1
|
Wang TW, Sofras D, Montelongo-Jauregui D, Paiva TO, Carolus H, Dufrêne YF, Alfaifi AA, McCracken C, Bruno VM, Van Dijck P, Jabra-Rizk MA. Functional redundancy in Candida auris cell surface adhesins crucial for cell-cell interaction and aggregation. Nat Commun 2024; 15:9212. [PMID: 39455573 PMCID: PMC11511831 DOI: 10.1038/s41467-024-53588-5] [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: 03/11/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Candida auris is an emerging nosocomial fungal pathogen associated with life-threatening invasive disease due to its persistent colonization, high level of transmissibility and multi-drug resistance. Aggregative and non-aggregative growth phenotypes for C. auris strains with different biofilm forming abilities, drug susceptibilities and virulence characteristics have been described. Using comprehensive transcriptional analysis we identified key cell surface adhesins that were highly upregulated in the aggregative phenotype during in vitro and in vivo grown biofilms using a mouse model of catheter infection. Phenotypic and functional evaluations of generated null mutants demonstrated crucial roles for the adhesins Als4112 and Scf1 in mediating cell-cell adherence, coaggregation and biofilm formation. While individual mutants were largely non-aggregative, in combination cells were able to co-adhere and aggregate, as directly demonstrated by measuring cell adhesion forces using single-cell atomic force spectroscopy. This co-adherence indicates their role as complementary adhesins, which despite their limited similarity, may function redundantly to promote cell-cell interaction and biofilm formation. Functional diversity of cell wall proteins may be a form of regulation that provides the aggregative phenotype of C. auris with flexibility and rapid adaptation to the environment, potentially impacting persistence and virulence.
Collapse
Affiliation(s)
- Tristan W Wang
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Dimitrios Sofras
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Telmo O Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Areej A Alfaifi
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- Department of Restorative and Prosthetic Dental Sciences, College of Dentistry King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vincent M Bruno
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD, USA
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium.
- KU Leuven One-Health Institute, KU Leuven, Leuven, Belgium.
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA.
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
2
|
Shivarathri R, Chauhan M, Datta A, Das D, Karuli A, Aptekmann A, Jenull S, Kuchler K, Thangamani S, Chowdhary A, Desai JV, Chauhan N. The Candida auris Hog1 MAP kinase is essential for the colonization of murine skin and intradermal persistence. mBio 2024:e0274824. [PMID: 39422509 DOI: 10.1128/mbio.02748-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Candida auris, a multidrug-resistant human fungal pathogen, was first identified in 2009 in Japan. Since then, systemic C. auris infections have now been reported in more than 50 countries, with mortality rates of 30%-60%. A major contributing factor to its high inter- and intrahospital clonal transmission is that C. auris, unlike most Candida species, displays unique skin tropism and can stay on human skin for a prolonged period. However, the molecular mechanisms responsible for C. auris skin colonization, intradermal persistence, and systemic virulence are poorly understood. Here, we report that C. auris Hog1 mitogen-activated protein kinase is essential for efficient skin colonization, intradermal persistence as well as systemic virulence. RNA-seq analysis of wild-type parental and hog1Δ mutant strains revealed marked downregulation of genes involved in processes such as cell adhesion, cell wall rearrangement, and pathogenesis in hog1Δ mutant compared to the wild-type parent. Consistent with these data, we found a prominent role for Hog1 in maintaining cell wall architecture, as the hog1Δ mutant demonstrated a significant increase in cell-surface β-glucan exposure and a concomitant reduction in chitin content. Additionally, we observed that Hog1 was required for biofilm formation in vitro and fungal survival when challenged with primary murine macrophages and neutrophils ex vivo. Collectively, these findings have important implications for understanding the C. auris skin adherence mechanisms and penetration of skin epithelial layers preceding bloodstream infections. IMPORTANCE Candida auris is a World Health Organization fungal priority pathogen and an urgent public health threat recognized by the Centers for Disease Control and Prevention. C. auris has a unique ability to colonize human skin. It also persists on abiotic surfaces in healthcare environments for an extended period of time. These attributes facilitate the inter- and intrahospital clonal transmission of C. auris. Therefore, understanding C. auris skin colonization mechanisms is critical for infection control, especially in hospitals and nursing homes. However, despite its profound clinical relevance, the molecular and genetic basis of C. auris skin colonization mechanisms are poorly understood. Herein, we present data on the identification of the Hog1 MAP kinase as a key regulator of C. auris skin colonization. These findings lay the foundation for further characterization of unique mechanisms that promote fungal persistence on human skin.
Collapse
Affiliation(s)
- Raju Shivarathri
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Manju Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Abhishek Datta
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Diprasom Das
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Adela Karuli
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Ariel Aptekmann
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Sabrina Jenull
- Department of Medical Biochemistry, Medical University Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karl Kuchler
- Department of Medical Biochemistry, Medical University Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Shankar Thangamani
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Anuradha Chowdhary
- Department of Medical Mycology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Jigar V Desai
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Neeraj Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
3
|
Holmes CL, Albin OR, Mobley HLT, Bachman MA. Bloodstream infections: mechanisms of pathogenesis and opportunities for intervention. Nat Rev Microbiol 2024:10.1038/s41579-024-01105-2. [PMID: 39420097 DOI: 10.1038/s41579-024-01105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
Bloodstream infections (BSIs) are common in hospitals, often life-threatening and increasing in prevalence. Microorganisms in the blood are usually rapidly cleared by the immune system and filtering organs but, in some cases, they can cause an acute infection and trigger sepsis, a systemic response to infection that leads to circulatory collapse, multiorgan dysfunction and death. Most BSIs are caused by bacteria, although fungi also contribute to a substantial portion of cases. Escherichia coli, Staphylococcus aureus, coagulase-negative Staphylococcus, Klebsiella pneumoniae and Candida albicans are leading causes of BSIs, although their prevalence depends on patient demographics and geographical region. Each species is equipped with unique factors that aid in the colonization of initial sites and dissemination and survival in the blood, and these factors represent potential opportunities for interventions. As many pathogens become increasingly resistant to antimicrobials, new approaches to diagnose and treat BSIs at all stages of infection are urgently needed. In this Review, we explore the prevalence of major BSI pathogens, prominent mechanisms of BSI pathogenesis, opportunities for prevention and diagnosis, and treatment options.
Collapse
Affiliation(s)
- Caitlyn L Holmes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Owen R Albin
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Harry L T Mobley
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael A Bachman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Huang Y, Su Y, Chen X, Xiao M, Xu Y. Insight into Virulence and Mechanisms of Amphotericin B Resistance in the Candida haemulonii Complex. J Fungi (Basel) 2024; 10:615. [PMID: 39330375 PMCID: PMC11433262 DOI: 10.3390/jof10090615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The Candida haemulonii complex includes emerging opportunistic human fungal pathogens with documented multidrug-resistance profiles. It comprises Candida haemulonii sensu stricto, Candida haemulonii var. vulnera, Candida duobushaemulonii, Candida pseudohaemulonii, and Candida vulturna. In recent years, rates of clinical isolation of strains from this complex have increased in multiple countries, including China, Malaysia, and Brazil. Biofilm formation, hydrolytic enzymes, surface interaction properties, phenotype switching and cell aggregation abilities, extracellular vesicles production, stress response, and immune evasion help these fungi to infect the host and exert pathological effects. Multidrug resistance profiles also enhance the threat they pose; they exhibit low susceptibility to echinocandins and azoles and an intrinsic resistance to amphotericin B (AMB), the first fungal-specific antibiotic. AMB is commonly employed in antifungal treatments, and it acts via several known mechanisms. Given the propensity of clinical Candida species to initiate bloodstream infections, clarifying how C. haemulonii resists AMB is of critical clinical importance. This review outlines our present understanding of the C. haemulonii complex's virulence factors, the mechanisms of action of AMB, and the mechanisms underlying AMB resistance.
Collapse
Affiliation(s)
- Yuyan Huang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- Graduate School, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yanyu Su
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- Graduate School, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Xinfei Chen
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Meng Xiao
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| |
Collapse
|
5
|
Perrier M, Barber AE. Unraveling the genomic diversity and virulence of human fungal pathogens through pangenomics. PLoS Pathog 2024; 20:e1012313. [PMID: 38990800 PMCID: PMC11238998 DOI: 10.1371/journal.ppat.1012313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Affiliation(s)
- Marion Perrier
- Junior Research Group Fungal Informatics, Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| | - Amelia E Barber
- Junior Research Group Fungal Informatics, Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
6
|
Naik B, Sasikumar J, Das SP. From Skin and Gut to the Brain: The Infectious Journey of the Human Commensal Fungus Malassezia and Its Neurological Consequences. Mol Neurobiol 2024:10.1007/s12035-024-04270-w. [PMID: 38871941 DOI: 10.1007/s12035-024-04270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
The human mycobiome encompasses diverse communities of fungal organisms residing within the body and has emerged as a critical player in shaping health and disease. While extensive research has focused on the skin and gut mycobiome, recent investigations have pointed toward the potential role of fungal organisms in neurological disorders. Among those fungal organisms, the presence of the commensal fungus Malassezia in the brain has created curiosity because of its commensal nature and primary association with the human skin and gut. This budding yeast is responsible for several diseases, such as Seborrheic dermatitis, Atopic dermatitis, Pityriasis versicolor, Malassezia folliculitis, dandruff, and others. However recent findings surprisingly show the presence of Malassezia DNA in the brain and have been linked to diseases like Alzheimer's disease, Parkinson's disease, Multiple sclerosis, and Amyotrophic lateral sclerosis. The exact role of Malassezia in these disorders is unknown, but its ability to infect human cells, travel through the bloodstream, cross the blood-brain barrier, and reside along with the lipid-rich neuronal cells are potential mechanisms responsible for pathogenesis. This also includes the induction of pro-inflammatory cytokines, disruption of the blood-brain barrier, gut-microbe interaction, and accumulation of metabolic changes in the brain environment. In this review, we discuss these key findings from studies linking Malassezia to neurological disorders, emphasizing the complex and multifaceted nature of these cases. Furthermore, we discuss potential mechanisms through which Malassezia might contribute to the development of neurological conditions. Future investigations will open up new avenues for our understanding of the fungal gut-brain axis and how it influences human behavior. Collaborative research efforts among microbiologists, neuroscientists, immunologists, and clinicians hold promise for unraveling the enigmatic connections between human commensal Malassezia and neurological disorders.
Collapse
Affiliation(s)
- Bharati Naik
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Jayaprakash Sasikumar
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Shankar Prasad Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| |
Collapse
|
7
|
Nishimura A, Tanahashi R, Nakagami K, Morioka Y, Takagi H. The arginine transporter Can1 negatively regulates biofilm formation in yeasts. Front Microbiol 2024; 15:1419530. [PMID: 38903792 PMCID: PMC11188447 DOI: 10.3389/fmicb.2024.1419530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
The arginine transporter Can1 is a multifunctional protein of the conventional yeast Saccharomyces cerevisiae. Apart from facilitating arginine uptake, Can1 plays a pivotal role in regulating proline metabolism and maintaining cellular redox balance. Here, we report a novel function of Can1 in the control of yeast biofilm formation. First, the S. cerevisiae CAN1 gene knockout strain displayed a significant growth delay compared to the wild-type strain. Our genetic screening revealed that the slow growth of the CAN1 knockout strain is rescued by a functional deficiency of the FLO8 gene, which encodes the master transcription factor associated with biofilm formation, indicating that Can1 is involved in biofilm formation. Intriguingly, the CAN1 knockout strain promoted the Flo11-dependent aggregation, leading to higher biofilm formation. Furthermore, the CAN1 knockout strain of the pathogenic yeast Candida glabrata exhibited slower growth and higher biofilm formation, similar to S. cerevisiae. More importantly, the C. glabrata CAN1 gene knockout strain showed severe toxicity to macrophage-like cells and nematodes. The present results could help to elucidate both the molecular mechanism underlying yeast biofilm formation and the role it plays. Future investigations may offer insights that contribute to development of antibiofilm agents.
Collapse
Affiliation(s)
- Akira Nishimura
- Institute for Research Initiatives, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Ryoya Tanahashi
- Institute for Research Initiatives, Nara Institute of Science and Technology, Ikoma, Nara, Japan
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Kazuki Nakagami
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Yuto Morioka
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Hiroshi Takagi
- Institute for Research Initiatives, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| |
Collapse
|
8
|
Xiong L, Goerlich K, Mitchell AP. Regulatory features of Candida albicans hemin-induced filamentation. G3 (BETHESDA, MD.) 2024; 14:jkae053. [PMID: 38470537 PMCID: PMC11075532 DOI: 10.1093/g3journal/jkae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
Candida albicans is a prominent fungal pathogen that can infect the bloodstream and deep tissues. One key pathogenicity trait is the ability to transition between yeast and hyphal growth. Hyphae are critical for the formation of biofilms, which in turn enable device-associated infection. Among signals that drive hypha formation is the presence of hemin, an oxidized Fe(III)-containing heme derivative found in blood. In this study, we asked 4 questions. First, how uniform is the filamentation response to hemin among C. albicans strains? We tested 26 diverse isolates and found that the strength of a strain's filamentation response to hemin reflected its filamentation level in the absence of hemin. Second, does hemin induce biofilm formation? Hemin biofilm induction was evident in 5 out of 10 isolates tested, including most of the weaker biofilm formers tested. Third, what is the gene expression response to hemin? We compared RNA-seq data for type strain SC5314 grown in pH 5.5 minimal media with or without hemin. We also compared that response to SC5314 grown in pH 7.0 minimal media, where it undergoes well-studied pH-dependent filamentation. We found a common set of 72 genes with upregulated RNA levels in response to both signals, including many known hypha-associated genes. Surprisingly, overlap among those 72 genes with 2 recent consensus definitions of hypha-associated genes was limited to only 16 genes. Fourth, which regulators govern hemin-induced filamentation? A mutant survey indicated that the response depends upon filamentation regulators Efg1, Brg1, and Rim101, but not upon heme acquisition regulator Hap1 or its target genes HMX1, RBT5, PGA10, PGA7, and CSA2. These findings argue that hemin induces hypha formation independently of its utilization.
Collapse
Affiliation(s)
- Liping Xiong
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Katharina Goerlich
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Aaron P Mitchell
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
9
|
Ichikawa T, Ikeda Y, Sadanaga J, Kikuchi A, Kawamura K, Ikeda R, Ishibashi Y. Identification of heparin-binding proteins expressed on Trichosporon asahii cell surface. Yeast 2024; 41:299-306. [PMID: 38297467 DOI: 10.1002/yea.3928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/02/2024] Open
Abstract
Trichosporon asahii is a pathogenic yeast that cause trichosporonosis. T. asahii exhibits several colony morphologies, such as white (W)- or off-white (O)-type, which may affect virulence. In this study, we compared the expression pattern of heparin-binding proteins in various colony morphologies and identified heparin-binding protein in T. asahii. Surface plasmon resonance analysis revealed that cell surface molecules attached more strongly to heparin in W- than O-type cells. We purified and identified a heparin-binding protein strongly expressed in W-type cells using heparin-Sepharose beads, named it heparin-binding protein 1 (HepBP1), and expressed Flag-tagged HepBP1 in mammalian cells. The heparin-binding ability of Flag-tagged HepBP1 was confirmed by pulldown assay using heparin-Sepharose beads. Thus, HepBP1 is a heparin-binding protein on T. asahii cell surface. These results suggest that several T. asahii cell surface proteins interact with glycosaminoglycans; therefore, they could contribute to infection.
Collapse
Affiliation(s)
- Tomoe Ichikawa
- Department of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Yokohama, Japan
| | - Yuka Ikeda
- Meiji Pharmaceutical University, Kiyose, Japan
| | | | | | | | - Reiko Ikeda
- Meiji Pharmaceutical University, Kiyose, Japan
| | - Yoshio Ishibashi
- Department of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Yokohama, Japan
| |
Collapse
|
10
|
Wang TW, Sofras D, Montelongo-Jauregui D, Paiva TO, Carolus H, Dufrêne YF, Alfaifi AA, McCracken C, Bruno VM, Van Dijck P, Jabra-Rizk MA. Functional Redundancy in Candida auris Cell Surface Adhesins Crucial for Cell-Cell Interaction and Aggregation. RESEARCH SQUARE 2024:rs.3.rs-4077218. [PMID: 38562859 PMCID: PMC10984083 DOI: 10.21203/rs.3.rs-4077218/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Candida auris is an emerging nosocomial fungal pathogen associated with life-threatening invasive disease due to its persistent colonization, high level of transmissibility and multi-drug resistance. Aggregative and non-aggregative growth phenotypes for C. auris strains with different biofilm forming abilities, drug susceptibilities and virulence characteristics have been described. Using comprehensive transcriptional analysis we identified key cell surface adhesins that were highly upregulated in the aggregative phenotype during in vitro and in vivo grown biofilms using a mouse model of catheter infection. Phenotypic and functional evaluations of generated null mutants demonstrated crucial roles for the adhesins Als5 and Scf1 in mediating cell-cell adherence, coaggregation and biofilm formation. While individual mutants were largely non-aggregative, in combination cells were able to co-adhere and aggregate, as directly demonstrated by measuring cell adhesion forces using single-cell atomic force spectroscopy. This co-adherence indicates their role as complementary adhesins, which despite their limited similarity, may function redundantly to promote cell-cell interaction and biofilm formation. Functional diversity of cell wall proteins may be a form of regulation that provides the aggregative phenotype of C. auris with flexibility and rapid adaptation to the environment, potentially impacting persistence and virulence.
Collapse
Affiliation(s)
- Tristan W. Wang
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Dimitrios Sofras
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Telmo O. Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Yves F. Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Areej A. Alfaifi
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Vincent M. Bruno
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD 21201, USA
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
11
|
Wang TW, Sofras D, Montelongo-Jauregui D, Paiva TO, Carolus H, Dufrêne YF, Alfaifi AA, McCracken C, Bruno VM, Van Dijck P, Jabra-Rizk MA. Functional Redundancy in Candida auris Cell Surface Adhesins Crucial for Cell-Cell Interaction and Aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586120. [PMID: 38562758 PMCID: PMC10983922 DOI: 10.1101/2024.03.21.586120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Candida auris is an emerging nosocomial fungal pathogen associated with life-threatening invasive disease due to its persistent colonization, high level of transmissibility and multi-drug resistance. Aggregative and non-aggregative growth phenotypes for C. auris strains with different biofilm forming abilities, drug susceptibilities and virulence characteristics have been described. Using comprehensive transcriptional analysis we identified key cell surface adhesins that were highly upregulated in the aggregative phenotype during in vitro and in vivo grown biofilms using a mouse model of catheter infection. Phenotypic and functional evaluations of generated null mutants demonstrated crucial roles for the adhesins Als5 and Scf1 in mediating cell-cell adherence, coaggregation and biofilm formation. While individual mutants were largely non-aggregative, in combination cells were able to co-adhere and aggregate, as directly demonstrated by measuring cell adhesion forces using single-cell atomic force spectroscopy. This co-adherence indicates their role as complementary adhesins, which despite their limited similarity, may function redundantly to promote cell-cell interaction and biofilm formation. Functional diversity of cell wall proteins may be a form of regulation that provides the aggregative phenotype of C. auris with flexibility and rapid adaptation to the environment, potentially impacting persistence and virulence.
Collapse
Affiliation(s)
- Tristan W. Wang
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Dimitrios Sofras
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Telmo O. Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Yves F. Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Areej A. Alfaifi
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Vincent M. Bruno
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD 21201, USA
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, School of Medicine University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Shivarathri R, Chauhan M, Datta A, Das D, Karuli A, Jenull S, Kuchler K, Thangamani S, Chowdhary A, Desai JV, Chauhan N. The Candida auris Hog1 MAP kinase is essential for the colonization of murine skin and intradermal persistence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585572. [PMID: 38562863 PMCID: PMC10983919 DOI: 10.1101/2024.03.18.585572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Candida auris , a multidrug-resistant human fungal pathogen, was first identified in 2009 in Japan. Since then, systemic C. auris infections have now been reported in more than 50 countries, with mortality rates of 30-60%. A major contributing factor to its high inter- and intrahospital clonal transmission is that C. auris, unlike most Candida species, displays unique skin tropism and can stay on human skin for a prolonged period. However, the molecular mechanisms responsible for C. auris skin colonization, intradermal persistence, and systemic virulence are poorly understood. Here, we report that C. auris Hog1 mitogen-activated protein kinase (MAPK) is essential for efficient skin colonization, intradermal persistence, as well as systemic virulence. RNA-seq analysis of wildtype parental and hog1 Δ mutant strains revealed marked down-regulation of genes involved in processes such as cell adhesion, cell-wall rearrangement, and pathogenesis in hog1 Δ mutant compared to the wildtype parent. Consistent with these data, we found a prominent role for Hog1 in maintaining cell-wall architecture, as the hog1 Δ mutant demonstrated a significant increase in cell-surface β-glucan exposure and a concomitant reduction in chitin content. Additionally, we observed that Hog1 was required for biofilm formation in vitro and fungal survival when challenged with primary murine macrophages and neutrophils ex vivo . Collectively, these findings have important implications for understanding the C. auris skin adherence mechanisms and penetration of skin epithelial layers preceding bloodstream infections. Importance Candida auris is a World Health Organization (WHO) fungal priority pathogen and an urgent public health threat recognized by the Centers for Disease Control and Prevention (CDC). C. auris has a unique ability to colonize human skin. It also persists on abiotic surfaces in healthcare environments for an extended period of time. These attributes facilitate the inter- and intrahospital clonal transmission of C. auris . Therefore, understanding C. auris skin colonization mechanisms are critical for infection control, especially in hospitals and nursing homes. However, despite its profound clinical relevance, the molecular and genetic basis of C. auris skin colonization mechanisms are poorly understood. Herein, we present data on the identification of the Hog1 MAP kinase as a key regulator of C. auris skin colonization. These findings lay foundation for further characterization of unique mechanisms that promote fungal persistence on human skin.
Collapse
|
13
|
John E, Chau MQ, Hoang CV, Chandrasekharan N, Bhaskar C, Ma LS. Fungal Cell Wall-Associated Effectors: Sensing, Integration, Suppression, and Protection. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2024; 37:196-210. [PMID: 37955547 DOI: 10.1094/mpmi-09-23-0142-fi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The cell wall (CW) of plant-interacting fungi, as the direct interface with host plants, plays a crucial role in fungal development. A number of secreted proteins are directly associated with the fungal CW, either through covalent or non-covalent interactions, and serve a range of important functions. In the context of plant-fungal interactions many are important for fungal development in the host environment and may therefore be considered fungal CW-associated effectors (CWAEs). Key CWAE functions include integrating chemical/physical signals to direct hyphal growth, interfering with plant immunity, and providing protection against plant defenses. In recent years, a diverse range of mechanisms have been reported that underpin their roles, with some CWAEs harboring conserved motifs or functional domains, while others are reported to have novel features. As such, the current understanding regarding fungal CWAEs is systematically presented here from the perspective of their biological functions in plant-fungal interactions. An overview of the fungal CW architecture and the mechanisms by which proteins are secreted, modified, and incorporated into the CW is first presented to provide context for their biological roles. Some CWAE functions are reported across a broad range of pathosystems or symbiotic/mutualistic associations. Prominent are the chitin interacting-effectors that facilitate fungal CW modification, protection, or suppression of host immune responses. However, several alternative functions are now reported and are presented and discussed. CWAEs can play diverse roles, some possibly unique to fungal lineages and others conserved across a broad range of plant-interacting fungi. [Formula: see text] Copyright © 2024 The Author(s). This is an open access article distributed under the CC BY 4.0 International license.
Collapse
Affiliation(s)
- Evan John
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Minh-Quang Chau
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Cuong V Hoang
- Centro de Biotecnología y Genómica de Plantas, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Universidad Politécnica de Madrid (UPM), Campus de Montegancedo UPM, 28223 Pozuelo de Alarcón, Spain
| | | | - Chibbhi Bhaskar
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Lay-Sun Ma
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
14
|
Shah K, Deshpande M, Shah P. Healthcare-associated fungal infections and emerging pathogens during the COVID-19 pandemic. FRONTIERS IN FUNGAL BIOLOGY 2024; 5:1339911. [PMID: 38465254 PMCID: PMC10920311 DOI: 10.3389/ffunb.2024.1339911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024]
Abstract
Historically, fungi were mainly identified as plant and insect pathogens since they grow at 28°C. At the same time, bacteria are known to be the most common human pathogens as they are compatible with the host body temperature of 37°C. Because of immunocompromised hosts, cancer therapy, and malnutrition, fungi are rapidly gaining attention as human pathogens. Over 150 million people have severe fungal infections, which lead to approximately more than one million deaths per year. Moreover, diseases like cancer involving long-term therapy and prophylactic use of antifungal drugs in high-risk patients have increased the emergence of drug-resistant fungi, including highly virulent strains such as Candida auris. This clinical spectrum of fungal diseases ranges from superficial mucocutaneous lesions to more severe and life-threatening infections. This review article summarizes the effect of hospital environments, especially during the COVID-19 pandemic, on fungal infections and emerging pathogens. The review also provides insights into the various antifungal drugs and their existing challenges, thereby driving the need to search for novel antifungal agents.
Collapse
Affiliation(s)
- Krish Shah
- Biological Sciences Bellarmine College Preparatory, San Jose, CA, United States
| | | | - P. Shah
- Science Ambassador/Bio-Rad Laboratories, Hercules, CA, United States
| |
Collapse
|
15
|
Yadav A, Yadav R, Sharma V, Dutta U. A comprehensive guide to assess gut mycobiome and its role in pathogenesis and treatment of inflammatory bowel disease. Indian J Gastroenterol 2024; 43:112-128. [PMID: 38409485 DOI: 10.1007/s12664-023-01510-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/20/2023] [Indexed: 02/28/2024]
Abstract
Inflammatory bowel disease (IBD) is an immune mediated chronic inflammatory disorder of gastrointestinal tract, which has underlying multifactorial pathogenic determinants such as environmental factors, susceptibility genes, gut microbial dysbiosis and a dysregulated immune response. Human gut is a frequent inhabitant of complex microbial ecosystem encompassing bacteria, viruses, parasites, fungi and other microorganisms that have an undisputable role in maintaining balanced homeostasis. All of these microbes interact with immune system and affect human gut physiology either directly or indirectly with interaction of each other. Intestinal fungi represent a smaller but crucial component of the human gut microbiome. Besides interaction with bacteriome and virome, it helps in balancing homoeostasis between pathophysiological and physiological processes, which is often dysregulated in patients with IBD. Understanding of gut mycobiome and its clinical implications are still in in its infancy as opposed to bacterial component of gut microbiome, which is more often focused. Modulation of gut mycobiome represents a novel and promising strategy in the management of patients with IBD. Emerging mycobiome-based therapies such as diet interventions, fecal microbiota transplantation (FMT), probiotics (both fungal and bacterial strains) and antifungals exhibit substantial effects in calibrating the gut mycobiome and restoring dysbalanced immune homeostasis by restoring the core gut mycobiome. In this review, we summarized compositional and functional diversity of the gut mycobiome in healthy individuals and patients with IBD, gut mycobiome dysbiosis in patients with IBD, host immune-fungal interactions and therapeutic role of modulation of intestinal fungi in patients with IBD.
Collapse
Affiliation(s)
- Amit Yadav
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160 012, India
| | - Renu Yadav
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, 110 029, India
| | - Vishal Sharma
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160 012, India
| | - Usha Dutta
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160 012, India.
| |
Collapse
|
16
|
Gómez-Gaviria M, García-Carnero LC, Baruch-Martínez DA, Mora-Montes HM. The Emerging Pathogen Candida metapsilosis: Biological Aspects, Virulence Factors, Diagnosis, and Treatment. Infect Drug Resist 2024; 17:171-185. [PMID: 38268929 PMCID: PMC10807450 DOI: 10.2147/idr.s448213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/16/2024] [Indexed: 01/26/2024] Open
Abstract
Fungal infections represent a constant and growing menace to public health. This concern is due to the emergence of new fungal species and the increase in antifungal drug resistance. Mycoses caused by Candida species are among the most common nosocomial infections and are associated with high mortality rates when the infection affects deep-seated organs. Candida metapsilosis is part of the Candida parapsilosis complex and has been described as part of the oral microbiota of healthy individuals. Within the complex, this species is considered the least virulent; however, the prevalence has been increasing in recent years, as well as an increment in the resistance to some antifungal drugs. One of the main concerns of candidiasis caused by this species is the wide range of clinical manifestations, ranging from tissue colonization to superficial infections, and in more severe cases it can spread, which makes diagnosis and treatment difficult. The study of virulence factors of this species is limited, however, proteomic comparisons between species indicate that virulence factors in this species could be similar to those already described for C. albicans. However, differences may exist, taking into account changes in the lifestyle of the species. Here, we provide a detailed review of the current literature about this organism, the caused disease, and some sharing aspects with other members of the complex, focusing on its biology, virulence factors, the host-fungus interaction, the identification, diagnosis, and treatment of infection.
Collapse
Affiliation(s)
- Manuela Gómez-Gaviria
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto., México
| | - Laura C García-Carnero
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto., México
| | - Dario A Baruch-Martínez
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto., México
| | - Héctor M Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Gto., México
| |
Collapse
|
17
|
Lim SJ, Muhd Noor ND, Sabri S, Mohamad Ali MS, Salleh AB, Oslan SN. Bibliometric analysis and thematic review of Candida pathogenesis: Fundamental omics to applications as potential antifungal drugs and vaccines. Med Mycol 2024; 62:myad126. [PMID: 38061839 DOI: 10.1093/mmy/myad126] [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: 09/03/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024] Open
Abstract
Invasive candidiasis caused by the pathogenic Candida yeast species has resulted in elevating global mortality. The pathogenicity of Candida spp. is not only originated from its primary invasive yeast-to-hyphal transition; virulence factors (transcription factors, adhesins, invasins, and enzymes), biofilm, antifungal drug resistance, stress tolerance, and metabolic adaptation have also contributed to a greater clinical burden. However, the current research theme in fungal pathogenicity could hardly be delineated with the increasing research output. Therefore, our study analysed the research trends in Candida pathogenesis over the past 37 years via a bibliometric approach against the Scopus and Web of Science databases. Based on the 3993 unique documents retrieved, significant international collaborations among researchers were observed, especially between Germany (Bernhard Hube) and the UK (Julian Naglik), whose focuses are on Candida proteinases, adhesins, and candidalysin. The prominent researchers (Neils Gow, Alistair Brown, and Frank Odds) at the University of Exeter and the University of Aberdeen (second top performing affiliation) UK contribute significantly to the mechanisms of Candida adaptation, tolerance, and stress response. However, the science mapping of co-citation analysis performed herein could not identify a hub representative of subsequent work since the clusters were semi-redundant. The co-word analysis that was otherwise adopted, revealed three research clusters; the cluster-based thematic analyses indicated the severeness of Candida biofilm and antifungal resistance as well as the elevating trend on molecular mechanism elucidation for drug screening and repurposing. Importantly, the in vivo pathogen adaptation and interactions with hosts are crucial for potential vaccine development.
Collapse
Affiliation(s)
- Si Jie Lim
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Noor Dina Muhd Noor
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Suriana Sabri
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Mohd Shukuri Mohamad Ali
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Abu Bakar Salleh
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Siti Nurbaya Oslan
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Enzyme and Microbial Technology (EMTech) Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
18
|
Zeng G, Xu X, Kok YJ, Deng FS, Ling Chow EW, Gao J, Bi X, Wang Y. Cytochrome c regulates hyphal morphogenesis by interfering with cAMP-PKA signaling in Candida albicans. Cell Rep 2023; 42:113473. [PMID: 37980562 DOI: 10.1016/j.celrep.2023.113473] [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: 06/19/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023] Open
Abstract
In the human fungal pathogen Candida albicans, invasive hyphal growth is a well-recognized virulence trait. We employed transposon-mediated genome-wide mutagenesis, revealing that inactivating CTM1 blocks hyphal growth. CTM1 encodes a lysine (K) methyltransferase, which trimethylates cytochrome c (Cyc1) at K79. Mutants lacking CTM1 or expressing cyc1K79A grow as yeast under hyphae-inducing conditions, indicating that unmethylated Cyc1 suppresses hyphal growth. Transcriptomic analyses detected increased levels of the hyphal repressor NRG1 and decreased levels of hyphae-specific genes in ctm1Δ/Δ and cyc1K79A mutants, suggesting cyclic AMP (cAMP)-protein kinase A (PKA) signaling suppression. Co-immunoprecipitation and in vitro kinase assays demonstrated that unmethylated Cyc1 inhibits PKA kinase activity. Surprisingly, hyphae-defective ctm1Δ/Δ and cyc1K79A mutants remain virulent in mice due to accelerated proliferation. Our results unveil a critical role for cytochrome c in maintaining the virulence of C. albicans by orchestrating proliferation, growth mode, and metabolism. Importantly, this study identifies a biological function for lysine methylation on cytochrome c.
Collapse
Affiliation(s)
- Guisheng Zeng
- A(∗)STAR Infectious Diseases Labs (A(∗)STAR ID Labs), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore.
| | - Xiaoli Xu
- A(∗)STAR Infectious Diseases Labs (A(∗)STAR ID Labs), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology Institute, 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Fu-Sheng Deng
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Eve Wai Ling Chow
- A(∗)STAR Infectious Diseases Labs (A(∗)STAR ID Labs), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore
| | - Jiaxin Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuezhi Bi
- Bioprocessing Technology Institute, 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
| | - Yue Wang
- A(∗)STAR Infectious Diseases Labs (A(∗)STAR ID Labs), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
19
|
Abramov VM, Kosarev IV, Machulin AV, Priputnevich TV, Deryusheva EI, Panin AN, Chikileva IO, Abashina TN, Melnikov VG, Suzina NE, Nikonov IN, Akhmetzyanova AA, Khlebnikov VS, Sakulin VK, Vasilenko RN, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. Protective Properties of S-layer Protein 2 from Lactobacillus crispatus 2029 against Candida albicans Infections. Biomolecules 2023; 13:1740. [PMID: 38136611 PMCID: PMC10741940 DOI: 10.3390/biom13121740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Previously, the protective role of the S-layer protein 2 (Slp2) of the vaginal Lactobacillus crispatus 2029 (LC2029) strain against foodborne pathogens Campylobacter jejuni, Salmonella enterica serovar Enteritidis, and Escherichia coli O157:H was demonstrated. We demonstrate the new roles of the Slp2-positive LC2029 strain and soluble Slp2 against C. albicans infections. We show that LC2029 bacteria can adhere to the surface of the cervical epithelial HeLa cells, prevent their contact with C. albicans, and block yeast transition to a pathogenic hyphal form. Surface-bound Slp2 provides the ability for LC2029 to co-aggregate with various C. albicans strains, including clinical isolates. C. albicans-induced necrotizing epithelial damage is reduced by colonization with the Slp2-positive LC2029 strain. Slp2 inhibits the adhesion of various strains of C. albicans to different human epithelial cells, blocks yeast transition to a pathogenic hyphal form, and prevents the colonization and pathogenic infiltration of mucosal barriers. Only Slp2 and LC2029 bacteria stimulate the production of protective human β-defensin 3 in various epithelial cells. These findings support the anti-Candida albicans potential of the probiotic LC2029 strain and Slp2 and form the basis for further research on their ability to prevent and manage invasive Candida infections.
Collapse
Affiliation(s)
- Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia (A.N.P.)
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia (A.N.P.)
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia;
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia (A.N.P.)
| | - Irina O. Chikileva
- Laboratory of Cell Immunity, Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia;
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ilia N. Nikonov
- Federal State Educational Institution of Higher Professional Education Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia
| | - Anna A. Akhmetzyanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia (A.N.P.)
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia (R.N.V.)
| | - Raisa N. Vasilenko
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia (R.N.V.)
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, School of Life Sciences, Chemistry and Pharmacy, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK;
| |
Collapse
|
20
|
Carlson SL, Mathew L, Savage M, Kok K, Lindsay JO, Munro CA, McCarthy NE. Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease. J Fungi (Basel) 2023; 9:1105. [PMID: 37998910 PMCID: PMC10672531 DOI: 10.3390/jof9111105] [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: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023] Open
Abstract
The gut microbiome is a diverse microbial community composed of bacteria, viruses, and fungi that plays a major role in human health and disease. Dysregulation of these gut organisms in a genetically susceptible host is fundamental to the pathogenesis of inflammatory bowel disease (IBD). While bacterial dysbiosis has been a predominant focus of research for many years, there is growing recognition that fungal interactions with the host immune system are an important driver of gut inflammation. Candida albicans is likely the most studied fungus in the context of IBD, being a near universal gut commensal in humans and also a major barrier-invasive pathogen. There is emerging evidence that intra-strain variation in C. albicans virulence factors exerts a critical influence on IBD pathophysiology. In this review, we describe the immunological impacts of variations in C. lbicans colonisation, morphology, genetics, and proteomics in IBD, as well as the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Sean L. Carlson
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
- Gastroenterology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Liya Mathew
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Michael Savage
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Klaartje Kok
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
- Gastroenterology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - James O. Lindsay
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
- Gastroenterology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Carol A. Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Neil E. McCarthy
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
21
|
Ndlovu E, Malpartida L, Sultana T, Dahms TES, Dague E. Host Cell Geometry and Cytoskeletal Organization Governs Candida-Host Cell Interactions at the Nanoscale. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37888912 DOI: 10.1021/acsami.3c09870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Candida is one of the most common opportunistic fungal pathogens in humans. Its adhesion to the host cell is required in parasitic states and is important for pathogenesis. Many studies have shown that there is an increased risk of developing candidiasis when normal tissue barriers are weakened or when immune defenses are compromised, for example, during cancer treatment that induces immunosuppression. The mechanical properties of malignant cells, such as adhesiveness and viscoelasticity, which contribute to cellular invasion and migration are different from those of noncancerous cells. To understand host invasion and its relationship with host cell health, we probed the interaction of Candida spp. with cancerous and noncancerous human cell lines using atomic force microscopy in the single-cell force spectroscopy mode. There was significant adhesion between Candida and human cells, with more adhesion to cancerous versus noncancerous cell lines. This increase in adhesion is related to the mechanobiological properties of cancer cells, which have a disorganized cytoskeleton and lower rigidity. Altered geometry and cytoskeletal disruption of the human cells impacted adhesion parameters, underscoring the role of cytoskeletal organization in Candida-human cell adhesion and implicating the manipulation of cell properties as a potential future therapeutic strategy.
Collapse
Affiliation(s)
- Easter Ndlovu
- Department of Chemistry and Biochemistry, University of Regina, 3737 Wascana Parkway, Regina S4S 0A2, Saskatchewan, Canada
| | - Lucas Malpartida
- National Centre for Scientific Research, Laboratory for Analysis and Architecture of Systems (LAAS), 7 Avenue du Colonel Roche, BP 54200, Toulouse cedex 4 31031, France
| | - Taranum Sultana
- Department of Chemistry and Biochemistry, University of Regina, 3737 Wascana Parkway, Regina S4S 0A2, Saskatchewan, Canada
| | - Tanya E S Dahms
- Department of Chemistry and Biochemistry, University of Regina, 3737 Wascana Parkway, Regina S4S 0A2, Saskatchewan, Canada
| | - Etienne Dague
- National Centre for Scientific Research, Laboratory for Analysis and Architecture of Systems (LAAS), 7 Avenue du Colonel Roche, BP 54200, Toulouse cedex 4 31031, France
| |
Collapse
|
22
|
Santana DJ, Anku JAE, Zhao G, Zarnowski R, Johnson CJ, Hautau H, Visser ND, Ibrahim AS, Andes D, Nett JE, Singh S, O'Meara TR. A Candida auris-specific adhesin, Scf1 , governs surface association, colonization, and virulence. Science 2023; 381:1461-1467. [PMID: 37769084 PMCID: PMC11235122 DOI: 10.1126/science.adf8972] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/23/2023] [Indexed: 09/30/2023]
Abstract
Candida auris is an emerging fungal pathogen responsible for health care-associated outbreaks that arise from persistent surface and skin colonization. We characterized the arsenal of adhesins used by C. auris and discovered an uncharacterized adhesin, Surface Colonization Factor (Scf1), and a conserved adhesin, Iff4109, that are essential for the colonization of inert surfaces and mammalian hosts. SCF1 is apparently specific to C. auris, and its expression mediates adhesion to inert and biological surfaces across isolates from all five clades. Unlike canonical fungal adhesins, which function through hydrophobic interactions, Scf1 relies on exposed cationic residues for surface association. SCF1 is required for C. auris biofilm formation, skin colonization, virulence in systemic infection, and colonization of inserted medical devices.
Collapse
Affiliation(s)
- Darian J Santana
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Juliet A E Anku
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Guolei Zhao
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Robert Zarnowski
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Chad J Johnson
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Haley Hautau
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles Medical Center, Torrance, CA, USA
| | - Noelle D Visser
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Ashraf S Ibrahim
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Andes
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Jeniel E Nett
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Shakti Singh
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Teresa R O'Meara
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Malinovská Z, Čonková E, Váczi P. Biofilm Formation in Medically Important Candida Species. J Fungi (Basel) 2023; 9:955. [PMID: 37888211 PMCID: PMC10607155 DOI: 10.3390/jof9100955] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/01/2023] [Accepted: 08/18/2023] [Indexed: 10/28/2023] Open
Abstract
Worldwide, the number of infections caused by biofilm-forming fungal pathogens is very high. In human medicine, there is an increasing proportion of immunocompromised patients with prolonged hospitalization, and patients with long-term inserted drains, cannulas, catheters, tubes, or other artificial devices, that exhibit a predisposition for colonization by biofilm-forming yeasts. A high percentage of mortality is due to candidemia caused by medically important Candida species. Species of major clinical significance include C. albicans, C. glabrata, C. tropicalis, C. parapsilosis, C. krusei, and C. auris. The association of these pathogenic species in the biofilm structure is a serious therapeutic problem. Candida cells growing in the form of a biofilm are able to resist persistent therapy thanks to a combination of their protective mechanisms and their ability to disseminate to other parts of the body, thus representing a threat from the perspective of a permanent source of infection. The elucidation of the key mechanisms of biofilm formation is essential to progress in the understanding and treatment of invasive Candida infections.
Collapse
Affiliation(s)
- Zuzana Malinovská
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81 Košice, Slovakia; (E.Č.); (P.V.)
| | | | | |
Collapse
|
24
|
Badrane H, Cheng S, Dupont CL, Hao B, Driscoll E, Morder K, Liu G, Newbrough A, Fleres G, Kaul D, Espinoza JL, Clancy CJ, Nguyen MH. Genotypic diversity and unrecognized antifungal resistance among populations of Candida glabrata from positive blood cultures. Nat Commun 2023; 14:5918. [PMID: 37739935 PMCID: PMC10516878 DOI: 10.1038/s41467-023-41509-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023] Open
Abstract
The longstanding model is that most bloodstream infections (BSIs) are caused by a single organism. We perform whole genome sequencing of five-to-ten strains from blood culture (BC) bottles in each of ten patients with Candida glabrata BSI. We demonstrate that BCs contain mixed populations of clonal but genetically diverse strains. Genetically distinct strains from two patients exhibit phenotypes that are potentially important during BSIs, including differences in susceptibility to antifungal agents and phagocytosis. In both patients, the clinical microbiology lab recovered a fluconazole-susceptible index strain, but we identify mixed fluconazole-susceptible and -resistant populations. Diversity in drug susceptibility is likely clinically relevant, as fluconazole-resistant strains were subsequently recovered by the clinical laboratory during persistent or relapsing infections. In one patient, unrecognized respiration-deficient small colony variants are fluconazole-resistant and significantly attenuated for virulence during murine candidiasis. Our data suggest a population-based model of C. glabrata genotypic and phenotypic diversity during BSIs.
Collapse
Affiliation(s)
| | | | | | - Binghua Hao
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Guojun Liu
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Drishti Kaul
- J. Craig Venter Institute, La Jolla, CA, 92037, USA
| | | | - Cornelius J Clancy
- University of Pittsburgh, Pittsburgh, PA, USA
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | | |
Collapse
|
25
|
Abstract
Candida auris is a multidrug-resistant fungal pathogen that presents a serious threat to global human health. Since the first reported case in 2009 in Japan, C. auris infections have been reported in more than 40 countries, with mortality rates between 30% and 60%. In addition, C. auris has the potential to cause outbreaks in health care settings, especially in nursing homes for elderly patients, owing to its efficient transmission via skin-to-skin contact. Most importantly, C. auris is the first fungal pathogen to show pronounced and sometimes untreatable clinical drug resistance to all known antifungal classes, including azoles, amphotericin B, and echinocandins. In this review, we explore the causes of the rapid spread of C. auris. We also highlight its genome organization and drug resistance mechanisms and propose future research directions that should be undertaken to curb the spread of this multidrug-resistant pathogen.
Collapse
Affiliation(s)
- Anuradha Chowdhary
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India;
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Kusum Jain
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India;
| | - Neeraj Chauhan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
26
|
Silva RRS, Malveira EA, Aguiar TKB, Neto NAS, Roma RR, Santos MHC, Santos ALE, Silva AFB, Freitas CDT, Rocha BAM, Souza PFN, Teixeira CS. DVL, lectin from Dioclea violacea seeds, has multiples mechanisms of action against Candida spp via carbohydrate recognition domain. Chem Biol Interact 2023; 382:110639. [PMID: 37468117 DOI: 10.1016/j.cbi.2023.110639] [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: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
Lectins are proteins of non-immunological origin with the ability to bind to carbohydrates reversibly. They emerge as an alternative to conventional antifungals, given the ability to interact with carbohydrates in the fungal cell wall inhibiting fungal growth. The lectin from D. violacea (DVL) already has its activity described as anti-candida in some species. Here, we observed the anti-candida effect of DVL on C. albicans, C. krusei and C. parapsilosis and its multiple mechanisms of action toward the yeasts. Additionally, it was observed that DVL induces membrane and cell wall damage and ROS overproduction. DVL was also able to cause an imbalance in the redox system of the cells, interact with ergosterol, inhibit ergosterol biosynthesis, and induce cytochrome c release from the mitochondrial membrane. These results endorse the potential application of DVL in developing a new antifungal drug to fight back against fungal resistance.
Collapse
Affiliation(s)
- Romério R S Silva
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Ellen A Malveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Tawanny K B Aguiar
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Nilton A S Neto
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Renato R Roma
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Maria H C Santos
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Ana L E Santos
- Medical School, Federal University of Cariri, Barbalha, Ceará, Brazil
| | - Ayrles F B Silva
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Cleverson D T Freitas
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Bruno A M Rocha
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil
| | - Pedro F N Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, 60451-970, CE, Brazil; Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.
| | - Claudener S Teixeira
- Center for Agricultural Sciences and Biodiversity, Federal University of Cariri, Crato, 63130-025, Brazil.
| |
Collapse
|
27
|
Pal K, Kundu S, Wang X. Macrophages form integrin-mediated adhesion rings to pinch off surface-bound objects for phagocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551462. [PMID: 37577702 PMCID: PMC10418108 DOI: 10.1101/2023.08.01.551462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Macrophages engulf micron-sized objects including pathogens and cell debris by phagocytosis, serving a fundamental role in immune defense and homeostasis 1, 2 . Although the internalization process of suspended particles has been thoroughly investigated 3, 4 , it is incompletely understood how macrophages internalize surface-bound objects by overcoming the surface binding. Here, we prepared a force-sensing platform which visualizes cell-substrate adhesive force by fluorescence. Macrophages are tested on this platform with micron-sized objects (E. coli, microbeads and silver nanorods) immobilized. By co-imaging integrin-transmitted forces and corresponding structural proteins, we discovered that macrophages consistently form integrin-mediated adhesion structures on the surface to encircle and pinch off surface-bound objects. We termed these structures phagocytic adhesion rings (PAR) and showed that integrin tensions in PARs are resulted from local actin polymerization, but not from myosin II. We further demonstrated that the intensity of integrin tensions in PARs is correlated with the object surface-bound strength, and the integrin ligand strength (dictating the upper limit of integrin tensions) determines the phagocytosis efficiency. Collectively, this study revealed a new phagocytosis mechanism that macrophages form PARs to provide physical anchorage for local F-actin polymerization that pushes and lifts off surface-bound objects during phagocytosis.
Collapse
|
28
|
Rapala-Kozik M, Surowiec M, Juszczak M, Wronowska E, Kulig K, Bednarek A, Gonzalez-Gonzalez M, Karkowska-Kuleta J, Zawrotniak M, Satała D, Kozik A. Living together: The role of Candida albicans in the formation of polymicrobial biofilms in the oral cavity. Yeast 2023; 40:303-317. [PMID: 37190878 DOI: 10.1002/yea.3855] [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: 10/31/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
The oral cavity of humans is colonized by diversity of microbial community, although dominated by bacteria, it is also constituted by a low number of fungi, often represented by Candida albicans. Although in the vast minority, this usually commensal fungus under certain conditions of the host (e.g., immunosuppression or antibiotic therapy), can transform into an invasive pathogen that adheres to mucous membranes and also to medical or dental devices, causing mucosal infections. This transformation is correlated with changes in cell morphology from yeast-like cells to hyphae and is supported by numerous virulence factors exposed by C. albicans cells at the site of infection, such as multifunctional adhesins, degradative enzymes, or toxin. All of them affect the surrounding host cells or proteins, leading to their destruction. However, at the site of infection, C. albicans can interact with different bacterial species and in its filamentous form may produce biofilms-the elaborated consortia of microorganisms, that present increased ability to host colonization and resistance to antimicrobial agents. In this review, we highlight the modification of the infectious potential of C. albicans in contact with different bacterial species, and also consider the mutual bacterial-fungal relationships, involving cooperation, competition, or antagonism, that lead to an increase in the propagation of oral infection. The mycofilm of C. albicans is an excellent hiding place for bacteria, especially those that prefer low oxygen availability, where microbial cells during mutual co-existence can avoid host recognition or elimination by antimicrobial action. However, these microbial relationships, identified mainly in in vitro studies, are modified depending on the complexity of host conditions and microbial dominance in vivo.
Collapse
Affiliation(s)
- Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Magdalena Surowiec
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Aneta Bednarek
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Miriam Gonzalez-Gonzalez
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satała
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
29
|
Ormsby MJ, Akinbobola A, Quilliam RS. Plastic pollution and fungal, protozoan, and helminth pathogens - A neglected environmental and public health issue? THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 882:163093. [PMID: 36996975 DOI: 10.1016/j.scitotenv.2023.163093] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/10/2023] [Accepted: 03/23/2023] [Indexed: 06/01/2023]
Abstract
Plastic waste is ubiquitous in the environment and can become colonised by distinct microbial biofilm communities, known collectively as the 'plastisphere.' The plastisphere can facilitate the increased survival and dissemination of human pathogenic prokaryotes (e.g., bacteria); however, our understanding of the potential for plastics to harbour and disseminate eukaryotic pathogens is lacking. Eukaryotic microorganisms are abundant in natural environments and represent some of the most important disease-causing agents, collectively responsible for tens of millions of infections, and millions of deaths worldwide. While prokaryotic plastisphere communities in terrestrial, freshwater, and marine environments are relatively well characterised, such biofilms will also contain eukaryotic species. Here, we critically review the potential for fungal, protozoan, and helminth pathogens to associate with the plastisphere, and consider the regulation and mechanisms of this interaction. As the volume of plastics in the environment continues to rise there is an urgent need to understand the role of the plastisphere for the survival, virulence, dissemination, and transfer of eukaryotic pathogens, and the effect this can have on environmental and human health.
Collapse
Affiliation(s)
- Michael J Ormsby
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK.
| | - Ayorinde Akinbobola
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Richard S Quilliam
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| |
Collapse
|
30
|
Li F, Gao Y, Cheng W, Su X, Yang R. Gut fungal mycobiome: A significant factor of tumor occurrence and development. Cancer Lett 2023; 569:216302. [PMID: 37451425 DOI: 10.1016/j.canlet.2023.216302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
A variety of bacteria, viruses, fungi, protists, archaea and protozoa coexists within the mammalian gastrointestinal (GI) tract such as that fungi are detectable in all intestinal and colon segments in almost all healthy adults. Although fungi can cause infectious diseases, they are also related to gut and systemic homeostasis. Importantly, through transformation of different forms such as from yeast to hyphae, interaction among gut microbiota such as fungal and bacterial interaction, host factors such as immune and host derived factors, and fungus genetic and epigenetic factors, fungi can be transformed from commensal into pathogenic lifestyles. Recent studies have shown that fungi play a significant role in the occurrence and development of tumors such as colorectal cancer. Indeed, evidences have shown that multiple species of different fungi exist in different tumors. Studies have also demonstrated that fungi are related to the occurrence and development of tumors, and also survival of patients. Here we summarize recent advances in the transformation of fungi from commensal into pathogenic lifestyles, and the effects of gut pathogenic fungi on the occurrence and development of tumors such as colorectal and pancreatic cancers.
Collapse
Affiliation(s)
- Fan Li
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yunhuan Gao
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Wenyue Cheng
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xiaomin Su
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Rongcun Yang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
31
|
Corrêa-Junior D, Bastos de Andrade I, Alves V, Avellar-Moura I, Brito de Souza Rabello V, Valdez AF, Nimrichter L, Zancopé-Oliveira RM, Ribeiro de Sousa Araújo G, Almeida-Paes R, Frases S. Unveiling the Morphostructural Plasticity of Zoonotic Sporotrichosis Fungal Strains: Possible Implications for Sporothrix brasiliensis Virulence and Pathogenicity. J Fungi (Basel) 2023; 9:701. [PMID: 37504690 PMCID: PMC10381685 DOI: 10.3390/jof9070701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Sporotrichosis is a fungal infection caused by Sporothrix species, with Sporothrix brasiliensis as a prevalent pathogen in Latin America. Despite its clinical importance, the virulence factors of S. brasiliensis and their impact on the pathogenesis of sporotrichosis are still poorly understood. This study evaluated the morphostructural plasticity of S. brasiliensis, a fungus that causes sporotrichosis. Three cell surface characteristics, namely cell surface hydrophobicity, Zeta potential, and conductance, were assessed. Biofilm formation was also analyzed, with measurements taken for biomass, extracellular matrix, and metabolic activity. In addition, other potential and poorly studied characteristics correlated with virulence such as lipid bodies, chitin, and cell size were evaluated. The results revealed that the major phenotsypic features associated with fungal virulence in the studied S. brasiliensis strains were chitin, lipid bodies, and conductance. The dendrogram clustered the strains based on their overall similarity in the production of these factors. Correlation analyses showed that hydrophobicity was strongly linked to the production of biomass and extracellular matrix, while there was a weaker association between Zeta potential and size, and lipid bodies and chitin. This study provides valuable insights into the virulence factors of S. brasiliensis and their potential role in the pathogenesis of sporotrichosis.
Collapse
Affiliation(s)
- Dario Corrêa-Junior
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Iara Bastos de Andrade
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Vinicius Alves
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Igor Avellar-Moura
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Vanessa Brito de Souza Rabello
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Alessandro Fernandes Valdez
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21040-900, Brazil
| | - Leonardo Nimrichter
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21040-900, Brazil
- Rede Micologia RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Rosely Maria Zancopé-Oliveira
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Glauber Ribeiro de Sousa Araújo
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Rodrigo Almeida-Paes
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
- Rede Micologia RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
- Rede Micologia RJ, FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Ennerfelt H, Holliday C, Shapiro D, Zengeler K, Bolte A, Ulland T, Lukens J. CARD9 attenuates Aβ pathology and modifies microglial responses in an Alzheimer's disease mouse model. Proc Natl Acad Sci U S A 2023; 120:e2303760120. [PMID: 37276426 PMCID: PMC10268238 DOI: 10.1073/pnas.2303760120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 06/07/2023] Open
Abstract
Recent advances have highlighted the importance of several innate immune receptors expressed by microglia in Alzheimer's disease (AD). In particular, mounting evidence from AD patients and experimental models indicates pivotal roles for TREM2, CD33, and CD22 in neurodegenerative disease progression. While there is growing interest in targeting these microglial receptors to treat AD, we still lack knowledge of the downstream signaling molecules used by these receptors to orchestrate immune responses in AD. Notably, TREM2, CD33, and CD22 have been described to influence signaling associated with the intracellular adaptor molecule CARD9 to mount downstream immune responses outside of the brain. However, the role of CARD9 in AD remains poorly understood. Here, we show that genetic ablation of CARD9 in the 5xFAD mouse model of AD results in exacerbated amyloid beta (Aβ) deposition, increased neuronal loss, worsened cognitive deficits, and alterations in microglial responses. We further show that pharmacological activation of CARD9 promotes improved clearance of Aβ deposits from the brains of 5xFAD mice. These results help to establish CARD9 as a key intracellular innate immune signaling molecule that regulates Aβ-mediated disease and microglial responses. Moreover, these findings suggest that targeting CARD9 might offer a strategy to improve Aβ clearance in AD.
Collapse
Affiliation(s)
- Hannah Ennerfelt
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA22908
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA22908
- Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA22908
| | - Coco Holliday
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA22908
| | - Daniel A. Shapiro
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA22908
| | - Kristine E. Zengeler
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA22908
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA22908
- Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA22908
| | - Ashley C. Bolte
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA22908
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA22908
| | - Tyler K. Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI53705
| | - John R. Lukens
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA22908
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA22908
- Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA22908
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA22908
| |
Collapse
|
33
|
Alvarado M, Gómez-Navajas JA, Blázquez-Muñoz MT, Gómez-Molero E, Berbegal C, Eraso E, Kramer G, De Groot PWJ. Integrated post-genomic cell wall analysis reveals floating biofilm formation associated with high expression of flocculins in the pathogen Pichia kudriavzevii. PLoS Pathog 2023; 19:e1011158. [PMID: 37196016 DOI: 10.1371/journal.ppat.1011158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/30/2023] [Accepted: 04/27/2023] [Indexed: 05/19/2023] Open
Abstract
The pathogenic yeast Pichia kudriavzevii, previously known as Candida krusei, is more distantly related to Candida albicans than clinically relevant CTG-clade Candida species. Its cell wall, a dynamic organelle that is the first point of interaction between pathogen and host, is relatively understudied, and its wall proteome remains unidentified to date. Here, we present an integrated study of the cell wall in P. kudriavzevii. Our comparative genomic studies and experimental data indicate that the general structure of the cell wall in P. kudriavzevii is similar to Saccharomyces cerevisiae and C. albicans and is comprised of β-1,3-glucan, β-1,6-glucan, chitin, and mannoproteins. However, some pronounced differences with C. albicans walls were observed, for instance, higher mannan and protein levels and altered protein mannosylation patterns. Further, despite absence of proteins with high sequence similarity to Candida adhesins, protein structure modeling identified eleven proteins related to flocculins/adhesins in S. cerevisiae or C. albicans. To obtain a proteomic comparison of biofilm and planktonic cells, P. kudriavzevii cells were grown to exponential phase and in static 24-h cultures. Interestingly, the 24-h static cultures of P. kudriavzevii yielded formation of floating biofilm (flor) rather than adherence to polystyrene at the bottom. The proteomic analysis of both conditions identified a total of 33 cell wall proteins. In line with a possible role in flor formation, increased abundance of flocculins, in particular Flo110, was observed in the floating biofilm compared to exponential cells. This study is the first to provide a detailed description of the cell wall in P. kudriavzevii including its cell wall proteome, and paves the way for further investigations on the importance of flor formation and flocculins in the pathogenesis of P. kudriavzevii.
Collapse
Affiliation(s)
- María Alvarado
- Regional Center for Biomedical Research, Castilla-La Mancha Science & Technology Park, University of Castilla-La Mancha, Albacete, Spain
| | - Jesús Alberto Gómez-Navajas
- Regional Center for Biomedical Research, Castilla-La Mancha Science & Technology Park, University of Castilla-La Mancha, Albacete, Spain
| | - María Teresa Blázquez-Muñoz
- Regional Center for Biomedical Research, Castilla-La Mancha Science & Technology Park, University of Castilla-La Mancha, Albacete, Spain
| | - Emilia Gómez-Molero
- Regional Center for Biomedical Research, Castilla-La Mancha Science & Technology Park, University of Castilla-La Mancha, Albacete, Spain
| | - Carmen Berbegal
- ENOLAB, Estructura de Recerca Interdisciplinar (ERI) BioTecMed and Departament de Microbiologia i Ecología, Universitat de València, Burjassot, Spain
| | - Elena Eraso
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Gertjan Kramer
- Mass Spectrometry of Biomolecules, University of Amsterdam, Swammerdam Institute for Life Sciences Amsterdam, Amsterdam, The Netherlands
| | - Piet W J De Groot
- Regional Center for Biomedical Research, Castilla-La Mancha Science & Technology Park, University of Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
34
|
Kulshrestha A, Gupta P. Secreted aspartyl proteases family: a perspective review on the regulation of fungal pathogenesis. Future Microbiol 2023; 18:295-309. [PMID: 37097060 DOI: 10.2217/fmb-2022-0143] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Secreted aspartyl proteases (SAPs) are important enzymes for fungal pathogenicity, playing a significant role in infection and survival. This article provides insight into how SAPs facilitate the transformation of yeast cells into hyphae and engage in biofilm formation, invasion and degradation of host cells and proteins. SAPs and their isoenzymes are prevalent during fungal infections, making them a potential target for antifungal and antibiofilm therapies. By targeting SAPs, critical stages of fungal pathogenesis such as adhesion, hyphal development, biofilm formation, host invasion and immune evasion can potentially be disrupted. Developing therapies that target SAPs could provide an effective treatment option for a wide range of fungal infections.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, India
| |
Collapse
|
35
|
Nouri N, Mohammadi SR, Beardsley J, Aslani P, Ghaffarifar F, Roudbary M, Rodrigues CF. Thymoquinone Antifungal Activity against Candida glabrata Oral Isolates from Patients in Intensive Care Units-An In Vitro Study. Metabolites 2023; 13:metabo13040580. [PMID: 37110238 PMCID: PMC10143056 DOI: 10.3390/metabo13040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The number of Candida spp. infections and drug resistance are dramatically increasing worldwide, particularly among immunosuppressed patients, and it is urgent to find novel compounds with antifungal activity. In this work, the antifungal and antibiofilm activity of thymoquinone (TQ), a key bioactive constituent of black cumin seed Nigella sativa L., was evaluated against Candida glabrata, a WHO 'high-priority' pathogen. Then, its effect on the expression of C. glabrata EPA6 and EPA7 genes (related to biofilm adhesion and development, respectively) were analyzed. Swab samples were taken from the oral cavity of 90 hospitalized patients in ICU wards, transferred to sterile falcon tubes, and cultured on Sabouraud Dextrose Agar (SDA) and Chromagar Candida for presumptive identification. Next, a 21-plex PCR was carried out for the confirmation of species level. C. glabrata isolates underwent antifungal drug susceptibility testing against fluconazole (FLZ), itraconazole (ITZ), amphotericin B (AMB), and TQ according to the CLSI microdilution method (M27, A3/S4). Biofilm formation was measured by an MTT assay. EPA6 and EPA7 gene expression was assessed by real-time PCR. From the 90 swab samples, 40 isolates were identified as C. glabrata with the 21-plex PCR. Most isolates were resistant to FLZ (n = 29, 72.5%), whereas 12.5% and 5% were ITZ and AMB resistant, respectively. The minimum inhibitory concentration (MIC50) of TQ against C. glabrata was 50 µg/mL. Importantly, TQ significantly inhibited the biofilm formation of C. glabrata isolates, and EPA6 gene expression was reduced significantly at MIC50 concentration of TQ. TQ seems to have some antifungal, antibiofilm (adhesion) effect on C. glabrata isolates, showing that this plant secondary metabolite is a promising agent to overcome Candida infections, especially oral candidiasis.
Collapse
Affiliation(s)
- Noura Nouri
- Department of Medical Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115111, Iran
| | - Shahla Roudbar Mohammadi
- Department of Medical Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115111, Iran
| | - Justin Beardsley
- Sydney Institute for Infectious Diseases, University of Sydney, Sydney, NSW 2145, Australia
- Westmead Hospital, NSW Health, Sydney, NSW 2145, Australia
| | - Peyman Aslani
- Department of Parasitology and Mycology, Faculty of Medicine, Aja University of Medical Sciences, Tehran 1411718541, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology and Entomology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115111, Iran
| | - Maryam Roudbary
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Célia Fortuna Rodrigues
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, 4585-116 Gandra PRD, Portugal
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
36
|
Smoak RA, Snyder LF, Fassler JS, He BZ. Parallel expansion and divergence of an adhesin family in pathogenic yeasts. Genetics 2023; 223:iyad024. [PMID: 36794645 PMCID: PMC10319987 DOI: 10.1093/genetics/iyad024] [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: 12/07/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Opportunistic yeast pathogens arose multiple times in the Saccharomycetes class, including the recently emerged, multidrug-resistant (MDR) Candida auris. We show that homologs of a known yeast adhesin family in Candida albicans, the Hyr/Iff-like (Hil) family, are enriched in distinct clades of Candida species as a result of multiple, independent expansions. Following gene duplication, the tandem repeat-rich region in these proteins diverged extremely rapidly and generated large variations in length and β-aggregation potential, both of which are known to directly affect adhesion. The conserved N-terminal effector domain was predicted to adopt a β-helical fold followed by an α-crystallin domain, making it structurally similar to a group of unrelated bacterial adhesins. Evolutionary analyses of the effector domain in C. auris revealed relaxed selective constraint combined with signatures of positive selection, suggesting functional diversification after gene duplication. Lastly, we found the Hil family genes to be enriched at chromosomal ends, which likely contributed to their expansion via ectopic recombination and break-induced replication. Combined, these results suggest that the expansion and diversification of adhesin families generate variation in adhesion and virulence within and between species and are a key step toward the emergence of fungal pathogens.
Collapse
Affiliation(s)
- Rachel A Smoak
- Civil and Environmental Engineering, The University of Iowa, Iowa City, IA 52242, USA
| | - Lindsey F Snyder
- Interdisciplinary Graduate Program in Genetics, The University of Iowa, Iowa City, IA 52242, USA
| | - Jan S Fassler
- Interdisciplinary Graduate Program in Genetics, The University of Iowa, Iowa City, IA 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA 52242, USA
| | - Bin Z He
- Interdisciplinary Graduate Program in Genetics, The University of Iowa, Iowa City, IA 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
37
|
Badrane H, Cheng S, Dupont CL, Hao B, Driscoll E, Morder K, Liu G, Newbrough A, Fleres G, Kaul D, Espinoza JL, Clancy CJ, Nguyen MH. Genotypic diversity and unrecognized antifungal resistance among populations of Candida glabrata from positive blood cultures. RESEARCH SQUARE 2023:rs.3.rs-2706400. [PMID: 37066226 PMCID: PMC10104189 DOI: 10.21203/rs.3.rs-2706400/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The longstanding paradigm is that most bloodstream infections (BSIs) are caused by a single organism. We performed whole genome sequencing of five-to-ten strains from blood culture (BC) bottles in each of ten patients with Candida glabrata BSI. We demonstrated that BCs contained mixed populations of clonal but genetically diverse strains. Genetically distinct strains from two patients exhibited phenotypes that were potentially important during BSIs, including differences in susceptibility to antifungal agents and phagocytosis. In both patients, the clinical microbiology lab recovered a fluconazole-susceptible index strain, but we identified mixed fluconazole-susceptible and â€"resistant populations. Diversity in drug susceptibility was likely clinically relevant, as fluconazole-resistant strains were subsequently recovered by the clinical laboratory during persistent or relapsing infections. In one patient, unrecognized respiration-deficient small colony variants were fluconazole-resistant and significantly attenuated for virulence during murine candidiasis. Our data suggest a new population-based paradigm of C. glabrata genotypic and phenotypic diversity during BSIs.
Collapse
Affiliation(s)
| | - Shaoji Cheng
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Binghua Hao
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Guojun Liu
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | - Cornelius J Clancy
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
38
|
Hsu C, Ghannoum M, Cominelli F, Martino LD. Mycobiome and Inflammatory Bowel Disease: Role in Disease Pathogenesis, Current Approaches and Novel Nutritional-based Therapies. Inflamm Bowel Dis 2023; 29:470-479. [PMID: 35851921 PMCID: PMC9977251 DOI: 10.1093/ibd/izac156] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Indexed: 12/09/2022]
Abstract
Inflammatory bowel disease (IBD), a disorder characterized by chronic inflammation of the gastrointestinal (GI) tract and a range of adverse health effects including diarrhea, abdominal pain, vomiting, and bloody stools, affects nearly 3.1 million genetically susceptible adults in the United States today. Although the etiology of IBD remains unclear, genetics, stress, diet, and gut microbiota dysbiosis- especially in immunocompromised individuals- have been identified as possible causes of disease. Although previous research has largely focused on the role of bacteria in IBD pathogenesis, recently observed alterations of fungal load and biodiversity in the GI tract of afflicted individuals suggest interkingdom interactions amongst different gut microbial communities, particularly between bacteria and fungi. These discoveries point to the potential utilization of treatment approaches such as antibiotics, antifungals, probiotics, and postbiotics that target both bacteria and fungi in managing IBD. In this review, we discuss the impact of specific fungi on disease pathogenesis, with a focus on the highly virulent genus Candida and how the presence of certain co-enzymes impacts its virulence. In addition, we evaluate current gut microbiome-based therapeutic approaches with the intention of better understanding the mechanisms behind novel therapies.
Collapse
Affiliation(s)
- Caitlyn Hsu
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
| | - Mahmoud Ghannoum
- Center for Medical Mycology and Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, and University Hospitals Cleveland Medical Center, Cleveland, Ohio, 44106, USA
| | - Fabio Cominelli
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
- Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
| | - Luca Di Martino
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
| |
Collapse
|
39
|
The Pga59 cell wall protein is an amyloid forming protein involved in adhesion and biofilm establishment in the pathogenic yeast Candida albicans. NPJ Biofilms Microbiomes 2023; 9:6. [PMID: 36697414 PMCID: PMC9877000 DOI: 10.1038/s41522-023-00371-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
The human commensal fungus Candida albicans can attach to epithelia or indwelling medical devices and form biofilms, that are highly tolerant to antifungal drugs and can evade the immune response. The cell surface protein Pga59 has been shown to influence adhesion and biofilm formation. Here, we present evidence that Pga59 displays amyloid properties. Using electron microscopy, staining with an amyloid fibre-specific dye and X-ray diffraction experiments, we showed that the predicted amyloid-forming region of Pga59 is sufficient to build up an amyloid fibre in vitro and that recombinant Pga59 can also adopt a cross-β amyloid fibre architecture. Further, mutations impairing Pga59 amyloid assembly led to diminished adhesion to substrates and reduced biofilm production. Immunogold labelling on amyloid structures extracted from C. albicans revealed that Pga59 is used by the fungal cell to assemble amyloids within the cell wall in response to adhesion. Altogether, our results suggest that Pga59 amyloid properties are used by the fungal cell to mediate cell-substrate interactions and biofilm formation.
Collapse
|
40
|
Branco J, Miranda IM, Rodrigues AG. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. J Fungi (Basel) 2023; 9:jof9010080. [PMID: 36675901 PMCID: PMC9862255 DOI: 10.3390/jof9010080] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Candida parapsilosis is the second most common Candida species isolated in Asia, Southern Europe, and Latin America and is often involved in invasive infections that seriously impact human health. This pathogen is part of the psilosis complex, which also includes Candida orthopsilosis and Candida metapsilosis. C. parapsilosis infections are particularly prevalent among neonates with low birth weights, individuals who are immunocompromised, and patients who require prolonged use of a central venous catheter or other indwelling devices, whose surfaces C. parapsilosis exhibits an enhanced capacity to adhere to and form biofilms. Despite this well-acknowledged prevalence, the biology of C. parapsilosis has not been as extensively explored as that of Candida albicans. In this paper, we describe the molecular mechanistic pathways of virulence in C. parapsilosis and show how they differ from those of C. albicans. We also describe the mode of action of antifungal drugs used for the treatment of Candida infections, namely, polyenes, echinocandins, and azoles, as well as the resistance mechanisms developed by C. parapsilosis to overcome them. Finally, we stress the importance of the ongoing search for species-specific features that may aid the development of effective control strategies and thus reduce the burden on patients and healthcare costs.
Collapse
Affiliation(s)
- Joana Branco
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Correspondence: ; Tel./Fax: +351-225513662
| | - Isabel M. Miranda
- Cardiovascular Research & Development Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Acácio G. Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
41
|
The Zinc Finger Transcription Factor Fts2 Represses the Yeast-to-Filament Transition in the Dimorphic Yeast Yarrowia lipolytica. mSphere 2022; 7:e0045022. [PMID: 36409080 PMCID: PMC9769893 DOI: 10.1128/msphere.00450-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The yeast-to-filament transition is an important cellular response to environmental stimulations in dimorphic fungi. In addition to activators, there are repressors in the cells to prevent filament formation, which is important to keep the cells in the yeast form when filamentation is not necessary. However, very few repressors of filamentation are known so far. Here, we identify a novel repressor of filamentation in the dimorphic yeast Yarrowia lipolytica, Fts2, which is a C2H2-type zinc finger transcription factor. We show that fts2Δ cells exhibited increased filamentation under mild filament-inducing conditions and formed filaments under non-filament-inducing conditions. We also show that Fts2 interacts with YlSsn6, component of the Tup1-Ssn6 transcriptional corepressor, and Fts2-LexA represses a lexAop-PYlACT1-lacZ reporter in a Tup1-Ssn6-dependent manner, suggesting that Fts2 has transcriptional repressor activity and represses gene expression via Tup1-Ssn6. In addition, we show that Fts2 represses a large number of cell wall protein genes and transcription factor genes, some of which are implicated in the filamentation response. Interestingly, about two-thirds of Fts2-repressed genes are also repressed by Tup1-Ssn6, suggesting that Fts2 may repress the bulk of its target genes via Tup1-Ssn6. Lastly, we show that Fts2 expression is downregulated in response to alkaline pH and the relief of negative control by Fts2 facilitates the induction of filamentation by alkaline pH. IMPORTANCE The repressors of filamentation are important negative regulators of the yeast-to-filament transition. However, except in Candida albicans, very few repressors of filamentation are known in dimorphic fungi. More importantly, how they repress filamentation is often not clear. In this paper, we report a novel repressor of filamentation in Y. lipolytica. Fts2 is not closely related in amino acid sequence to CaNrg1 and Rfg1, two major repressors of filamentation in C. albicans, yet it represses gene expression via the transcriptional corepressor Tup1-Ssn6, similar to CaNrg1 and Rfg1. Using transcriptome sequencing, we determined the whole set of genes regulated by Fts2 and identified the major targets of Fts2 repression, which provide clues to the mechanism by which Fts2 represses filamentation. Our results have important implications for understanding the negative control of the yeast-to-filament transition in dimorphic fungi.
Collapse
|
42
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
43
|
The Role of Sfp1 in Candida albicans Cell Wall Maintenance. J Fungi (Basel) 2022; 8:jof8111196. [PMID: 36422017 PMCID: PMC9692975 DOI: 10.3390/jof8111196] [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: 10/07/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
The cell wall is the first interface for Candida albicans interaction with the surrounding environment and the host cells. Therefore, maintenance of cell wall integrity (CWI) is crucial for C. albicans survival and host-pathogen interaction. In response to environmental stresses, C. albicans undergoes cell wall remodeling controlled by multiple signaling pathways and transcription regulators. Here, we explored the role of the transcription factor Sfp1 in CWI. A deletion of the SFP1 gene not only caused changes in cell wall properties, cell wall composition and structure but also modulated expression of cell wall biosynthesis and remodeling genes. In addition, Cas5 is a known transcription regulator for C. albicans CWI and cell wall stress response. Interestingly, our results indicated that Sfp1 negatively controls the CAS5 gene expression by binding to its promoter element. Together, this study provides new insights into the regulation of C. albicans CWI and stress response.
Collapse
|
44
|
Marcet-Houben M, Alvarado M, Ksiezopolska E, Saus E, de Groot PWJ, Gabaldón T. Chromosome-level assemblies from diverse clades reveal limited structural and gene content variation in the genome of Candida glabrata. BMC Biol 2022; 20:226. [PMID: 36209154 PMCID: PMC9548116 DOI: 10.1186/s12915-022-01412-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Background Candida glabrata is an opportunistic yeast pathogen thought to have a large genetic and phenotypic diversity and a highly plastic genome. However, the lack of chromosome-level genome assemblies representing this diversity limits our ability to accurately establish how chromosomal structure and gene content vary across strains. Results Here, we expanded publicly available assemblies by using long-read sequencing technologies in twelve diverse strains, obtaining a final set of twenty-one chromosome-level genomes spanning the known C. glabrata diversity. Using comparative approaches, we inferred variation in chromosome structure and determined the pan-genome, including an analysis of the adhesin gene repertoire. Our analysis uncovered four new adhesin orthogroups and inferred a rich ancestral adhesion repertoire, which was subsequently shaped through a still ongoing process of gene loss, gene duplication, and gene conversion. Conclusions C. glabrata has a largely stable pan-genome except for a highly variable subset of genes encoding cell wall-associated functions. Adhesin repertoire was established for each strain and showed variability among clades. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01412-1.
Collapse
Affiliation(s)
- Marina Marcet-Houben
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
| | - María Alvarado
- Regional Center for Biomedical Research, University of Castilla-La Mancha, E-02008, Albacete, Spain
| | - Ewa Ksiezopolska
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
| | - Ester Saus
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
| | - Piet W J de Groot
- Regional Center for Biomedical Research, University of Castilla-La Mancha, E-02008, Albacete, Spain.,Castilla-La Mancha Science & Technology Park, E-02006, Albacete, Spain
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain. .,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain. .,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain. .,Centro Investigación Biomédica En Red de Enfermedades Infecciosas, Barcelona, Spain.
| |
Collapse
|
45
|
Collaboration between Antagonistic Cell Type Regulators Governs Natural Variation in the Candida albicans Biofilm and Hyphal Gene Expression Network. mBio 2022; 13:e0193722. [PMID: 35993746 PMCID: PMC9600859 DOI: 10.1128/mbio.01937-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is among the most significant human fungal pathogens. However, the vast majority of C. albicans studies have focused on a single clinical isolate and its marked derivatives. We investigated natural variation among clinical C. albicans isolates in gene regulatory control of biofilm formation, a process crucial to virulence. The transcription factor Efg1 is required for biofilm-associated gene expression and biofilm formation. Previously, we found extensive variation in Efg1-responsive gene expression among 5 diverse clinical isolates. However, chromatin immunoprecipitation sequencing analysis showed that Efg1 binding to genomic loci was uniform among the isolates. Functional dissection of strain differences identified three transcription factors, Brg1, Tec1, and Wor1, for which small changes in expression levels reshaped the Efg1 regulatory network. Brg1 and Tec1 are known biofilm activators, and their role in Efg1 network variation may be expected. However, Wor1 is a known repressor of EFG1 expression and an inhibitor of biofilm formation. In contrast, we found that a modest increase in WOR1 RNA levels, reflecting the expression differences between C. albicans strains, could augment biofilm formation and expression of biofilm-related genes. The analysis of natural variation here reveals a novel function for a well-characterized gene and illustrates that strain diversity offers a unique resource for elucidation of network interactions.
Collapse
|
46
|
Yang Y, Huang J, Dornbusch D, Grundmeier G, Fahmy K, Keller A, Cheung DL. Effect of Surface Hydrophobicity on the Adsorption of a Pilus-Derived Adhesin-like Peptide. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:9257-9265. [PMID: 35876027 PMCID: PMC9352356 DOI: 10.1021/acs.langmuir.2c01016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/12/2022] [Indexed: 06/15/2023]
Abstract
Bacterial colonization of abiotic surfaces such as those of medical implants, membrane filters, and everyday household items is a process of tremendous importance for public health. Bacteria use adhesive cell surface structures called adhesins to establish contact with abiotic surfaces. Among them, protein filaments called type IV pili are particularly important and found in many Gram-negative pathogens such as Pseudomonas aeruginosa. Understanding the interaction of such adhesin proteins with different abiotic surfaces at the molecular level thus represents a fundamental prerequisite for impeding bacterial colonization and preventing the spread of infectious diseases. In this work, we investigate the interaction of a synthetic adhesin-like peptide, PAK128-144ox, derived from the type IV pilus of P. aeruginosa with hydrophilic and hydrophobic self-assembled monolayers (SAMs). Using a combination of molecular dynamics (MD) simulations, quartz crystal microbalance with dissipation monitoring (QCM-D), and spectroscopic investigations, we find that PAK128-144ox has a higher affinity for hydrophobic than for hydrophilic surfaces. Additionally, PAK128-144ox adsorption on the hydrophobic SAM is furthermore accompanied by a strong increase in α-helix content. Our results show a clear influence of surface hydrophobicity and further indicate that PAK128-144ox adsorption on the hydrophobic surface is enthalpically favored, while on the hydrophilic surface, entropic contributions are more significant. However, our spectroscopic investigations also suggest aggregation of the peptide under the employed experimental conditions, which is not considered in the MD simulations and should be addressed in more detail in future studies.
Collapse
Affiliation(s)
- Yu Yang
- Technical
and Macromolecular Chemistry, Paderborn
University, Warburger Str. 100, 33098 Paderborn, Germany
| | - Jingyuan Huang
- Technical
and Macromolecular Chemistry, Paderborn
University, Warburger Str. 100, 33098 Paderborn, Germany
| | - Daniel Dornbusch
- Institute
of Resource Ecology, Biophysics Department, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany
- Center
for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01062 Dresden, Germany
| | - Guido Grundmeier
- Technical
and Macromolecular Chemistry, Paderborn
University, Warburger Str. 100, 33098 Paderborn, Germany
| | - Karim Fahmy
- Institute
of Resource Ecology, Biophysics Department, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany
- Center
for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01062 Dresden, Germany
| | - Adrian Keller
- Technical
and Macromolecular Chemistry, Paderborn
University, Warburger Str. 100, 33098 Paderborn, Germany
| | - David L. Cheung
- School
of Chemistry, National University of Ireland
Galway, Galway H91 TK33, Ireland
| |
Collapse
|
47
|
Mendoza-Reyes DF, Gómez-Gaviria M, Mora-Montes HM. Candida lusitaniae: Biology, Pathogenicity, Virulence Factors, Diagnosis, and Treatment. Infect Drug Resist 2022; 15:5121-5135. [PMID: 36068831 PMCID: PMC9441179 DOI: 10.2147/idr.s383785] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/25/2022] [Indexed: 12/30/2022] Open
Abstract
The incidence of fungal infections is increasing at an alarming rate and has posed a great challenge for science in recent years. The rise in these infections has been related to the increase in immunocompromised patients and the resistance of different species to antifungal drugs. Infections caused by the different Candida species, especially Candida albicans, are one of the most common mycoses in humans, and the etiological agents are considered opportunistic pathogens associated with high mortality rates when disseminated infections occur. Candida lusitaniae is considered an emerging opportunistic pathogen that most frequently affects immunocompromised patients with some comorbidity. Although it is a low-frequency pathogen, and the mortality rate of C. lusitaniae-caused candidemia does not exceed 5%, some isolates are known to be resistant to antifungals such as amphotericin B, 5-fluorocytosine, and fluconazole. In this paper, a detailed review of the current literature on this organism and its different aspects, such as its biology, possible virulence factors, pathogen-host interaction, diagnosis, and treatment of infection, is provided. Of particular interest, through Blastp analysis we predicted possible virulence factors in this species.
Collapse
Affiliation(s)
- Diana F Mendoza-Reyes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Guanajuato, Gto, C.P. 36050, México
| | - Manuela Gómez-Gaviria
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Guanajuato, Gto, C.P. 36050, México
- Correspondence: Manuela Gómez-Gaviria; Héctor M Mora-Montes, Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, Guanajuato, Gto, C.P. 36050, México, Tel +52 473-7320006 Ext. 8193, Fax +52 473-7320006 Ext. 8153, Email ;
| | - Héctor M Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Guanajuato, Gto, C.P. 36050, México
| |
Collapse
|
48
|
Le Bars P, Kouadio AA, Bandiaky ON, Le Guéhennec L, de La Cochetière MF. Host's Immunity and Candida Species Associated with Denture Stomatitis: A Narrative Review. Microorganisms 2022; 10:microorganisms10071437. [PMID: 35889156 PMCID: PMC9323190 DOI: 10.3390/microorganisms10071437] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
Denture-related Candida stomatitis, which has been described clinically in the literature, is either localized or generalized inflammation of the oral mucosa in connection with a removable prosthesis. During this inflammatory process, the mycobacterial biofilm and the host’s immune response play an essential role. Among microorganisms of this mixed biofilm, the Candida species proliferates easily and changes from a commensal to an opportunistic pathogen. In this situation, the relationship between the Candida spp. and the host is influenced by the presence of the denture and conditioned both by the immune response and the oral microbiota. Specifically, this fungus is able to hijack the innate immune system of its host to cause infection. Additionally, older edentulous wearers of dentures may experience an imbalanced and decreased oral microbiome diversity. Under these conditions, the immune deficiency of these aging patients often promotes the spread of commensals and pathogens. The present narrative review aimed to analyze the innate and adaptive immune responses of patients with denture stomatitis and more particularly the involvement of Candida albicans sp. associated with this pathology.
Collapse
Affiliation(s)
- Pierre Le Bars
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
- Correspondence: authors:
| | - Alain Ayepa Kouadio
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
- Department of Prosthetic Dentistry, Faculty of Dentistry, CHU, Abidjan P.O. Box 612, Côte d’Ivoire
| | - Octave Nadile Bandiaky
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
| | - Laurent Le Guéhennec
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
| | - Marie-France de La Cochetière
- EA 3826 Thérapeutiques Cliniques Et expérimentales des Infections, Faculté de Médecine, CHU Hôtel-Dieu, Université de Nantes, 1, rue G. Veil, 44000 Nantes, France;
| |
Collapse
|
49
|
Tec1 and Ste12 transcription factors play a role in adaptation to low pH stress and biofilm formation in the human opportunistic fungal pathogen Candida glabrata. INTERNATIONAL MICROBIOLOGY : THE OFFICIAL JOURNAL OF THE SPANISH SOCIETY FOR MICROBIOLOGY 2022; 25:789-802. [PMID: 35829973 DOI: 10.1007/s10123-022-00264-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 01/18/2023]
Abstract
Eukaryotic cells respond to environmental cues through mitogen activated protein kinase (MAPK) signaling pathways. Each MAPK cascade is specific to particular stimuli and mediates specialized responses through activation of transcription factors. In the budding yeast, Saccharomyces cerevisiae, the pheromone-induced mating pathway and the starvation-responsive invasive growth/filamentation pathway generate their distinct outputs through the transcription factors Ste12 and Tec1, respectively. In this study, we report the functional characterization of these transcription factors in the closely related human opportunistic pathogenic yeast Candida glabrata. Two homologues each for S. cerevisiae TEC1 and STE12 were identified in C. glabrata. Both C. glabrata Tec1 proteins contain the N-terminal TEA DNA-binding domain characteristic of the TEA/ATTS transcription factor family. Similarly, the DNA-binding homeodomain shared by members of the highly conserved fungal Ste12 transcription factor family is present in N-terminus of both C. glabrata Ste12 transcription factors. We show that both C. glabrata STE12 genes are at least partial functional orthologues of S. cerevisiae STE12 as they can rescue the mating defect of haploid S. cerevisiae ste12 null mutant. Knockout of one of the STE12 genes (ORF CAGL0H02145g) leads to decreased biofilm development; a stronger biofilm-impaired phenotype results from loss of both CgSTE12 genes in the double deletion mutant (Cgste12ΔΔ). The transcript levels of one of the TEC1 genes (ORF CAGL0M01716g) were found to be upregulated upon exposure to low pH; its deletion causes slightly increased sensitivity to higher concentrations of acetic acid. Heat shock leads to increase in mRNA levels of one of the STE12 genes (ORF CAGL0M01254g). These findings suggest a role of Tec1 and Ste12 transcription factors in the regulation of some traits (biofilm formation, response to low pH stress and elevated temperature) that contribute to C. glabrata's ability to colonize various host niches and to occasionally cause disease.
Collapse
|
50
|
Gabius H, Cudic M, Diercks T, Kaltner H, Kopitz J, Mayo KH, Murphy PV, Oscarson S, Roy R, Schedlbauer A, Toegel S, Romero A. What is the Sugar Code? Chembiochem 2022; 23:e202100327. [PMID: 34496130 PMCID: PMC8901795 DOI: 10.1002/cbic.202100327] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/07/2021] [Indexed: 12/18/2022]
Abstract
A code is defined by the nature of the symbols, which are used to generate information-storing combinations (e. g. oligo- and polymers). Like nucleic acids and proteins, oligo- and polysaccharides are ubiquitous, and they are a biochemical platform for establishing molecular messages. Of note, the letters of the sugar code system (third alphabet of life) excel in coding capacity by making an unsurpassed versatility for isomer (code word) formation possible by variability in anomery and linkage position of the glycosidic bond, ring size and branching. The enzymatic machinery for glycan biosynthesis (writers) realizes this enormous potential for building a large vocabulary. It includes possibilities for dynamic editing/erasing as known from nucleic acids and proteins. Matching the glycome diversity, a large panel of sugar receptors (lectins) has developed based on more than a dozen folds. Lectins 'read' the glycan-encoded information. Hydrogen/coordination bonding and ionic pairing together with stacking and C-H/π-interactions as well as modes of spatial glycan presentation underlie the selectivity and specificity of glycan-lectin recognition. Modular design of lectins together with glycan display and the nature of the cognate glycoconjugate account for the large number of post-binding events. They give an entry to the glycan vocabulary its functional, often context-dependent meaning(s), hereby building the dictionary of the sugar code.
Collapse
Affiliation(s)
- Hans‐Joachim Gabius
- Institute of Physiological ChemistryFaculty of Veterinary MedicineLudwig-Maximilians-University MunichVeterinärstr. 1380539MunichGermany
| | - Maré Cudic
- Department of Chemistry and BiochemistryCharles E. Schmidt College of ScienceFlorida Atlantic University777 Glades RoadBoca RatonFlorida33431USA
| | - Tammo Diercks
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)Bizkaia Technology Park, Building 801 A48160DerioBizkaiaSpain
| | - Herbert Kaltner
- Institute of Physiological ChemistryFaculty of Veterinary MedicineLudwig-Maximilians-University MunichVeterinärstr. 1380539MunichGermany
| | - Jürgen Kopitz
- Institute of PathologyDepartment of Applied Tumor BiologyFaculty of MedicineRuprecht-Karls-University HeidelbergIm Neuenheimer Feld 22469120HeidelbergGermany
| | - Kevin H. Mayo
- Department of BiochemistryMolecular Biology & BiophysicsUniversity of MinnesotaMinneapolisMN 55455USA
| | - Paul V. Murphy
- CÚRAM – SFI Research Centre for Medical Devices and theSchool of ChemistryNational University of Ireland GalwayUniversity RoadGalwayH91 TK33Ireland
| | - Stefan Oscarson
- Centre for Synthesis and Chemical BiologyUniversity College DublinBelfieldDublin 4Ireland
| | - René Roy
- Département de Chimie et BiochimieUniversité du Québec à MontréalCase Postale 888Succ. Centre-Ville MontréalQuébecH3C 3P8Canada
| | - Andreas Schedlbauer
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)Bizkaia Technology Park, Building 801 A48160DerioBizkaiaSpain
| | - Stefan Toegel
- Karl Chiari Lab for Orthopaedic BiologyDepartment of Orthopedics and Trauma SurgeryMedical University of ViennaViennaAustria
| | - Antonio Romero
- Department of Structural and Chemical BiologyCIB Margarita Salas, CSICRamiro de Maeztu 928040MadridSpain
| |
Collapse
|