of Technology (VIT), Tamil Nadu, India
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Mohammad Shabib Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Md. Afroz Bakht
- Chemistry Department, College of Science and Humanity Studies, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Alfonso J. Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas—Institución Universitaria Visión de las Américas, Pereira, Colombia
- Faculties of Health Sciences and Environmental Sciences, Universidad Científica del Sur, Lima, Peru
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Luis Andres Salas-Matta
- Faculties of Health Sciences and Environmental Sciences, Universidad Científica del Sur, Lima, Peru
| | - Aroop Mohanty
- Department of Clinical Microbiology, All India Institute of Medical Sciences, Gorakhpur, India
| | | | - Ranjit Sah
- Institute of Medicine, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
- Department of Clinical Microbiology, DY Patil Medical College, Hospital and Research Centre, DY Patil Vidyapeeth, Pune, Maharashtra, India
- Datta Meghe Institute of Higher Education and Research, Jawaharlal Nehru Medical College, Wardha, India
| |
Collapse
|
24
|
Marquez L, Lee Y, Duncan D, Whitesell L, Cowen LE, Quave C. Potent Antifungal Activity of Penta- O-galloyl-β-d-Glucose against Drug-Resistant Candida albicans, Candida auris, and Other Non- albicans Candida Species. ACS Infect Dis 2023; 9:1685-1694. [PMID: 37607350 PMCID: PMC10496123 DOI: 10.1021/acsinfecdis.3c00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Indexed: 08/24/2023]
Abstract
Among fungal pathogens, infections by drug-resistant Candida species continue to pose a major challenge to healthcare. This study aimed to evaluate the activity of the bioactive natural product, penta-O-galloyl-β-d-glucose (PGG) against multidrug-resistant (MDR) Candida albicans, MDR Candida auris, and other MDR non-albicans Candida species. Here, we show that PGG has a minimum inhibitory concentration (MIC) of 0.25-8 μg mL-1 (0.265-8.5 μM) against three clinical strains of C. auris and a MIC of 0.25-4 μg mL-1 (0.265-4.25 μM) against a panel of other MDR Candida species. Our cytotoxicity studies found that PGG was well tolerated by human kidney, liver, and epithelial cells with an IC50 > 256 μg mL-1 (>272 μM). We also show that PGG is a high-capacity iron chelator and that deletion of key iron homeostasis genes in C. albicans rendered strains hypersensitive to PGG. In conclusion, PGG displayed potent anti-Candida activity with minimal cytotoxicity for human cells. We also found that the antifungal activity of PGG is mediated through an iron-chelating mechanism, suggesting that the compound could prove useful as a topical treatment for superficial Candida infections.
Collapse
Affiliation(s)
- Lewis Marquez
- Molecular
and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, Georgia 30322, United States
- Jones
Center at Ichauway, Newton, Georgia 39870, United States
| | - Yunjin Lee
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Dustin Duncan
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
- Department
of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| | - Luke Whitesell
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Leah E. Cowen
- Department
of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Cassandra Quave
- Center
for the Study of Human Health, Emory University, Atlanta, Georgia 30322, United States
- Department
of Dermatology, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
25
|
Pandey SS. The Role of Iron in Phytopathogenic Microbe-Plant Interactions: Insights into Virulence and Host Immune Response. PLANTS (BASEL, SWITZERLAND) 2023; 12:3173. [PMID: 37687419 PMCID: PMC10563075 DOI: 10.3390/plants12173173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
Iron is an essential element required for the growth and survival of nearly all forms of life. It serves as a catalytic component in multiple enzymatic reactions, such as photosynthesis, respiration, and DNA replication. However, the excessive accumulation of iron can result in cellular toxicity due to the production of reactive oxygen species (ROS) through the Fenton reaction. Therefore, to maintain iron homeostasis, organisms have developed a complex regulatory network at the molecular level. Besides catalyzing cellular redox reactions, iron also regulates virulence-associated functions in several microbial pathogens. Hosts and pathogens have evolved sophisticated strategies to compete against each other over iron resources. Although the role of iron in microbial pathogenesis in animals has been extensively studied, mechanistic insights into phytopathogenic microbe-plant associations remain poorly understood. Recent intensive research has provided intriguing insights into the role of iron in several plant-pathogen interactions. This review aims to describe the recent advances in understanding the role of iron in the lifestyle and virulence of phytopathogenic microbes, focusing on bacteria and host immune responses.
Collapse
Affiliation(s)
- Sheo Shankar Pandey
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati 781035, India; ; Tel.: +91-361-2270095 (ext. 216)
- Citrus Research and Education Center (CREC), Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL 33850, USA
| |
Collapse
|
26
|
Askari F, Vasavi B, Kaur R. Phosphatidylinositol 3-phosphate regulates iron transport via PI3P-binding CgPil1 protein. Cell Rep 2023; 42:112855. [PMID: 37490387 DOI: 10.1016/j.celrep.2023.112855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 05/23/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023] Open
Abstract
Iron homeostasis, which is pivotal to virulence, is regulated by the phosphatidylinositol 3-kinase CgVps34 in the human fungal pathogen Candida glabrata. Here, we identify CgPil1 as a phosphatidylinositol 3-phosphate (PI3P)-binding protein and unveil its role in retaining the high-affinity iron transporter CgFtr1 at the plasma membrane (PM), with PI3P negatively regulating CgFtr1-CgPil1 interaction. PI3P production and its PM localization are elevated in the high-iron environment. Surplus iron also leads to intracellular distribution and vacuolar delivery of CgPil1 and CgFtr1, respectively, from the PM. Loss of CgPil1 or CgFtr1 ubiquitination at lysines 391 and 401 results in CgFtr1 trafficking to the endoplasmic reticulum and a decrease in vacuole-localized CgFtr1. The E3-ubiquitin ligase CgRsp5 interacts with CgFtr1 and forms distinct CgRsp5-CgFtr1 puncta at the PM, with high iron resulting in their internalization. Finally, PI3P controls retrograde transport of many PM proteins. Altogether, we establish PI3P as a key regulator of membrane transport in C. glabrata.
Collapse
Affiliation(s)
- Fizza Askari
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India; Graduate Studies, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Bhogadi Vasavi
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India
| | - Rupinder Kaur
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India.
| |
Collapse
|
27
|
Xue P, Hu G, Jung WH, Kronstad JW. Metals and the cell surface of Cryptococcus neoformans. Curr Opin Microbiol 2023; 74:102331. [PMID: 37257400 PMCID: PMC10513164 DOI: 10.1016/j.mib.2023.102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/18/2023] [Accepted: 05/01/2023] [Indexed: 06/02/2023]
Abstract
Recent studies in pathogenic yeasts reinforce our appreciation of the influence of metal homeostasis on the fungal cell surface. To illustrate this influence, we focus on recent studies on Cryptococcus neoformans, a fungal pathogen with a complex surface of a cell wall with embedded melanin and an attached polysaccharide capsule. Copper and iron are essential yet toxic metals, and current efforts demonstrate the importance of these metals for modulating the surface structure of C. neoformans cells in ways that contribute to fungal-host interactions during disease in vertebrate hosts. In this review, we briefly summarize mechanisms of acquisition and regulation for copper and iron, and then discuss recent insights into the connections between the metals and the cell surface.
Collapse
Affiliation(s)
- Peng Xue
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Guanggan Hu
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Won Hee Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - James W Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.
| |
Collapse
|
28
|
Liu H, Zhang Y, Chen J. Whole-genome sequencing and functional annotation of pathogenic Paraconiothyrium brasiliense causing human cellulitis. Hum Genomics 2023; 17:65. [PMID: 37461066 DOI: 10.1186/s40246-023-00512-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 07/11/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND A pathogenic filamentous fungus causing eyelid cellulitis was isolated from the secretion from a patient's left eyelid, and a phylogenetic analysis based on the rDNA internal transcribed spacer region (ITS) and single-copy gene families identified the isolated strain as Paraconiothyrium brasiliense. The genus Paraconiothyrium contains the major plant pathogenic fungi, and in our study, P. brasiliense was identified for the first time as causing human infection. To comprehensively analyze the pathogenicity, and proteomics of the isolated strain from a genetic perspective, whole-genome sequencing was performed with the Illumina NovaSeq and Oxford Nanopore Technologies platforms, and a bioinformatics analysis was performed with BLAST against genome sequences in various publicly available databases. RESULTS The genome of P. brasiliense GGX 413 is 39.49 Mb in length, with a 51.2% GC content, and encodes 13,057 protein-coding genes and 181 noncoding RNAs. Functional annotation showed that 592 genes encode virulence factors that are involved in human disease, including 61 lethal virulence factors and 30 hypervirulence factors. Fifty-four of these 592 virulence genes are related to carbohydrate-active enzymes, including 46 genes encoding secretory CAZymes, and 119 associated with peptidases, including 70 genes encoding secretory peptidases, and 27 are involved in secondary metabolite synthesis, including four that are associated with terpenoid metabolism. CONCLUSIONS This study establishes the genomic resources of P. brasiliense and provides a theoretical basis for future studies of the pathogenic mechanism of its infection of humans, the treatment of the diseases caused, and related research.
Collapse
Affiliation(s)
- Haibing Liu
- Department of Clinical Laboratory, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yue Zhang
- Department of Clinical Laboratory, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jianguo Chen
- Department of Clinical Laboratory, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
29
|
Nagy L, Vonk P, Künzler M, Földi C, Virágh M, Ohm R, Hennicke F, Bálint B, Csernetics Á, Hegedüs B, Hou Z, Liu X, Nan S, Pareek M, Sahu N, Szathmári B, Varga T, Wu H, Yang X, Merényi Z. Lessons on fruiting body morphogenesis from genomes and transcriptomes of Agaricomycetes. Stud Mycol 2023; 104:1-85. [PMID: 37351542 PMCID: PMC10282164 DOI: 10.3114/sim.2022.104.01] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/02/2022] [Indexed: 01/09/2024] Open
Abstract
Fruiting bodies (sporocarps, sporophores or basidiomata) of mushroom-forming fungi (Agaricomycetes) are among the most complex structures produced by fungi. Unlike vegetative hyphae, fruiting bodies grow determinately and follow a genetically encoded developmental program that orchestrates their growth, tissue differentiation and sexual sporulation. In spite of more than a century of research, our understanding of the molecular details of fruiting body morphogenesis is still limited and a general synthesis on the genetics of this complex process is lacking. In this paper, we aim at a comprehensive identification of conserved genes related to fruiting body morphogenesis and distil novel functional hypotheses for functionally poorly characterised ones. As a result of this analysis, we report 921 conserved developmentally expressed gene families, only a few dozens of which have previously been reported to be involved in fruiting body development. Based on literature data, conserved expression patterns and functional annotations, we provide hypotheses on the potential role of these gene families in fruiting body development, yielding the most complete description of molecular processes in fruiting body morphogenesis to date. We discuss genes related to the initiation of fruiting, differentiation, growth, cell surface and cell wall, defence, transcriptional regulation as well as signal transduction. Based on these data we derive a general model of fruiting body development, which includes an early, proliferative phase that is mostly concerned with laying out the mushroom body plan (via cell division and differentiation), and a second phase of growth via cell expansion as well as meiotic events and sporulation. Altogether, our discussions cover 1 480 genes of Coprinopsis cinerea, and their orthologs in Agaricus bisporus, Cyclocybe aegerita, Armillaria ostoyae, Auriculariopsis ampla, Laccaria bicolor, Lentinula edodes, Lentinus tigrinus, Mycena kentingensis, Phanerochaete chrysosporium, Pleurotus ostreatus, and Schizophyllum commune, providing functional hypotheses for ~10 % of genes in the genomes of these species. Although experimental evidence for the role of these genes will need to be established in the future, our data provide a roadmap for guiding functional analyses of fruiting related genes in the Agaricomycetes. We anticipate that the gene compendium presented here, combined with developments in functional genomics approaches will contribute to uncovering the genetic bases of one of the most spectacular multicellular developmental processes in fungi. Citation: Nagy LG, Vonk PJ, Künzler M, Földi C, Virágh M, Ohm RA, Hennicke F, Bálint B, Csernetics Á, Hegedüs B, Hou Z, Liu XB, Nan S, M. Pareek M, Sahu N, Szathmári B, Varga T, Wu W, Yang X, Merényi Z (2023). Lessons on fruiting body morphogenesis from genomes and transcriptomes of Agaricomycetes. Studies in Mycology 104: 1-85. doi: 10.3114/sim.2022.104.01.
Collapse
Affiliation(s)
- L.G. Nagy
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - P.J. Vonk
- Microbiology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands;
| | - M. Künzler
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland;
| | - C. Földi
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - M. Virágh
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - R.A. Ohm
- Microbiology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands;
| | - F. Hennicke
- Project Group Genetics and Genomics of Fungi, Chair Evolution of Plants and Fungi, Ruhr-University Bochum, 44780, Bochum, North Rhine-Westphalia, Germany;
| | - B. Bálint
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - Á. Csernetics
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - B. Hegedüs
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - Z. Hou
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - X.B. Liu
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - S. Nan
- Institute of Applied Mycology, Huazhong Agricultural University, 430070 Hubei Province, PR China
| | - M. Pareek
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - N. Sahu
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - B. Szathmári
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - T. Varga
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - H. Wu
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| | - X. Yang
- Institute of Applied Mycology, Huazhong Agricultural University, 430070 Hubei Province, PR China
| | - Z. Merényi
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, 6726, Hungary;
| |
Collapse
|
30
|
Meagher RB, Lewis ZA, Ambati S, Lin X. DectiSomes: C-type lectin receptor-targeted liposomes as pan-antifungal drugs. Adv Drug Deliv Rev 2023; 196:114776. [PMID: 36934519 PMCID: PMC10133202 DOI: 10.1016/j.addr.2023.114776] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023]
Abstract
Combatting the ever-increasing threat from invasive fungal pathogens faces numerous fundamental challenges, including constant human exposure to large reservoirs of species in the environment, the increasing population of immunocompromised or immunosuppressed individuals, the unsatisfactory efficacy of current antifungal drugs and their associated toxicity, and the scientific and economic barriers limiting a new antifungal pipeline. DectiSomes represent a new drug delivery platform that enhances antifungal efficacy for diverse fungal pathogens and reduces host toxicity for current and future antifungals. DectiSomes employ pathogen receptor proteins - C-type lectins - to target drug-loaded liposomes to conserved fungal cognate ligands and away from host cells. DectiSomes represent one leap forward for urgently needed effective pan-antifungal therapy. Herein, we discuss the problems of battling fungal diseases and the state of DectiSome development.
Collapse
Affiliation(s)
- Richard B Meagher
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Zachary A Lewis
- Department of Genetics, University of Georgia, Athens, GA 30602, USA; Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Suresh Ambati
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
31
|
Xie S, Wang C, Zeng T, Wang H, Suo H. Whole-genome and comparative genome analysis of Mucor racemosus C isolated from Yongchuan Douchi. Int J Biol Macromol 2023; 234:123397. [PMID: 36739051 DOI: 10.1016/j.ijbiomac.2023.123397] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/10/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023]
Abstract
Mucor racemosus is the predominant fungal in the zhiqu stage of the fermentation of Yongchuan Douchi (Mucor-type), which plays an important role in the fermentation process of Yongchuan Douchi. However, there is a lack of information on the genetic analysis of M. racemosus. In this study, we isolated and identified M. racemosus C (accession no JAPEHQ000000000) from Yongchuan Douchi and analyzed the physiological indicators, then genomic information of the strain to perform a comprehensive analysis of its fermentation capacity and safety. M. racemosus C had neutral protease activity up to 68.051 U/mL at 30 °C and alkaline protease activity up to 57.367 U/mL at 25 °C. In addition, comparing the genomic data with the COGs database (NCBI), it was predicted that M. racemosus C undergoes extensive amino acid metabolism, making C suitable for the production of fermented foods (e.g., Douchi, Syoyu, and sufu). Finally, we performed virulence genes and resistance genes analysis, hemolysis experiment, aflatoxins assay, antibiotic resistance assay to evaluate the safety of M. racemosus C, and the results showed that M. racemosus C was safe, non-toxin-producing and non-hemolytic.
Collapse
Affiliation(s)
- Shicai Xie
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China
| | - Tao Zeng
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China
| | - Hongwei Wang
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China.
| |
Collapse
|
32
|
Wu JJ, Wu PC, Yago JI, Chung KR. The Regulatory Hub of Siderophore Biosynthesis in the Phytopathogenic Fungus Alternaria alternata. J Fungi (Basel) 2023; 9:jof9040427. [PMID: 37108881 PMCID: PMC10146468 DOI: 10.3390/jof9040427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
A GATA zinc finger-containing repressor (AaSreA) suppresses siderophore biosynthesis in the phytopathogenic fungus Alternaria alternata under iron-replete conditions. In this study, targeted gene deletion revealed two bZIP-containing transcription factors (AaHapX and AaAtf1) and three CCAAT-binding proteins (AaHapB, AaHapC, and AaHapE) that positively regulate gene expression in siderophore production. This is a novel phenotype regarding Atf1 and siderophore biosynthesis. Quantitative RT-PCR analyses revealed that only AaHapX and AaSreA were regulated by iron. AaSreA and AaHapX form a transcriptional feedback negative loop to regulate iron acquisition in response to the availability of environmental iron. Under iron-limited conditions, AaAtf1 enhanced the expression of AaNps6, thus playing a positive role in siderophore production. However, under nutrient-rich conditions, AaAtf1 plays a negative role in resistance to sugar-induced osmotic stress, and AaHapX plays a negative role in resistance to salt-induced osmotic stress. Virulence assays performed on detached citrus leaves revealed that AaHapX and AaAtf1 play no role in fungal pathogenicity. However, fungal strains carrying the AaHapB, AaHapC, or AaHapE deletion failed to incite necrotic lesions, likely due to severe growth deficiency. Our results revealed that siderophore biosynthesis and iron homeostasis are regulated by a well-organized network in A. alternata.
Collapse
|
33
|
XUE P, SÁNCHEZ-LEÓN E, DAMOO D, HU G, JUNG WH, KRONSTAD JW. Heme sensing and trafficking in fungi. FUNGAL BIOL REV 2023; 43:100286. [PMID: 37781717 PMCID: PMC10540271 DOI: 10.1016/j.fbr.2022.09.002] [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] [Indexed: 11/04/2022]
Abstract
Fungal pathogens cause life-threatening diseases in humans, and the increasing prevalence of these diseases emphasizes the need for new targets for therapeutic intervention. Nutrient acquisition during infection is a promising target, and recent studies highlight the contributions of endomembrane trafficking, mitochondria, and vacuoles in the sensing and acquisition of heme by fungi. These studies have been facilitated by genetically encoded biosensors and other tools to quantitate heme in subcellular compartments and to investigate the dynamics of trafficking in living cells. In particular, the applications of biosensors in fungi have been extended beyond the detection of metabolites, cofactors, pH, and redox status to include the detection of heme. Here, we focus on studies that make use of biosensors to examine mechanisms of heme uptake and degradation, with guidance from the model fungus Saccharomyces cerevisiae and an emphasis on the pathogenic fungi Candida albicans and Cryptococcus neoformans that threaten human health. These studies emphasize a role for endocytosis in heme uptake, and highlight membrane contact sites involving mitochondria, the endoplasmic reticulum and vacuoles as mediators of intracellular iron and heme trafficking.
Collapse
Affiliation(s)
- Peng XUE
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eddy SÁNCHEZ-LEÓN
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Djihane DAMOO
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guanggan HU
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Won Hee JUNG
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| | - James W. KRONSTAD
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
34
|
Aguiar M, Orasch T, Shadkchan Y, Caballero P, Pfister J, Sastré-Velásquez LE, Gsaller F, Decristoforo C, Osherov N, Haas H. Uptake of the Siderophore Triacetylfusarinine C, but Not Fusarinine C, Is Crucial for Virulence of Aspergillus fumigatus. mBio 2022; 13:e0219222. [PMID: 36125294 PMCID: PMC9600649 DOI: 10.1128/mbio.02192-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/02/2022] [Indexed: 11/20/2022] Open
Abstract
Siderophores play an important role in fungal virulence, serving as trackers for in vivo imaging and as biomarkers of fungal infections. However, siderophore uptake is only partially characterized. As the major cause of aspergillosis, Aspergillus fumigatus is one of the most common airborne fungal pathogens of humans. Here, we demonstrate that this mold species mediates the uptake of iron chelated by the secreted siderophores triacetylfusarinine C (TAFC) and fusarinine C by the major facilitator-type transporters MirB and MirD, respectively. In a murine aspergillosis model, MirB but not MirD was found to be crucial for virulence, indicating that TAFC-mediated uptake plays a dominant role during infection. In the absence of MirB, TAFC becomes inhibitory by decreasing iron availability because the mutant is not able to recognize iron that is chelated by TAFC. MirB-mediated transport was found to tolerate the conjugation of fluorescein isothiocyanate to triacetylfusarinine C, which might aid in the development of siderophore-based antifungals in a Trojan horse approach, particularly as the role of MirB in pathogenicity restrains its mutational inactivation. Taken together, this study identified the first eukaryotic siderophore transporter that is crucial for virulence and elucidated its translational potential as well as its evolutionary conservation. IMPORTANCE Aspergillus fumigatus is responsible for thousands of cases of invasive fungal disease annually. For iron uptake, A. fumigatus secretes so-called siderophores, which are taken up after the binding of environmental iron. Moreover, A. fumigatus can utilize siderophore types that are produced by other fungi or bacteria. Fungal siderophores raised considerable interest due to their role in virulence and their potential for the diagnosis and treatment of fungal infections. Here, we demonstrate that the siderophore transporter MirB is crucial for the virulence of A. fumigatus, which reveals that its substrate, triacetylfusarinine C, is the most important siderophore during infection. We found that in the absence of MirB, TAFC becomes inhibitory by decreasing the availability of environmental iron and that MirB-mediated transport tolerates the derivatization of its substrate, which might aid in the development of siderophore-based antifungals. This study significantly improved the understanding of fungal iron homeostasis and the role of siderophores in interactions with the host.
Collapse
Affiliation(s)
- Mario Aguiar
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Orasch
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Yana Shadkchan
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine Ramat-Aviv, Tel Aviv, Israel
| | - Patricia Caballero
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Joachim Pfister
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | - Fabio Gsaller
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Nir Osherov
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine Ramat-Aviv, Tel Aviv, Israel
| | - Hubertus Haas
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
35
|
Andrawes N, Weissman Z, Pinsky M, Moshe S, Berman J, Kornitzer D. Regulation of heme utilization and homeostasis in Candida albicans. PLoS Genet 2022; 18:e1010390. [PMID: 36084128 PMCID: PMC9491583 DOI: 10.1371/journal.pgen.1010390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/21/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
Heme (iron-protoporphyrin IX) is an essential but potentially toxic cellular cofactor. While most organisms are heme prototrophs, many microorganisms can utilize environmental heme as iron source. The pathogenic yeast Candida albicans can utilize host heme in the iron-poor host environment, using an extracellular cascade of soluble and anchored hemophores, and plasma membrane ferric reductase-like proteins. To gain additional insight into the C. albicans heme uptake pathway, we performed an unbiased genetic selection for mutants resistant to the toxic heme analog Ga3+-protoporphyrin IX at neutral pH, and a secondary screen for inability to utilize heme as iron source. Among the mutants isolated were the genes of the pH-responsive RIM pathway, and a zinc finger transcription factor related to S. cerevisiae HAP1. In the presence of hemin in the medium, C. albicans HAP1 is induced, the Hap1 protein is stabilized and Hap1-GFP localizes to the nucleus. In the hap1 mutant, cytoplasmic heme levels are elevated, while influx of extracellular heme is lower. Gene expression analysis indicated that in the presence of extracellular hemin, Hap1 activates the heme oxygenase HMX1, which breaks down excess cytoplasmic heme, while at the same time it also activates all the known heme uptake genes. These results indicate that Hap1 is a heme-responsive transcription factor that plays a role both in cytoplasmic heme homeostasis and in utilization of extracellular heme. The induction of heme uptake genes by C. albicans Hap1 under iron satiety indicates that preferential utilization of host heme can be a dietary strategy in a heme prototroph.
Collapse
Affiliation(s)
- Natalie Andrawes
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion–I.I.T., Haifa, Israel
| | - 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
| | - Shilat Moshe
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion–I.I.T., Haifa, Israel
| | - Judith Berman
- School of Molecular Microbiology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion–I.I.T., Haifa, Israel
| |
Collapse
|
36
|
Abstract
Heme (protoheme IX) is an essential cofactor for a large variety of proteins whose functions vary from one electron reactions to binding gases. While not ubiquitous, heme is found in the great majority of known life forms. Unlike most cofactors that are acquired from dietary sources, the vast majority of organisms that utilize heme possess a complete pathway to synthesize the compound. Indeed, dietary heme is most frequently utilized as an iron source and not as a source of heme. In Nature there are now known to exist three pathways to synthesize heme. These are the siroheme dependent (SHD) pathway which is the most ancient, but least common of the three; the coproporphyrin dependent (CPD) pathway which with one known exception is found only in gram positive bacteria; and the protoporphyrin dependent (PPD) pathway which is found in gram negative bacteria and all eukaryotes. All three pathways share a core set of enzymes to convert the first committed intermediate, 5-aminolevulinate (ALA) into uroporphyrinogen III. In the current review all three pathways are reviewed as well as the two known pathways to synthesize ALA. In addition, interesting features of some heme biosynthesis enzymes are discussed as are the regulation and disorders of heme biosynthesis.
Collapse
Affiliation(s)
- Harry A Dailey
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602-1111, USA
- Department of Microbiology, University of Georgia, Athens, GA 30602-1111, USA
| | - Amy E Medlock
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602-1111, USA
- Augusta University/University of Georgia Medical Partnership, University of Georgia, Athens, GA, USA
| |
Collapse
|
37
|
Song M, Thak EJ, Kang HA, Kronstad JW, Jung WH. Cryptococcus neoformans can utilize ferritin as an iron source. Med Mycol 2022; 60:myac056. [PMID: 35943215 PMCID: PMC9387142 DOI: 10.1093/mmy/myac056] [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: 05/03/2022] [Revised: 07/11/2022] [Accepted: 08/06/2022] [Indexed: 01/21/2023] Open
Abstract
UNLABELLED Ferritin, a major iron storage protein in vertebrates, supplies iron upon iron deficiency. Ferritin is also found extracellularly, and acts as an iron carrier and a contributor to the immune response to invading microbes. Some microbial pathogens take advantage of ferritin as an iron source upon infection. However, no information is currently available on whether the human fungal pathogen Cryptococcus neoformans can acquire iron from ferritin. Here, we found that C. neoformans grew well in the presence of ferritin as a sole iron source. We showed that the binding of ferritin to the surface of C. neoformans is necessary and that acidification may contribute to ferritin-iron utilization by the fungus. Our data also revealed that the high-affinity reductive iron uptake system in C. neoformans is required for ferritin-iron acquisition. Furthermore, phagocytosis of C. neoformans by macrophages led to increased intracellular ferritin levels, suggesting that iron is sequestered by ferritin in infected macrophages. The increase in intracellular ferritin levels was reversed upon infection with a C. neoformans mutant deficient in the high-affinity reductive iron uptake system, indicating that this system plays a major role in iron acquisition in the phagocytosed C. neoformans in macrophages. LAY SUMMARY Cryptococcus neoformans is an opportunistic fungal pathogen causing life-threatening pulmonary disease and cryptococcal meningitis, mainly in immunocompromised patients. In this study, we found that C. neoformans can use ferritin, a major iron storage protein in vertebrates, as a sole iron source.
Collapse
Affiliation(s)
- Moonyong Song
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| | - Eun Jung Thak
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Hyun Ah Kang
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - James W Kronstad
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Won Hee Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| |
Collapse
|
38
|
Lapiere A, Richard ML. Bacterial-fungal metabolic interactions within the microbiota and their potential relevance in human health and disease: a short review. Gut Microbes 2022; 14:2105610. [PMID: 35903007 PMCID: PMC9341359 DOI: 10.1080/19490976.2022.2105610] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The composition of the microbiota is the focus of many recent publications describing the effects of the microbiota on host health. In recent years, research has progressed further, investigating not only the diversity of genes and functions but also metabolites produced by microorganisms composing the microbiota of various niches and how these metabolites affect and shape the microbial community. While an abundance of data has been published on bacterial interactions, much less data are available on the interactions of bacteria with another component of the microbiota: the fungal community. Although present in smaller numbers, fungi are essential to the balance of this complex microbial ecosystem. Both bacterial and fungal communities produce metabolites that influence their own population but also that of the other. However, to date, interkingdom interactions occurring through metabolites produced by bacteria and fungi have rarely been described. In this review, we describe the major metabolites produced by both kingdoms and discuss how they influence each other, by what mechanisms and with what consequences for the host.
Collapse
Affiliation(s)
- Alexia Lapiere
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France,Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, France
| | - Mathias L Richard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France,Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, France,CONTACT Mathias L Richard INRAE, Micalis Institute, Probihote Team, Domaine de Vilvert, 78352, Jouy en Josas, France
| |
Collapse
|
39
|
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
|
40
|
Duan X, Xie Z, Ma L, Jin X, Zhang M, Xu Y, Liu Y, Lou H, Chang W. Selective Metal Chelation by a Thiosemicarbazone Derivative Interferes with Mitochondrial Respiration and Ribosome Biogenesis in Candida albicans. Microbiol Spectr 2022; 10:e0195121. [PMID: 35412374 PMCID: PMC9241695 DOI: 10.1128/spectrum.01951-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/04/2022] [Indexed: 11/20/2022] Open
Abstract
Metal chelation is generally considered as a promising antifungal approach but its specific mechanisms are unclear. Here, we identify 13 thiosemicarbazone derivatives that exert broad-spectrum antifungal activity with potency comparable or superior to that of fluconazole in vitro by screening a small compound library comprising 89 thiosemicarbazone derivatives as iron chelators. Among the hits, 19ak exhibits minimal cytotoxicity and potent activity against either azole-sensitive or azole-resistant fungal pathogens. Mechanism investigations reveal that 19ak inhibits mitochondrial respiration mainly by retarding mitochondrial respiratory chain complex I activity through iron chelation, and further reduces mitochondrial membrane potential and ATP synthesis in Candida albicans. In addition, 19ak inhibits fungal ribosome biogenesis mainly by disrupting intracellular zinc homeostasis. 19ak also stimulates the activities of antioxidant enzymes and decreases reactive oxygen species formation in C. albicans, resulting in an increase in detrimental intracellular reductive stress. However, 19ak has minor effects on mammalian cells in depleting intracellular iron and zinc. Moreover, 19ak exhibits low capacity to induce drug resistance and in vivo efficacy in a Galleria mellonella infection model. These findings uncover retarded fungal mitochondrial respiration and ribosome biogenesis as downstream effects of disruption of iron and zinc homeostasis in C. albicans and provide a basis for the thiosemicarbazone 19ak in antifungal application. IMPORTANCE The increasing incidence of fungal infections and resistance to existing antifungals call for the development of broad-spectrum antifungals with novel mechanisms of action. In this study, we demonstrate that a thiosemicarbazone derivative 19ak selectively inhibits mitochondrial respiration mainly by retarding mitochondrial respiratory chain complex I activity through iron chelation and inhibits ribosome biogenesis mainly by disrupting intracellular zinc homeostasis in C. albicans. In addition, 19ak exhibits low capacity to induce fungal resistance, minimal cytotoxicity, and in vivo antifungal efficacy. This study provides the basis of thiosemicarbazone derivative 19ak as a metal chelator for the treatment of fungal infections.
Collapse
Affiliation(s)
- Ximeng Duan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Zhiyu Xie
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, Henan, People’s Republic of China
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Pharmaceutical Research and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xueyang Jin
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ming Zhang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuliang Xu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Yue Liu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Wenqiang Chang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
41
|
Ye J, Wang Y, Li X, Wan Q, Zhang Y, Lu L. Synergistic Antifungal Effect of a Combination of Iron Deficiency and Calcium Supplementation. Microbiol Spectr 2022; 10:e0112122. [PMID: 35674440 PMCID: PMC9241635 DOI: 10.1128/spectrum.01121-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/19/2022] [Indexed: 01/10/2023] Open
Abstract
Fungal diseases have become a major public health issue worldwide. Increasing drug resistance and the limited number of available antifungals result in high morbidity and mortality. Metal-based drugs have been reported to be therapeutic agents against major protozoan diseases, but knowledge of their ability to function as antifungals is limited. In this study, we found that calcium supplementation combined with iron deficiency causes dramatic growth inhibition of the human fungal pathogens Aspergillus fumigatus, Candida albicans, and Cryptococcus neoformans. Calcium induces the downregulation of iron uptake-related genes and, in particular, causes a decrease in the expression of the transcription factor HapX, which tends to transcriptionally activate siderophore-mediated iron acquisition under iron-deficient conditions. Iron deficiency causes calcium overload and the overproduction of intracellular reactive oxygen species (ROS), and perturbed ion homeostasis suppresses fungal growth. These phenomena are consistently identified in azole-resistant A. fumigatus isolates. The findings here imply that low iron availability lets cells mistakenly absorb calcium as a substitute, causing calcium abnormalities. Thus, there is a mutual effect between iron and calcium in fungal pathogens, and the combination of calcium with an iron chelator could serve to improve antifungal therapy. IMPORTANCE Millions of immunocompromised people are at a higher risk of developing different types of severe fungal diseases. The limited number of antifungals and the emergence of antimicrobial resistance highlight an urgent need for new strategies against invasive fungal infections. Here, we report that calcium can interfere with iron absorption of fungal pathogens, especially in iron-limited environments. Thus, a combination of calcium supplementation with an iron chelator inhibits the growth of human fungal pathogens, including Aspergillus fumigatus, Candida albicans, and Cryptococcus neoformans. Moreover, we demonstrate that iron deficiency induces a nonspecific calcium uptake response, which results in toxic levels of metal. Findings in this study suggest that a microenvironment with excess calcium and limited iron is an efficient strategy to curb the growth of fungal pathogens, especially for drug-resistant isolates.
Collapse
Affiliation(s)
- Jing Ye
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yamei Wang
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xinyu Li
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Qinyi Wan
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yuanwei Zhang
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ling Lu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
42
|
Lee SR, Schalk F, Schwitalla JW, Guo H, Yu JS, Song M, Jung WH, de Beer ZW, Beemelmanns C, Kim KH. GNPS‐Guided Discovery of Madurastatin Siderophores from the Termite‐Associated
Actinomadura
sp. RB99**. Chemistry 2022; 28:e202200612. [PMID: 35404539 PMCID: PMC9325478 DOI: 10.1002/chem.202200612] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 12/14/2022]
Abstract
In this study, we analyzed if Actinomadura sp. RB99 produces siderophores that that could be responsible for the antimicrobial activity observed in co‐cultivation studies. Dereplication of high‐resolution tandem mass spectrometry (HRMS/MS) and global natural product social molecular networking platform (GNPS) analysis of fungus‐bacterium co‐cultures resulted in the identification of five madurastatin derivatives (A1, A2, E1, F, and G1), of which were four new derivatives. Chemical structures were unambiguously confirmed by HR‐ESI‐MS, 1D and 2D NMR experiments, as well as MS/MS data and their absolute structures were elucidated based on Marfey's analysis, DP4+ probability calculation and total synthesis. Structure analysis revealed that madurastatin E1 (2) contained a rare 4‐imidazolidinone cyclic moiety and madurastatin A1 (5) was characterized as a Ga3+‐complex. The function of madurastatins as siderophores was evaluated using the fungal pathogen Cryptococcus neoformans as model organism. Based on homology models, we identified the putative NRPS‐based gene cluster region of the siderophores in Actinomadura sp. RB99.
Collapse
Affiliation(s)
- Seoung Rak Lee
- School of Pharmacy Sungkyunkwan University Suwon 16419 (Republic of Korea
- Department of Chemistry Princeton University New Jersey 08544 USA
| | - Felix Schalk
- Chemical Biology of Microbe-Host Interactions Hans-Knöll Institute (HKI) Beutenbergstraße 11a 07745 Jena Germany
| | - Jan W. Schwitalla
- Chemical Biology of Microbe-Host Interactions Hans-Knöll Institute (HKI) Beutenbergstraße 11a 07745 Jena Germany
| | - Huijuan Guo
- Chemical Biology of Microbe-Host Interactions Hans-Knöll Institute (HKI) Beutenbergstraße 11a 07745 Jena Germany
| | - Jae Sik Yu
- School of Pharmacy Sungkyunkwan University Suwon 16419 (Republic of Korea
| | - Moonyong Song
- Department of Systems Biotechnology Chung-Ang University Anseong 17546 Republic of Korea
| | - Won Hee Jung
- Department of Systems Biotechnology Chung-Ang University Anseong 17546 Republic of Korea
| | - Z. Wilhelm de Beer
- Department of Biochemistry Genetics and Microbiology Forestry and Agricultural Biotechnology Institute (FABI) University of Pretoria Hatfield 0028 Pretoria South Africa
| | - Christine Beemelmanns
- Chemical Biology of Microbe-Host Interactions Hans-Knöll Institute (HKI) Beutenbergstraße 11a 07745 Jena Germany
- Biochemistry of Microbial Metabolism Institute of Biochemistry Leipzig University Johannisallee 21–23 Leipzig 04103 Germany
| | - Ki Hyun Kim
- School of Pharmacy Sungkyunkwan University Suwon 16419 (Republic of Korea
| |
Collapse
|
43
|
Secretion of the siderophore rhizoferrin is regulated by the cAMP-PKA pathway and is involved in the virulence of Mucor lusitanicus. Sci Rep 2022; 12:10649. [PMID: 35739200 PMCID: PMC9226013 DOI: 10.1038/s41598-022-14515-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022] Open
Abstract
Mucormycosis is a fungal infection caused by Mucorales, with a high mortality rate. However, only a few virulence factors have been described in these organisms. This study showed that deletion of rfs, which encodes the enzyme for the biosynthesis of rhizoferrin, a siderophore, in Mucor lusitanicus, led to a lower virulence in diabetic mice and nematodes. Upregulation of rfs correlated with the increased toxicity of the cell-free supernatants of the culture broth (SS) obtained under growing conditions that favor oxidative metabolism, such as low glucose levels or the presence of H2O2 in the culture, suggesting that oxidative metabolism enhances virulence through rhizoferrin production. Meanwhile, growing M. lusitanicus in the presence of potassium cyanide, N-acetylcysteine, a higher concentration of glucose, or exogenous cAMP, or the deletion of the gene encoding the regulatory subunit of PKA (pkaR1), correlated with a decrease in the toxicity of SS, downregulation of rfs, and reduction in rhizoferrin production. These observations indicate the involvement of the cAMP-PKA pathway in the regulation of rhizoferrin production and virulence in M. lusitanicus. Moreover, rfs upregulation was observed upon macrophage interaction or during infection with spores in mice, suggesting a pivotal role of rfs in M. lusitanicus infection.
Collapse
|
44
|
de Souza AF, Pigosso LL, Silva LOS, Galo IDC, Paccez JD, e Silva KSF, de Oliveira MAP, Pereira M, Soares CMDA. Iron Deprivation Modulates the Exoproteome in Paracoccidioides brasiliensis. Front Cell Infect Microbiol 2022; 12:903070. [PMID: 35719340 PMCID: PMC9205457 DOI: 10.3389/fcimb.2022.903070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022] Open
Abstract
Fungi of the Paracoccidioides genus are the etiological agents of the systemic mycosis paracoccidioidomycosis and, when in the host, they find a challenging environment that is scarce in nutrients and micronutrients, such as Fe, which is indispensable for the survival of the pathogen. Previous studies have shown that fungi of this genus, in response to Fe deprivation, are able to synthesize and capture siderophores (Fe3+ chelators), use Fe-containing host proteins as a source of the metal, and use a non-canonical reductive pathway for Fe3+ assimilation. Despite all of these findings, there are still gaps that need to be filled in the pathogen response to metal deprivation. To contribute to the knowledge related to this subject, we obtained the exoproteome of Paracoccidioides brasiliensis (Pb18) undergoing Fe deprivation and by nanoUPLC-MSE. One hundred forty-one proteins were identified, and out of these, 64 proteins were predicted to be secreted. We also identified the regulation of several virulence factors. Among the results, we highlight Cyb5 as a secreted molecule of Paracoccidioides in the exoproteome obtained during Fe deprivation. Cyb5 is described as necessary for the Fe deprivation response of Saccharomyces cerevisiae and Aspergillus fumigatus. Experimental data and molecular modeling indicated that Cyb5 can bind to Fe ions in vitro, suggesting that it can be relevant in the arsenal of molecules related to iron homeostasis in P. brasiliensis.
Collapse
Affiliation(s)
- Aparecido Ferreira de Souza
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | - Laurine Lacerda Pigosso
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | - Lana O’Hara Souza Silva
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | - Italo Dany Cavalcante Galo
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | - Juliano Domiraci Paccez
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | - Kleber Santiago Freitas e Silva
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | | | - Maristela Pereira
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
| | - Célia Maria de Almeida Soares
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, Goiânia, Brazil
- *Correspondence: Célia Maria de Almeida Soares,
| |
Collapse
|
45
|
Peng YJ, Hou J, Zhang H, Lei JH, Lin HY, Ding JL, Feng MG, Ying SH. Systematic contributions of CFEM domain-containing proteins to iron acquisition are essential for interspecies interaction of the filamentous pathogenic fungus Beauveria bassiana. Environ Microbiol 2022; 24:3693-3704. [PMID: 35523457 DOI: 10.1111/1462-2920.16032] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/13/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022]
Abstract
Common in fungal extracellular membrane (CFEM) domain is unique in fungal proteins and some of which contribute to iron acquisition in yeast. However, their roles in iron acquisition remain largely unknown in filamentous fungi. In this study, 12 CFEM-containing proteins were bioinformatically identified in the filamentous entomopathogenic fungus Beauveria bassiana, and the roles of 11 genes were genetically characterized. Transmembrane helices were critical for their association with intracellular membranes, and their number varied among proteins. Eleven CFEM genes significantly contribute to vegetative growth under iron starvation and virulence. Notably, the virulence of most disruptants could be significantly weakened by a decrease in iron availability, in which the virulence of ΔBbcfem7 and 8 strains was partially recovered by exogenous hemin. ΔBbcfem7 and 8 mutants displayed defective competitiveness against the sister entomopathogenic fungus Beauveria brongniartii. All 11 disruptants displayed impaired growth in the antagonistic assay with the saprotrophic fungus Aspergillus niger, which could be repressed by exogenous ferric ions. These findings not only reveal the systematic contributions of CFEM proteins to acquire two forms of iron (i.e. heme and ferric ion) in the entire lifecycle of entomopathogenic fungi but also help to better understand the mechanisms of fungus-host and inter-fungus interactions.
Collapse
Affiliation(s)
- Yue-Jin Peng
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia Hou
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hao Zhang
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia-Hui Lei
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hai-Yan Lin
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jin-Li Ding
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ming-Guang Feng
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sheng-Hua Ying
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
46
|
Cryptococcus spp. and Cryptococcosis: focusing on the infection in Brazil. Braz J Microbiol 2022; 53:1321-1337. [PMID: 35486354 PMCID: PMC9433474 DOI: 10.1007/s42770-022-00744-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 03/25/2022] [Indexed: 11/02/2022] Open
Abstract
Cryptococcosis is a global fungal infection caused by the Cryptococcus neoformans/Cryptococcus gattii yeast complex. This infection is acquired by inhalation of propagules such as basidiospores or dry yeast, initially causing lung infections with the possibility of progressing to the meninges. This infection mainly affects immunocompromised HIV and transplant patients; however, immunocompetent patients can also be affected. This review proposes to evaluate cryptococcosis focusing on studies of this mycosis in Brazilian territory; moreover, recent advances in the understanding of its virulence mechanism, animal models in research are also assessed. For this, literature review as realized in PubMed, Scielo, and Brazilian legislation. In Brazil, cryptococcosis has been identified as one of the most lethal fungal infections among HIV patients and C. neoformans VNI and C. gattii VGII are the most prevalent genotypes. Moreover, different clinical settings published in Brazil were described. As in other countries, cryptococcosis is difficult to treat due to a limited therapeutic arsenal, which is highly toxic and costly. The presence of a polysaccharide capsule, thermo-tolerance, production of melanin, biofilm formation, mechanisms for iron use, and morphological alterations is an important virulence mechanism of these yeasts. The introduction of cryptococcosis as a compulsory notification disease could improve data regarding incidence and help in the management of these infections.
Collapse
|
47
|
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
|
48
|
Yamaki J, Chawla S, Tong S, Lozada KA, Yang S. Iron Effects on Clostridioides difficile Toxin Production and Antimicrobial Susceptibilities. Antibiotics (Basel) 2022; 11:537. [PMID: 35625180 PMCID: PMC9137654 DOI: 10.3390/antibiotics11050537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 03/30/2022] [Accepted: 04/18/2022] [Indexed: 12/10/2022] Open
Abstract
Despite the benefits of red blood cell (RBC) transfusion therapy, it can render patients vulnerable to iron overload. The excess iron deposits in various body tissues cause severe complications and organ damage such as cardiotoxicity and mold infections. Clostridioides difficile infection (CDI) is the most common cause of nosocomial diarrhea among cancer patients and is associated with significant morbidity and mortality. Our study aims to determine the role of iron overload and the effects of iron chelators on CDI. Our results demonstrated that iron (Fe3+) stimulated the growth of C. difficile with increased colony formation units (CFU) in a dose-dependent manner. Exposure to excess iron also increased the gene expression levels of tcdA and tcdB. The production of C. difficile toxin A, necessary for the pathogenesis of C. difficile, was also elevated after iron treatment. In the presence of excess iron, C. difficile becomes less susceptible to metronidazole with significantly elevated minimum inhibitory concentration (MIC) but remains susceptible to vancomycin. Iron-stimulated colony formation and production of C. difficile toxins were effectively diminished by iron chelator deferoxamine co-treatment. Incorporating iron overload status as a potential factor in developing a risk prediction model of CDI and antibiotic treatment response may aid clinical practitioners in optimizing CDI management in oncology patients.
Collapse
Affiliation(s)
- Jason Yamaki
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (J.Y.); (S.C.)
| | - Swati Chawla
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (J.Y.); (S.C.)
| | - Shirley Tong
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| | - Kate Alison Lozada
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| | - Sun Yang
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (J.Y.); (S.C.)
| |
Collapse
|
49
|
Soliman SSM, El-Labbad EM, Abu-Qiyas A, Fayed B, Hamoda AM, Al-Rawi AM, Dakalbab S, El-Shorbagi ANA, Hamad M, Ibrahim AS, Mohammad MG. Novel Secreted Peptides From Rhizopus arrhizus var. delemar With Immunomodulatory Effects That Enhance Fungal Pathogenesis. Front Microbiol 2022; 13:863133. [PMID: 35387075 PMCID: PMC8977774 DOI: 10.3389/fmicb.2022.863133] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/01/2022] [Indexed: 11/16/2022] Open
Abstract
Secreted fungal peptides are known to influence the interactions between the pathogen and host innate immunity. The aim of this study is to screen and evaluate secreted peptides from the fungus Rhizopus arrhizus var. delemar for their immunomodulatory activity. By using mass spectrometry and immuno-informatics analysis, we identified three secreted peptides CesT (S16), Colicin (S17), and Ca2+/calmodulin-dependent protein kinase/ligand (CAMK/CAMKL; S27). Culturing peripheral blood-derived monocytic macrophages (PBMMs) in the presence of S16 or S17 caused cell clumping, while culturing them with S27 resulted in the formation of spindle-shaped cells. S27-treated PBMMs showed cell cycle arrest at G0 phase and exhibited alternatively activated macrophage phenotype with pronounced reduction in scavenger receptors CD163 and CD206. Homology prediction indicated that IL-4/IL-13 is the immunomodulatory target of S27. Confirming this prediction, S27 initiated macrophage activation through phosphorylation of STAT-6; STAT-6 inhibition reversed the activity of S27 and reduced the formation of spindle-shaped PBMMs. Lastly, S27 treatment of PBMMs was associated with altered expression of key iron regulatory genes including hepcidin, ferroportin, transferrin receptor 1, and ferritin in a pattern consistent with increased cellular iron release; a condition known to enhance Rhizopus infection. Collectively, R. arrhizus var. delemar secretes peptides with immunomodulatory activities that support fungal pathogenesis. Targeting the IL-4/IL-13R/STAT-6 axis is a potential therapeutic approach to enhance the PBMM-mediated fungal phagocytosis. This represents a potential new approach to overcome lethal mucormycosis.
Collapse
Affiliation(s)
- Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Eman M El-Labbad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.,Pharmaceutical Sciences Department, College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
| | - Ameera Abu-Qiyas
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medical Laboratory Sciences, Collage of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Chemistry of Natural and Microbial Product Department, National Research Centre, Cairo, Egypt
| | - Alshaimaa M Hamoda
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Ahmed M Al-Rawi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medical Laboratory Sciences, Collage of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Salam Dakalbab
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medical Laboratory Sciences, Collage of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdel-Nasser A El-Shorbagi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates.,Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medical Laboratory Sciences, Collage of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ashraf S Ibrahim
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation, Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Mohammad G Mohammad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medical Laboratory Sciences, Collage of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
50
|
Palmieri F, Koutsokera A, Bernasconi E, Junier P, von Garnier C, Ubags N. Recent Advances in Fungal Infections: From Lung Ecology to Therapeutic Strategies With a Focus on Aspergillus spp. Front Med (Lausanne) 2022; 9:832510. [PMID: 35386908 PMCID: PMC8977413 DOI: 10.3389/fmed.2022.832510] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/22/2022] [Indexed: 12/15/2022] Open
Abstract
Fungal infections are estimated to be the main cause of death for more than 1.5 million people worldwide annually. However, fungal pathogenicity has been largely neglected. This is notably the case for pulmonary fungal infections, which are difficult to diagnose and to treat. We are currently facing a global emergence of antifungal resistance, which decreases the chances of survival for affected patients. New therapeutic approaches are therefore needed to face these life-threatening fungal infections. In this review, we will provide a general overview on respiratory fungal infections, with a focus on fungi of the genus Aspergillus. Next, the immunological and microbiological mechanisms of fungal pathogenesis will be discussed. The role of the respiratory mycobiota and its interactions with the bacterial microbiota on lung fungal infections will be presented from an ecological perspective. Finally, we will focus on existing and future innovative approaches for the treatment of respiratory fungal infections.
Collapse
Affiliation(s)
- Fabio Palmieri
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
- *Correspondence: Fabio Palmieri,
| | - Angela Koutsokera
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Eric Bernasconi
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Pilar Junier
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Christophe von Garnier
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Niki Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Niki Ubags,
| |
Collapse
|