1
|
Zarnowski R, Horton MV, Johnson CJ, Vang PC, Uram J, Fernando LDP, Vlach J, Heiss C, Azadi P, Nett JE, Andes DR. Dual function of Candida auris mannosyltransferase, MNT5, in biofilm community protection from antifungal therapy and the host. mBio 2025; 16:e0034625. [PMID: 39998206 PMCID: PMC11980391 DOI: 10.1128/mbio.00346-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Screen of mutants from a mannosyltransferase family identified the importance of MNT5 for C. auris biofilm drug resistance and neutrophil evasion. Biochemical analysis of the mnt5∆ mutant matrix and cell wall identified alterations in the mannan structures. Resistance and matrix for mnt5∆ were restored with delivery of wild-type matrix via extracellular vesicles. Analysis of the mnt5∆ cell wall revealed a reduction in mannan and compensatory increase in cell surface glucan and chitin, suggesting a role for MNT5 in mannan masking of pathogen-associated molecular patterns. IMPORTANCE C. auris recalcitrance is linked to biofilm drug resistance and immune evasion. The mannosyltransferase encoded by MNT5 is necessary for both phenotypes and may serve as a useful therapeutic target.
Collapse
Affiliation(s)
- Robert Zarnowski
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark V. Horton
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chad J. Johnson
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Por Choua Vang
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jeremy Uram
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Jiri Vlach
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Christian Heiss
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Jeniel E. Nett
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David R. Andes
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Katsiari M, Nikolaou C, Palla E, Theodoridou K, Tsakris A, Vrioni G. Impact of Candida auris on Critically Ill Patients: A Three-Year Observational Study in a Greek Intensive Care Unit. Pathogens 2025; 14:328. [PMID: 40333088 PMCID: PMC12030536 DOI: 10.3390/pathogens14040328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Candida auris has emerged as a multidrug-resistant yeast implicated in healthcare-associated invasive infections and hospital outbreaks. The aim of the current 38-month period observational study in a multidisciplinary Intensive Care Unit (ICU) was to analyze the epidemiology, potential risk factors, management strategies, and patient outcomes of patients with C. auris. During the study period, 32 patients were identified with C. auris infection (6 patients) or colonization (26 patients) and their clinical characteristics and treatment-related factors were compared. Identification of C. auris isolates was confirmed by MALDI-TOF spectrometry. According to our results, regarding patient-related factors, no significant differences were identified. Regarding treatment-related factors, the proportion of patients already receiving corticosteroids (34.6% vs. 83.3%, p = 0.064) or being on renal replacement treatment (7.7% vs. 33.3%) was higher in infected patients. Median time elapsed from ICU admission to first positive culture was 7 (1-21) days and half of cases were ICU-imported. All strains were resistant to fluconazole and susceptible to echinocandines and amphotericin B. Crude mortality of the study population was 43.75%, similar to other previously reported candidemias. Rapid identification of C. auris, continued surveillance, and infection control practices are important elements for controlling successfully its spread in the hospital setting and for establishing promptly its transition from commensalism to infection.
Collapse
Affiliation(s)
- Maria Katsiari
- Intensive Care Unit, Konstantopouleio-Patision General Hospital, 3-5 Theodorou Konstantopoulou Street, N. Ionia, 14233 Athens, Greece;
| | - Charikleia Nikolaou
- Intensive Care Unit, Konstantopouleio-Patision General Hospital, 3-5 Theodorou Konstantopoulou Street, N. Ionia, 14233 Athens, Greece;
| | - Eleftheria Palla
- Department of Microbiology, Konstantopouleio- Patision General Hospital, 3-5 Theodorou Konstantopoulou Street, N. Ionia, 14233 Athens, Greece;
| | - Kalliopi Theodoridou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (K.T.); (A.T.)
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (K.T.); (A.T.)
| | - Georgia Vrioni
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (K.T.); (A.T.)
| |
Collapse
|
3
|
Tamai R, Kiyoura Y. Candida Infections: The Role of Saliva in Oral Health-A Narrative Review. Microorganisms 2025; 13:717. [PMID: 40284554 PMCID: PMC12029948 DOI: 10.3390/microorganisms13040717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Candida species, particularly Candida albicans, are causative agents of oral infections to which immunocompromised patients are especially susceptible. Reduced saliva flow (xerostomia) can lead to Candida overgrowth, as saliva contains antibacterial components such as histatins and β-defensins that inhibit fungal growth and adhesion to the oral mucosa. Candida adheres to host tissues, forms biofilms, and secretes enzymes required for tissue invasion and immune evasion. Secretory asparaginyl proteinases (Saps) and candidalysin, a cytolytic peptide toxin, are vital to Candida virulence, and agglutinin-like sequence (Als) proteins are crucial for adhesion, invasion, and biofilm formation. C. albicans is a risk factor for dental caries and may increase periodontal disease virulence when it coexists with Porphyromonas gingivalis. Candida infections have been suggested to heighten the risk of oral cancer based on a relationship between Candida species and oral squamous cell carcinoma (OSCC) or oral potentially malignant disorder (OPMD). Meanwhile, β-glucan in the Candida cell wall has antitumor effects. In addition, Candida biofilms protect viruses such as herpesviruses and coxsackieviruses. Understanding the intricate interactions between Candida species, host immune responses, and coexisting microbial communities is essential for developing preventive and therapeutic strategies against oral Candida infections, particularly in immunocompromised individuals.
Collapse
Affiliation(s)
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| |
Collapse
|
4
|
Bhargava A, Klamer K, Sharma M, Ortiz D, Saravolatz L. Candida auris: A Continuing Threat. Microorganisms 2025; 13:652. [PMID: 40142543 PMCID: PMC11946832 DOI: 10.3390/microorganisms13030652] [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: 02/01/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Candida auris is a World Health Organization critical-priority fungal pathogen that has variable resistance to antifungal treatments. Multiple clades have been identified through genomic analysis and have appeared in different geographic locations simultaneously. Due to a combination of factors including antifungal resistance, ability to colonize and persist in the environment, and thermotolerance, it can thrive. Infected patients are associated with a high mortality rate, especially those with multiple health risk factors like those associated with other Candida species. This review highlights the current situation of this pathogen to help provide guidance for future work.
Collapse
Affiliation(s)
- Ashish Bhargava
- Thomas Mackey Center of Infectious Diseases, Henry Ford Health—St. John Hospital, Detroit, MI 48236, USA
- School of Medicine, Wayne State University, Detroit, MI 48202, USA
| | - Katherine Klamer
- Thomas Mackey Center of Infectious Diseases, Henry Ford Health—St. John Hospital, Detroit, MI 48236, USA
| | - Mamta Sharma
- Thomas Mackey Center of Infectious Diseases, Henry Ford Health—St. John Hospital, Detroit, MI 48236, USA
| | - Daniel Ortiz
- LabCorp—Health Systems Operating Division, Troy, MI 48083, USA
| | - Louis Saravolatz
- Thomas Mackey Center of Infectious Diseases, Henry Ford Health—St. John Hospital, Detroit, MI 48236, USA
- School of Medicine, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
5
|
Barbian HJ, Lie L, Kittner A, Harrington A, Carson J, Frias M, Slade DH, Kim DY, Black S, Parada JP, Hayden MK. Candida auris Outbreak and Epidemiologic Response in Burn Intensive Care Unit, Illinois, USA, 2021-2023. Emerg Infect Dis 2025; 31:438-447. [PMID: 40023787 PMCID: PMC11878305 DOI: 10.3201/eid3103.241195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025] Open
Abstract
Candida auris is an emerging fungal pathogen associated with outbreaks in healthcare settings. We report a multiyear outbreak of C. auris in a burn intensive care unit in Illinois, USA, during 2021-2023. We identified 28 C. auris cases in the unit over a 2-year period, despite outbreak response and multimodal mitigation measures. Of the 28 case-patients, 15 (53.6%) were considered colonized and 13 (46.4%) had clinical infections. Phylogenetic analysis of whole-genome sequences revealed 4 distinct clusters of closely related (0-6 SNP differences) genomes containing 3-6 cases. Clusters generally contained temporally related isolates from patients with epidemiologic links; this finding suggests that multiple introductions and within-unit spread over a limited time were responsible for the outbreak, rather than transmission from a long-term source (e.g., persistent environmental contamination or staff carriage). Here, integrated traditional and genomic epidemiology supported C. auris outbreak investigation and response and informed targeted interventions.
Collapse
|
6
|
Biswas B, Asif S, Puria R, Thakur A. A Novel and Robust Method for Investigating Fungal Biofilm. Bio Protoc 2025; 15:e5146. [PMID: 39803325 PMCID: PMC11717715 DOI: 10.21769/bioprotoc.5146] [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: 07/10/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 01/16/2025] Open
Abstract
Candida auris, labeled an urgent threat by the CDC, shows significant resilience to treatments and disinfectants via biofilm formation, complicating treatment/disease management. The inconsistencies in biofilm architecture observed across studies hinder the understanding of its role in pathogenesis. Our novel in vitro technique cultivates C. auris biofilms on gelatin-coated coverslips, reliably producing multilayer biofilms with extracellular polymeric substances (EPS). This method, applicable to other Candida species like C. glabrata and C. albicans, is cost-effective and mimics the niche of biofilm formation. It is suitable for high-throughput drug screening and repurposing efforts, aiding in the development of new therapeutics. Our technique represents a significant advancement in Candida biofilm research, addressing the need for consistent, reproducible biofilm models. We detail a step-by-step procedure for creating a substratum for biofilm growth and measuring biofilm thickness using confocal laser scanning microscopy (CLSM) and ultrastructure by scanning electron microscopy (SEM). This method provides consistent outcomes across various Candida species. Key features • The biofilm formed on gelatin surfaces mimics host conditions, replicating the multilayered structure and EPS, offering a more accurate model for studying C. auris biofilms. • This method is highly reproducible and suitable for drug screening and biofilm analysis through three-dimensional (3D) reconstruction. • This in vitro technique aids in studying biofilm formation, related virulence properties, and drug tolerance of C. auris and other Candida species. • The simple, cost-effective technique is ideal for screening novel inhibitors and repurposed drug libraries, facilitating the design/identification of new therapeutics against Candida species.
Collapse
Affiliation(s)
- Biswambhar Biswas
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, India
| | - Shumaiza Asif
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, India
| | - Rekha Puria
- Department of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, India
| | - Anil Thakur
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
7
|
Honorato L, Artunduaga Bonilla JJ, Ribeiro da Silva L, Kornetz J, Zamith-Miranda D, Valdez AF, Nosanchuk JD, Gonçalves Paterson Fox E, Nimrichter L. Alkaloids solenopsins from fire ants display in vitro and in vivo activity against the yeast Candida auris. Virulence 2024; 15:2413329. [PMID: 39370781 PMCID: PMC11469440 DOI: 10.1080/21505594.2024.2413329] [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: 10/06/2023] [Revised: 09/01/2024] [Accepted: 09/07/2024] [Indexed: 10/08/2024] Open
Abstract
The urgency surrounding Candida auris as a public health threat is highlighted by both the Center for Disease Control (CDC) and World Health Organization (WHO) that categorized this species as a priority fungal pathogen. Given the current limitations of antifungal therapy for C. auris, particularly due to its multiple resistance to the current antifungals, the identification of new drugs is of paramount importance. Some alkaloids abundant in the venom of the red invasive fire ant (Solenopsis invicta), known as solenopsins, have garnered attention as potent inhibitors of bacterial biofilms, and there are no studies demonstrating such effects against fungal pathogens. Thus, we herein investigated the antibiotic efficacy of solenopsin alkaloids against C. auris biofilms and planktonic cells. Both natural and synthetic solenopsins inhibited the growth of C. auris strains from different clades, including fluconazole and amphotericin B-resistant isolates. Such alkaloids also inhibited matrix deposition and altered cellular metabolic activity of C. auris in biofilm conditions. Mechanistically, the alkaloids compromised membrane integrity as measured by propidium iodide uptake in exposed planktonic cells. Additionally, combining the alkaloids with AMB yielded an additive antifungal effect, even against AMB-resistant strains. Finally, both extracted solenopsins and the synthetic analogues demonstrated protective effect in vivo against C. auris infection in the invertebrate model Galleria mellonella. These findings underscore the potent antifungal activities of solenopsins against C. auris and suggest their inclusion in future drug development. Furthermore, exploring derivatives of solenopsins could reveal novel compounds with therapeutic promise.
Collapse
Affiliation(s)
- Leandro Honorato
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Rio de Janeiro, Brazil
| | - Jhon Jhamilton Artunduaga Bonilla
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Rio de Janeiro, Brazil
| | - Larissa Ribeiro da Silva
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Rio de Janeiro, Brazil
| | - Julio Kornetz
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Rio de Janeiro, Brazil
| | - Daniel Zamith-Miranda
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Alessandro F. Valdez
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Rio de Janeiro, Brazil
| | - Joshua D. Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Leonardo Nimrichter
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
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; 15:e0274824. [PMID: 39422509 PMCID: PMC11558994 DOI: 10.1128/mbio.02748-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
9
|
Louvet M, Li J, Brandalise D, Bachmann D, Sala de Oyanguren F, Labes D, Jacquier N, Genoud C, Mucciolo A, Coste AT, Sanglard D, Lamoth F. Ume6-dependent pathways of morphogenesis and biofilm formation in Candida auris. Microbiol Spectr 2024; 12:e0153124. [PMID: 39297645 PMCID: PMC11537075 DOI: 10.1128/spectrum.01531-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/05/2024] [Indexed: 11/07/2024] Open
Abstract
Candida auris is a yeast pathogen causing nosocomial outbreaks of candidemia. Its ability to adhere to inert surfaces and to be transmitted from one patient to another via medical devices is of particular concern. Like other Candida spp., C. auris has the ability to transition from the yeast form to pseudohyphae and to build biofilms. Moreover, some isolates have a unique capacity to form aggregates. These morphogenetic changes may impact virulence. In this study, we demonstrated the role of the transcription factor Ume6 in C. auris morphogenesis. Genetic hyperactivation of Ume6 induced filamentation and aggregation. The Ume6-hyperactivated strain (UME6HA) also exhibited increased adhesion to inert surface and formed biofilms of higher biomass compared to the parental strain. Transcriptomic analyses of UME6HA revealed enrichment of genes encoding for adhesins, proteins involved in cell wall organization, sterol biosynthesis, and aspartic protease activities. The three most upregulated genes compared to wild-type were those encoding for the agglutin-like sequence adhesin Als4498, the C. auris-specific adhesin Scf1, and the hypha-specific G1 cyclin-related protein Hgc1. The deletion of these genes in the UME6HA background showed that Ume6 controls filamentation via Hgc1 and aggregation via Als4498 and Scf1. Adhesion to inert surface was essentially triggered by Scf1. However, Als4498 and Hgc1 were also crucial for biofilm formation. Our data show that Ume6 is a universal regulator of C. auris morphogenesis via distinct modulators.IMPORTANCEC. auris represents a public health threat because of its ability to cause difficult-to-treat infections and hospital outbreaks. The morphogenetic plasticity of C. auris, including its ability to filament, to form aggregates or biofilms on inert surfaces, is important to the fungus for interhuman transmission, skin or catheter colonization, tissue invasion, antifungal resistance, and escape of the host immune system. This work deciphered the importance of Ume6 in the control of distinct pathways involved in filamentation, aggregation, adhesion, and biofilm formation of C. auris. A better understanding of the mechanisms of C. auris morphogenesis may help identify novel antifungal targets.
Collapse
Affiliation(s)
- Marine Louvet
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jizhou Li
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Danielle Brandalise
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel Bachmann
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Danny Labes
- Flow Cytometry Facility, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Jacquier
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christel Genoud
- Electron Microscopy Facility, University of Lausanne, Lausanne, Switzerland
| | - Antonio Mucciolo
- Electron Microscopy Facility, University of Lausanne, Lausanne, Switzerland
| | - Alix T. Coste
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dominique Sanglard
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Frederic Lamoth
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Weerasinghe H, Stölting H, Rose AJ, Traven A. Metabolic homeostasis in fungal infections from the perspective of pathogens, immune cells, and whole-body systems. Microbiol Mol Biol Rev 2024; 88:e0017122. [PMID: 39230301 PMCID: PMC11426019 DOI: 10.1128/mmbr.00171-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
SUMMARYThe ability to overcome metabolic stress is a major determinant of outcomes during infections. Pathogens face nutrient and oxygen deprivation in host niches and during their encounter with immune cells. Immune cells require metabolic adaptations for producing antimicrobial compounds and mounting antifungal inflammation. Infection also triggers systemic changes in organ metabolism and energy expenditure that range from an enhanced metabolism to produce energy for a robust immune response to reduced metabolism as infection progresses, which coincides with immune and organ dysfunction. Competition for energy and nutrients between hosts and pathogens means that successful survival and recovery from an infection require a balance between elimination of the pathogen by the immune systems (resistance), and doing so with minimal damage to host tissues and organs (tolerance). Here, we discuss our current knowledge of pathogen, immune cell and systemic metabolism in fungal infections, and the impact of metabolic disorders, such as obesity and diabetes. We put forward the idea that, while our knowledge of the use of metabolic regulation for fungal proliferation and antifungal immune responses (i.e., resistance) has been growing over the years, we also need to study the metabolic mechanisms that control tolerance of fungal pathogens. A comprehensive understanding of how to balance resistance and tolerance by metabolic interventions may provide insights into therapeutic strategies that could be used adjunctly with antifungal drugs to improve patient outcomes.
Collapse
Affiliation(s)
- Harshini Weerasinghe
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| | - Helen Stölting
- Department of Biochemistry and Molecular Biology and the Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Adam J Rose
- Department of Biochemistry and Molecular Biology and the Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
11
|
Martinez M, Garsin DA, Lorenz MC. Vertebrate and invertebrate animal infection models of Candida auris pathogenicity. Curr Opin Microbiol 2024; 80:102506. [PMID: 38925077 PMCID: PMC11432150 DOI: 10.1016/j.mib.2024.102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Candida auris is an emerging fungal pathogen with several concerning qualities. First recognized in 2009, it has arisen in multiple geographically distinct genomic clades nearly simultaneously. C. auris strains are typically multidrug resistant and colonize the skin much better than most other pathogenic fungi; it also persists on abiotic surfaces, enabling outbreaks due to transmission in health care facilities. All these suggest a biology substantially different from the 'model' fungal pathogen, Candida albicans and support intensive investigation of C. auris biology directly. To uncover novel virulence mechanisms in this species requires the development of appropriate animal infection models. Various studies using mice, the definitive model, are inconsistent due to differences in mouse and fungal strains, immunosuppressive regimes, doses, and outcome metrics. At the same time, developing models of skin colonization present a route to new insights into an aspect of fungal pathogenesis that has not been well studied in other species. We also discuss the growing use of nonmammalian model systems, including both vertebrates and invertebrates, such as zebrafish, C. elegans, Drosophila, and Galleria mellonella, that have been productively employed in virulence studies with other fungal species. This review will discuss progress in developing appropriate animal models, outline current challenges, and highlight opportunities in demystifying this curious species.
Collapse
Affiliation(s)
- Melissa Martinez
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School and the University of Texas Graduate School of Biomedical Sciences, USA
| | - Danielle A Garsin
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School and the University of Texas Graduate School of Biomedical Sciences, USA
| | - Michael C Lorenz
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School and the University of Texas Graduate School of Biomedical Sciences, USA.
| |
Collapse
|
12
|
Li C, Wang J, Li H, Wang Y, Wu H, Wei W, Wu D, Shao J, Wang T, Wang C. Suppressing the virulence factors of Candida auris with baicalein through multifaceted mechanisms. Arch Microbiol 2024; 206:349. [PMID: 38992278 DOI: 10.1007/s00203-024-04038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 07/13/2024]
Abstract
Candida auris, a rapidly spreading multi-drug-resistant fungus, is causing lethal infections under certain conditions globally. Baicalin (BE), an active ingredient extracted from the dried root of Scutellaria baicalensis Georgi, exhibits antifungal activity. However, studies have shown the distinctive advantages of Traditional Chinese medicine in combating fungal infections, while the effect of BE, an active ingredient extracted from the dried roots of Scutellaria baicalensis Georgi, on C. auris, remains unknown. Therefore, this study aims to evaluate the potential of BE as an antifungal agent against the emerging multidrug-resistant C. auris. Various assays and models, including microbroth dilution, time growth curve analysis, spot assays, adhesion tests, flocculation test, cell surface hydrophobicity assay, hydrolase activity assays, XTT assay, violet crystal assay, scanning electron microscope (SEM), confocal laser scanning microscope (CLSM), flow cytometry, Live/dead fluorescent staining, reactive oxygen species (ROS), cell wall assay, aggregation assay, porcine skin model, Galleria mellonella larvae (G. mellonella larvae) infection model, and reverse transcription-quantitative polymerase chain reaction (RT-PCR) were utilized to investigate how baicalein suppresses C. auris through possible multifaceted mechanisms. The findings indicate that BE strongly inhibited C. auris growth, adhesion, and biofilm formation. It also effectively reduced drug resistance and aggregation by disrupting the cell membrane and cell wall while reducing colonization and invasion of the host. Transcriptome analysis showed significant modulation in gene expression related to different virulence factors post-BE treatment. In conclusion, BE exhibits significant effectiveness against C. auris, suggesting its potential as a viable treatment option due to its multifaceted suppression mechanisms.
Collapse
Affiliation(s)
- Can Li
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Wang
- Anhui Provincial Institute for Food and Drug Control, Hefei, China
| | - Hao Li
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yemei Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hui Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Wenfan Wei
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Shao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tianming Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Changzhong Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, China.
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
13
|
Fayed B, Shakartalla SB, Sabbah H, Dalle H, Tannira M, Senok A, Soliman SSM. Transcriptome Analysis of Human Dermal Cells Infected with Candida auris Identified Unique Pathogenesis/Defensive Mechanisms Particularly Ferroptosis. Mycopathologia 2024; 189:65. [PMID: 38990436 DOI: 10.1007/s11046-024-00868-9] [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: 12/23/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Candida auris is an emerging multi-drug resistant yeast that can cause life-threatening infections. A recent report clarified the ability of C. auris to form a biofilm with enhanced drug resistance properties in the host skin's deep layers. The formed biofilm may initiate further bloodstream spread and immune escape. Therefore, we propose that secreted chemicals from the biofilm may facilitate fungal pathogenesis. In response to this interaction, the host skin may develop potential defensive mechanisms. Comparative transcriptomics was performed on the host dermal cells in response to indirect interaction with C. auris biofilm through Transwell inserts compared to planktonic cells. Furthermore, the effect of antifungals including caspofungin and fluconazole was studied. The obtained data showed that the dermal cells exhibited different transcriptional responses. Kyoto Encyclopedia of Genes and Genomes and Reactome analyses identified potential defensive responses employed by the dermal cells and potential toxicity induced by C. auris. Additionally, our data indicated that the dominating toxic effect was mediated by ferroptosis; which was validated by qRT-PCR, cytotoxicity assay, and flow cytometry. On the other hand, the viability of C. auris biofilm was enhanced and accompanied by upregulation of MDR1, and KRE6 upon interaction with dermal cells; both genes play significant roles in drug resistance and biofilm maturation, respectively. This study for the first-time shed light on the dominating defensive responses of human dermal cells, microbe colonization site, to C. auris biofilm and its toxic effects. Further, it demonstrates how C. auris biofilm responds to the defensive mechanisms developed by the human dermal cells.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Chemistry of Natural and Microbial Products, National Research Centre, Cairo, Egypt
| | - Sarra B Shakartalla
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Faculty of Pharmacy, University of Gezira, P.O.Box. 21111, Wad Medani, Sudan
| | - Hassan Sabbah
- AbbVie BioPharmaceuticals, P.O. Box 118052, Dubai, UAE
| | - Hala Dalle
- AbbVie BioPharmaceuticals, Kuwait City, Kuwait
| | | | - Abiola Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14 Dubai Healthcare City, P.O.Box 505055, Dubai, UAE
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
| |
Collapse
|
14
|
Chalin A, Arvor A, Hervault AS, Plaisance M, Niol L, Simon S, Volland H. A lateral flow immunoassay for the rapid identification of Candida auris from isolates or directly from surveillance enrichment broths. Front Microbiol 2024; 15:1439273. [PMID: 39021636 PMCID: PMC11252032 DOI: 10.3389/fmicb.2024.1439273] [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: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Candida auris is a recently discovered yeast with a multi-drug resistant profile associated with high mortality rates. The rapid identification of Candida auris in hospital settings is crucial to allow appropriate therapeutic and rapid implementation of infection management measures. The aim of this study was to develop a lateral flow immunoassay (LFIA) for the rapid identification of Candida auris. Methods Highly specific monoclonal antibodies were obtained by immunizing mice with membrane proteins from Candida auris which were then used to develop a LFIA whose performance was assessed by testing 12 strains of Candida auris and 37 strains of other Candida species. Isolates were grown on either Sabouraud dextrose, CHROMagarTM Candida Plus or HardyCHROMTM Candida + auris agar plates. The strains were also cultured on salt sabouraud-dextrose with chloramphenicol or a commercially available Salt-Sabouraud Dulcitol Broth with chloramphenicol and gentamicin, and processed using a simple centrifugation protocol to recover a pellet. Finally, the colonies or yeast extract were transferred to the LFIA to determine the specificity and sensitivity of the assay. Results The LFIA reached 100% specificity and sensitivity from solid agar plates. For both enrichment broths, some Candida non-auris species were able to grow, but the LFIA remained 100% specific. The use of a dextrose-based sabouraud broth resulted in earlier identification with the LFIA, with most of the Candida auris strains detected at 24 h. Conclusion The developed LFIA prototype represents a powerful tool to fight the emerging threat of Candida auris. Clinical validation represents the next step.
Collapse
Affiliation(s)
- Arnaud Chalin
- NG Biotech – Research and Development Department, Guipry-Messac, France
| | - Antoine Arvor
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Médicaments et Technologies pour la Santé (MTS), Service de Pharmacologie et d'Immunoanalyse (SPI), Laboratoire d'Etudes et de Recherches en Immunoanalyse (LERI), Gif-sur-Yvette, France
| | | | - Marc Plaisance
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Médicaments et Technologies pour la Santé (MTS), Service de Pharmacologie et d'Immunoanalyse (SPI), Laboratoire d'Etudes et de Recherches en Immunoanalyse (LERI), Gif-sur-Yvette, France
| | - Léa Niol
- NG Biotech – Research and Development Department, Guipry-Messac, France
| | - Stéphanie Simon
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Médicaments et Technologies pour la Santé (MTS), Service de Pharmacologie et d'Immunoanalyse (SPI), Laboratoire d'Etudes et de Recherches en Immunoanalyse (LERI), Gif-sur-Yvette, France
| | - Hervé Volland
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Médicaments et Technologies pour la Santé (MTS), Service de Pharmacologie et d'Immunoanalyse (SPI), Laboratoire d'Etudes et de Recherches en Immunoanalyse (LERI), Gif-sur-Yvette, France
| |
Collapse
|
15
|
Deng Y, Xu M, Li S, Bing J, Zheng Q, Huang G, Liao W, Pan W, Tao L. A single gene mutation underpins metabolic adaptation and acquisition of filamentous competence in the emerging fungal pathogen Candida auris. PLoS Pathog 2024; 20:e1012362. [PMID: 38976759 PMCID: PMC11257696 DOI: 10.1371/journal.ppat.1012362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/18/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Filamentous cell growth is a vital property of fungal pathogens. The mechanisms of filamentation in the emerging multidrug-resistant fungal pathogen Candida auris are poorly understood. Here, we show that exposure of C. auris to glycerol triggers a rod-like filamentation-competent (RL-FC) phenotype, which forms elongated filamentous cells after a prolonged culture period. Whole-genome sequencing analysis reveals that all RL-FC isolates harbor a mutation in the C2H2 zinc finger transcription factor-encoding gene GFC1 (Gfc1 variants). Deletion of GFC1 leads to an RL-FC phenotype similar to that observed in Gfc1 variants. We further demonstrate that GFC1 mutation causes enhanced fatty acid β-oxidation metabolism and thereby promotes RL-FC/filamentous growth. This regulation is achieved through a Multiple Carbon source Utilizer (Mcu1)-dependent mechanism. Interestingly, both the evolved RL-FC isolates and the gfc1Δ mutant exhibit an enhanced ability to colonize the skin. Our results reveal that glycerol-mediated GFC1 mutations are beneficial during C. auris skin colonization and infection.
Collapse
Affiliation(s)
- Yuchen Deng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ming Xu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuaihu Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Bing
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiushi Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Guanghua Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanqing Liao
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Weihua Pan
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Li Tao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Das S, Singh S, Tawde Y, Dutta TK, Rudramurthy SM, Kaur H, Shaw T, Ghosh A. Comparative fitness trade-offs associated with azole resistance in Candida auris clinical isolates. Heliyon 2024; 10:e32386. [PMID: 38988564 PMCID: PMC11233892 DOI: 10.1016/j.heliyon.2024.e32386] [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: 02/23/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Multidrug-resistant yeast Candida auris is a serious threat to public health with documented survival in various hospital niches. The dynamics of this survival benefit and its trade off with drug resistance are still unknown for this pathogen. In this study we investigate the oxidative stress response (OSR) in fluconazole-resistant C. auris and compare its relative fitness with fluconazole-susceptible strains. A total of 351 C. auris clinical isolates (61 fluconazole-susceptible and 290 fluconazole-resistant) were screened for stress tolerance by spot assay and 95.08 % fluconazole-susceptible isolates were hyper-resistant to oxidative stress while majority (94.5 %) fluconazole-resistant isolates had lower oxidative tolerance. Expression of Hog1 and Cta1 gene transcript levels and cellular catalase levels were significantly higher in fluconazole-susceptible isolates and a corresponding higher intracellular reactive oxygen species level (iROS) was accumulated in the fluconazole-resistant isolates. Biofilm formation and cell viability under oxidative stress revealed higher biofilm formation and better viability in fluconazole-susceptible isolates. Fluconazole-resistant isolates had higher basal cell wall chitin. On comparison of virulence, the % cytotoxicity in A549 cell line was higher in fluconazole-susceptible isolates and the median survival of the infected larvae in G. mellonella infection model was higher in fluconazole-resistant (5; IQR:4.5-5 days) vs. fluconazole-susceptible C. auris (2; IQR:1.5-2.5 days). All organisms evolve with changes in their environmental conditions, to ensure an optimal balance between proliferation and survival. Development of tolerance to a certain kind of stress example antifungal exposure in yeast can leads to a compensatory decrease in tolerance for other stresses. This study provides useful insights into the comparative fitness and antifungal susceptibility trade off in C. auris. We report a negative association between H2O2 tolerance and fluconazole susceptibility. Using in-vitro cell cytotoxicity and in-vivo survival assays we also demonstrate the higher virulence potential of fluconazole-susceptible C. auris isolates corroborating the negative correlation between susceptibility and pathogen survival or virulence. These findings could also be translated to clinical practice by investigating the possibility of using molecules targeting stress response and fitness regulating pathways for management of this serious infection.
Collapse
Affiliation(s)
- Sourav Das
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shreya Singh
- Department of Microbiology. Dr. B. R. Ambedkar State Institute of Medical Science, Mohali, Punjab, India
| | - Yamini Tawde
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Tushar K. Dutta
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Shivaprakash M. Rudramurthy
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harsimran Kaur
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Tushar Shaw
- Department of Life and Allied Health Sciences, Ramaiah university of Applied sciences, Bangalore, India
| | - Anup Ghosh
- Department of Medical Microbiology. Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
17
|
Stindt KR, McClean MN. Tuning interdomain conjugation to enable in situ population modification in yeasts. mSystems 2024; 9:e0005024. [PMID: 38747597 PMCID: PMC11326116 DOI: 10.1128/msystems.00050-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/15/2024] [Indexed: 05/28/2024] Open
Abstract
The ability to modify and control natural and engineered microbiomes is essential for biotechnology and biomedicine. Fungi are critical members of most microbiomes, yet technology for modifying the fungal members of a microbiome has lagged far behind that for bacteria. Interdomain conjugation (IDC) is a promising approach, as DNA transfer from bacterial cells to yeast enables in situ modification. While such genetic transfers have been known to naturally occur in a wide range of eukaryotes and are thought to contribute to their evolution, IDC has been understudied as a technique to control fungal or fungal-bacterial consortia. One major obstacle to the widespread use of IDC is its limited efficiency. In this work, we manipulated metabolic and physical interactions between genetically tractable Escherichia coli and Saccharomyces cerevisiae to control the incidence of IDC. We test the landscape of population interactions between the bacterial donors and yeast recipients to find that bacterial commensalism leads to maximized IDC, both in culture and in mixed colonies. We demonstrate the capacity of cell-to-cell binding via mannoproteins to assist both IDC incidence and bacterial commensalism in culture and model how these tunable controls can predictably yield a range of IDC outcomes. Furthermore, we demonstrate that these controls can be utilized to irreversibly alter a recipient yeast population, by both "rescuing" a poor-growing recipient population and collapsing a stable population via a novel IDC-mediated CRISPR/Cas9 system.IMPORTANCEFungi are important but often unaddressed members of most natural and synthetic microbial communities. This work highlights opportunities for modifying yeast microbiome populations through bacterial conjugation. While conjugation has been recognized for its capacity to deliver engineerable DNA to a range of cells, its dependence on cell contact has limited its efficiency. Here, we find "knobs" to control DNA transfer, by engineering the metabolic dependence between bacterial donors and yeast recipients and by changing their ability to physically adhere to each other. Importantly, we functionally validate these "knobs" by irreversibly altering yeast populations. We use these controls to "rescue" a failing yeast population, demonstrate the capacity of conjugated CRISPR/Cas9 to depress or collapse populations, and show that conjugation can be easily interrupted by disrupting cell-to-cell binding. These results offer building blocks toward in situ mycobiome editing, with significant implications for clinical treatments of fungal pathogens and other fungal system engineering.
Collapse
Affiliation(s)
- Kevin R Stindt
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Doctoral Program in Biophysics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Megan N McClean
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
18
|
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] [Grants] [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
|
19
|
Hansanant N, Cao K, Tenorio A, Joseph T, Ju M, McNally N, Kummari E, Williams M, Cothrell A, Buhrow AR, Shin R, Orugunty R, Smith L. Previously Uncharacterized Variants, OCF-E-OCF-J, of the Antifungal Occidiofungin Produced by Burkholderia contaminans MS14. JOURNAL OF NATURAL PRODUCTS 2024; 87:186-194. [PMID: 38277493 PMCID: PMC10897925 DOI: 10.1021/acs.jnatprod.3c00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/05/2023] [Accepted: 12/17/2023] [Indexed: 01/28/2024]
Abstract
The rise of multidrug resistant fungal infections highlights the need to identify and develop novel antifungal agents. Occidiofungin is a nonribosomally synthesized glycolipopeptide that has a unique mechanism of action, disrupting actin-mediated functions and inducing cellular apoptosis. Antifungal activity has been observed in vitro against various fungal species, including multidrug resistant Candida auris, and in vivo efficacy has been demonstrated in a murine vulvovaginal candidiasis model. Occidiofungin, a cyclic glycolipopeptide, is composed of eight amino acids and in previous studies, an asparagine residue was assigned at position 7 (ASN7). In this study, new structural variants of occidiofungin have been characterized which have aspartic acid (ASP7), glutamine (GLN7), or glutamic acid (GLU7) at position 7. The side chain of the ASP7 variant contains the only terminal carboxylic acid in the peptide and provides a useful site for selective chemical modifications. Analogues were synthesized at the ASP7 position and tested for antifungal activity. These analogues were shown to be more active as compared to the ASP7 variant against a panel of Candida species. The naturally occurring variants of occidiofungin with a side chain containing a carboxylic acid at the seventh amino acid position can be used to develop semisynthetic analogues with enhanced therapeutic properties.
Collapse
Affiliation(s)
- Nopakorn Hansanant
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Kevin Cao
- Sano
Chemicals Incorporated, Bryan, Texas 77803, United States
| | - Abraham Tenorio
- Sano
Chemicals Incorporated, Bryan, Texas 77803, United States
| | - Thushinari Joseph
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Min Ju
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Noah McNally
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Evangel Kummari
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - McKinley Williams
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Andrew Cothrell
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Andrew R. Buhrow
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
| | - Ronald Shin
- Central
Alabama High-Field NMR Facility, Structural Biology Shared Facility,
Cancer Center, University of Alabama at
Birmingham, Birmingham, Alabama 35294-1240, United States
| | - Ravi Orugunty
- Sano
Chemicals Incorporated, Bryan, Texas 77803, United States
| | - Leif Smith
- Department
of Biology, Texas A&M University, College Station, Texas 77843, United States
- Sano
Chemicals Incorporated, Bryan, Texas 77803, United States
| |
Collapse
|
20
|
Omardien S, Teska P. Skin and hard surface disinfection against Candida auris - What we know today. Front Med (Lausanne) 2024; 11:1312929. [PMID: 38384416 PMCID: PMC10879571 DOI: 10.3389/fmed.2024.1312929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/05/2024] [Indexed: 02/23/2024] Open
Abstract
Candida auris has emerged as a global healthcare threat, displaying resistance to important healthcare antifungal therapies. Infection prevention and control protocols have become paramount in reducing transmission of C. auris in healthcare, of which cleaning and disinfection plays an important role. Candida albicans is used as a surrogate yeast for yeasticidal claims of disinfection products, but reports have been made that sensitivity to disinfectants by C. auris differs from its surrogate. In this review, we aimed to compile the information reported for products used for skin and hard surface disinfection against C. auris in its planktonic or biofilm form. A comparison was made with other Candida species, and information were gathered from laboratory studies and observations made in healthcare settings.
Collapse
Affiliation(s)
| | - Peter Teska
- Diversey Holdings Ltd., Fort Mill, SC, United States
| |
Collapse
|
21
|
Schapira AJ, Dramé M, Olive C, Marion-Sanchez K. Bacterial viability in dry-surface biofilms in healthcare facilities: a systematic review. J Hosp Infect 2024; 144:94-110. [PMID: 38029859 DOI: 10.1016/j.jhin.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Bacteria are known to live inside architectural structures called biofilms. Though standard biofilms have been studied extensively for more than 50 years, little is known about dry-surface biofilms (DSBs). Since 2012, DSBs have been described in several scientific papers, but basic knowledge about the viability and culturability of bacteria remains limited. AIM To conduct a systematic review to determine whether bacteria inside DSBs are viable, culturable, and enumerable. METHODS Eligible articles had to deal with DSBs containing at least one bacterial species involved in healthcare-associated infections, which developed in actual healthcare environments (in-situ) or with the help of any biofilm model (in-vitro). FINDINGS Twenty-four articles were included in the review. Whereas most of them isolated viable bacteria (87% in situ; 100% in vitro), no in-situ study quantified culturable bacteria in the biofilm per unit area. Conversely, 100% of in-vitro studies cultured the bacteria from controls and 94.4% supplied an enumeration of them. Culturable bacteria also grew after 78% of the cleaning, disinfection, or sterilization protocols tested. Microscopic observations after staining the samples with live/dead fluorescent probes (Baclight®) showed large amounts of viable cells in culture-negative samples. CONCLUSION Our study questions the efficacy of current methods for microbiological monitoring of surfaces, since these methods are only based on bacterial culturability. To improve both surface monitoring and cleaning and disinfection protocols, it is necessary to integrate the concept of DSBs which appears to contain a significant amount of viable but non-culturable bacteria.
Collapse
Affiliation(s)
- A-J Schapira
- CHU Martinique, Department of Bacteriology, Hygiene and Environment Laboratory, Fort-de-France Cedex, Martinique; Paris Cité University, Faculty of Health, Paris, France
| | - M Dramé
- CHU Martinique, Department of Clinical Research and Innovation, Fort-de-France Cedex, Martinique; University of the French West Indies, Faculty of Medicine, EpiCliV Research Unit, Martinique
| | - C Olive
- CHU Martinique, Department of Bacteriology, Hygiene and Environment Laboratory, Fort-de-France Cedex, Martinique
| | - K Marion-Sanchez
- CHU Martinique, Department of Bacteriology, Hygiene and Environment Laboratory, Fort-de-France Cedex, Martinique; PCCEI, University of Montpellier, University of the Antilles, INSERM, EFS, Montpellier, France.
| |
Collapse
|
22
|
Jaromin A, Zarnowski R, Markowski A, Zagórska A, Johnson CJ, Etezadi H, Kihara S, Mota-Santiago P, Nett JE, Boyd BJ, Andes DR. Liposomal formulation of a new antifungal hybrid compound provides protection against Candida auris in the ex vivo skin colonization model. Antimicrob Agents Chemother 2024; 68:e0095523. [PMID: 38092678 PMCID: PMC10777852 DOI: 10.1128/aac.00955-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/28/2023] [Indexed: 01/11/2024] Open
Abstract
The newly emerged pathogen, Candida auris, presents a serious threat to public health worldwide. This multidrug-resistant yeast often colonizes and persists on the skin of patients, can easily spread from person to person, and can cause life-threatening systemic infections. New antifungal therapies are therefore urgently needed to limit and control both superficial and systemic C. auris infections. In this study, we designed a novel antifungal agent, PQA-Az-13, that contains a combination of indazole, pyrrolidine, and arylpiperazine scaffolds substituted with a trifluoromethyl moiety. PQA-Az-13 demonstrated antifungal activity against biofilms of a set of 10 different C. auris clinical isolates, representing all four geographical clades distinguished within this species. This compound showed strong activity, with MIC values between 0.67 and 1.25 µg/mL. Cellular proteomics indicated that PQA-Az-13 partially or completely inhibited numerous enzymatic proteins in C. auris biofilms, particularly those involved in both amino acid biosynthesis and metabolism processes, as well as in general energy-producing processes. Due to its hydrophobic nature and limited aqueous solubility, PQA-Az-13 was encapsulated in cationic liposomes composed of soybean phosphatidylcholine (SPC), 1,2-dioleoyloxy-3-trimethylammonium-propane chloride (DOTAP), and N-(carbonyl-methoxypolyethylene glycol-2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine, sodium salt (DSPE-PEG 2000), and characterized by biophysical and spectral techniques. These PQA-Az-13-loaded liposomes displayed a mean size of 76.4 nm, a positive charge of +45.0 mV, a high encapsulation efficiency of 97.2%, excellent stability, and no toxicity to normal human dermal fibroblasts. PQA-Az-13 liposomes demonstrated enhanced antifungal activity levels against both C. auris in in vitro biofilms and ex vivo skin colonization models. These initial results suggest that molecules like PQA-Az-13 warrant further study and development.
Collapse
Affiliation(s)
- Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Robert Zarnowski
- Department of Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Adam Markowski
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Agnieszka Zagórska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, Cracow, Poland
| | - Chad J. Johnson
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Haniyeh Etezadi
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Shinji Kihara
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | - Jeniel E. Nett
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, USA
| | - Ben J. Boyd
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Victoria, Australia
| | - David R. Andes
- Department of Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
23
|
Balakumar A, Bernstein D, Thangamani S. The adhesin SCF1 mediates Candida auris colonization. Trends Microbiol 2024; 32:4-5. [PMID: 37951769 PMCID: PMC10872899 DOI: 10.1016/j.tim.2023.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023]
Abstract
Candida auris is an emerging human fungal pathogen that can rapidly spread and cause outbreaks of invasive infections. Santana et al. discovered that a novel surface colonization factor (SCF1), and a conserved adhesin, Iff4109, mediates C. auris colonization on abiotic surfaces, skin, and virulence in vivo.
Collapse
Affiliation(s)
- Abishek Balakumar
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | | | - Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA; Purdue Institute for Immunology, Inflammation, and Infectious Diseases (PI4D), Purdue University, West Lafayette, IN 47906, USA.
| |
Collapse
|
24
|
Seiser S, Arzani H, Ayub T, Phan-Canh T, Staud C, Worda C, Kuchler K, Elbe-Bürger A. Native human and mouse skin infection models to study Candida auris-host interactions. Microbes Infect 2024; 26:105234. [PMID: 37813159 DOI: 10.1016/j.micinf.2023.105234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
The World Health Organization (WHO) declared certain fungal pathogens as global health threats for the next decade. Candida auris (C. auris) is a newly emerging skin-tropic multidrug-resistant fungal pathogen that can cause life-threatening infections of high mortality in hospitals and healthcare settings. Here, we address an unmet need and present novel native ex vivo skin models, thus extending previous C. auris-host interaction studies. We exploit histology and immunofluorescence analysis of ex vivo skin biopsies of human adult and fetal, as well as mouse origin infected with C. auris via distinct routes. We demonstrate that an intact skin barrier efficiently protects from C. auris penetration and invasion. Although C. auris readily grows on native human skin, it can reach deeper layers only upon physical disruption of the barrier by needling or through otherwise damaged skin. By contrast, a barrier disruption is not necessary for C. auris penetration of native mouse skin. Importantly, we show that C. auris undergoes morphogenetic changes upon skin penetration, as it acquires pseudohyphal growth phenotypes in deeper human and mouse dermis. Taken together, this new human and mouse skin model toolset yields new insights into C. auris colonization, adhesion, growth and invasion properties of native versus damaged human skin. The results form a crucial basis for future studies on skin immune defense to colonizing pathogens, and offer new options for testing the action and efficacy of topical antimicrobial compound formulations.
Collapse
Affiliation(s)
- Saskia Seiser
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Hossein Arzani
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Tanya Ayub
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Trinh Phan-Canh
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Clement Staud
- Medical University of Vienna, Department of Plastic and Reconstructive Surgery, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Christof Worda
- Medical University of Vienna, Department of Obstetrics and Gynecology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria.
| | - Adelheid Elbe-Bürger
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
25
|
Akinbobola A, Kean R, Quilliam RS. Plastic pollution as a novel reservoir for the environmental survival of the drug resistant fungal pathogen Candida auris. MARINE POLLUTION BULLETIN 2024; 198:115841. [PMID: 38061145 DOI: 10.1016/j.marpolbul.2023.115841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024]
Abstract
The WHO recently classified Candida auris as a fungal pathogen of "critical concern". Evidence suggests that C. auris emerged from the natural environment, yet the ability of this pathogenic yeast to survive in the natural environment is still poorly understood. The aim of this study, therefore, was to quantify the persistence of C. auris in simulated environmental matrices and explore the role of plastic pollution for facilitating survival and potential transfer of C. auris. Multi-drug resistant strains of C. auris persisted for over 30 days in river water or seawater, either planktonically, or in biofilms colonising high-density polyethylene (HDPE) or glass. C. auris could be transferred from plastic beads onto simulated beach sand, particularly when the sand was wet. Importantly, all C. auris cells recovered from plastics retained their pathogenicity; therefore, plastic pollution could play a significant role in the widescale environmental dissemination of this recently emerged pathogen.
Collapse
Affiliation(s)
- Ayorinde Akinbobola
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK.
| | - Ryan Kean
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Richard S Quilliam
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| |
Collapse
|
26
|
Cox CA, Manavathu EK, Wakade S, Myntti M, Vazquez JA. Efficacy of biofilm disrupters against Candida auris and other Candida species in monomicrobial and polymicrobial biofilms. Mycoses 2024; 67:e13684. [PMID: 38214428 DOI: 10.1111/myc.13684] [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: 04/07/2023] [Revised: 10/19/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
Candida species are now considered global threats by the CDC and WHO. Candida auris specifically is on the critical pathogen threat list along with Candida albicans. In addition, it is not uncommon to find Candida spp. in a mixed culture with bacterial organisms, especially Staphylococcus aureus producing polymicrobial infections. To eradicate these organisms from the environment and from patient surfaces, surface agents such as chlorhexidine (CHD) and Puracyn are used. Biofilm disrupters (BDs) are novel agents with a broad spectrum of antimicrobial activity and have been used in the management of chronic wounds and to sterilise environmental surfaces for the past several years. The goal of this study was to evaluate BDs (BlastX, Torrent, NSSD) and CHD against Candida spp. and S. aureus using zone of inhibition assays, biofilm and time-kill assays. All BDs and CHD inhibited C. auris growth effectively in a concentration-dependent manner. Additionally, CHD and the BDs showed excellent antimicrobial activity within polymicrobial biofilms. A comparative analysis of the BDs and CHD against C. auris and C. albicans using biofilm kill-curves showed at least 99.999% killing. All three BDs and CHD have excellent activity against different Candida species, including C. auris. However, one isolate of C. auris in a polymicrobial biofilm assay showed resistance/tolerance to CHD, but not to the BDs. The fungicidal activity of these novel agents will be valuable in eradicating surface colonisation of Candida spp, especially C. auris from colonised environmental surfaces and from wounds in colonised patients.
Collapse
Affiliation(s)
- Claudia A Cox
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Elias K Manavathu
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Sushama Wakade
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | | | - Jose A Vazquez
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
27
|
Wells KM, Ciftci Y, Peddinti BST, Ghiladi RA, Vediyappan G, Spontak RJ, Govind R. Preventing the spread of life-threatening gastrointestinal microbes on the surface of a continuously self-disinfecting block polymer. J Colloid Interface Sci 2023; 652:718-726. [PMID: 37611471 DOI: 10.1016/j.jcis.2023.08.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/03/2023] [Accepted: 08/12/2023] [Indexed: 08/25/2023]
Abstract
Highly persistent, drug-resistant and transmissible healthcare pathogens such as Clostridioides difficile (C. difficile) and Candida auris (C. auris) are responsible for causing antibiotic-associated fatal diarrhea and invasive candidiasis, respectively. In this study, we demonstrate that these potentially lethal gastrointestinal microbes can be rapidly inactivated on the solid surface of a self-disinfecting anionic block polymer that inherently generates a water surface layer that is highly acidic (pH < 1) upon hydration. Due to thermodynamic incompatibility between its chemical sequences, the polymer spontaneously self-organizes into a nanostructure that enables proton migration from the interior of a film to the surface via contiguous nanoscale hydrophilic channels, as discerned here by scanning electron and atomic force microscopies, as well as X-ray photoelectron spectroscopy. Here, we report that two strains of C. difficile in the vegetative state and two species of Candida, Candida albicans (C. albicans) and C. auris, are, in most cases, inactivated to the limit of minimum detection. Corresponding electron and optical microscopy images reveal that, upon exposure to the hydrated polymer, the outer microbial membranes display evidence of damage and intracellular material is expelled. Combined with our previous studies of rapid bacterial and viral inactivation, these antimicrobial results are highly encouraging and, if translatable to clinical conditions in the form of self-standing polymer films or coatings, are expected to benefit the welfare of patients in healthcare facilities by continuously preventing the spread of such potentially dangerous microbes.
Collapse
Affiliation(s)
- Kacie M Wells
- Fiber & Polymer Science Program, North Carolina State University, Raleigh, NC 27695, United States
| | - Yusuf Ciftci
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States
| | - Bharadwaja S T Peddinti
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Reza A Ghiladi
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, United States
| | | | - Richard J Spontak
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States; Department of Materials Science & Engineering, North Carolina State University, Raleigh, NC 27695, United States.
| | - Revathi Govind
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States.
| |
Collapse
|
28
|
Rapti V, Iliopoulou K, Poulakou G. The Gordian Knot of C. auris: If You Cannot Cut It, Prevent It. Pathogens 2023; 12:1444. [PMID: 38133327 PMCID: PMC10747958 DOI: 10.3390/pathogens12121444] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Since its first description in 2009, Candida auris has, so far, resulted in large hospital outbreaks worldwide and is considered an emerging global public health threat. Exceptionally for yeast, it is gifted with a profoundly worrying invasive potential and high inter-patient transmissibility. At the same time, it is capable of colonizing and persisting in both patients and hospital settings for prolonged periods of time, thus creating a vicious cycle of acquisition, spreading, and infection. It exhibits various virulence qualities and thermotolerance, osmotolerance, filamentation, biofilm formation and hydrolytic enzyme production, which are mainly implicated in its pathogenesis. Owing to its unfavorable profile of resistance to diverse antifungal agents and the lack of effective treatment options, the implementation of robust infection prevention and control (IPC) practices is crucial for controlling and minimizing intra-hospital transmission of C. auris. Rapid and accurate microbiological identification, adherence to hand hygiene, use of adequate personal protective equipment (PPE), proper handling of catheters and implantable devices, contact isolation, periodical environmental decontamination, targeted screening, implementation of antimicrobial stewardship (AMS) programs and communication between healthcare facilities about residents' C. auris colonization status are recognized as coherent strategies for preventing its spread. Current knowledge on C. auris epidemiology, clinical characteristics, and its mechanisms of pathogenicity are summarized in the present review and a comprehensive overview of IPC practices ensuring yeast prevention is also provided.
Collapse
Affiliation(s)
- Vasiliki Rapti
- Third Department of Internal Medicine, School of Medicine, National & Kapodistrian University of Athens, Sotiria General Hospital, 115 27 Athens, Greece;
| | | | - Garyfallia Poulakou
- Third Department of Internal Medicine, School of Medicine, National & Kapodistrian University of Athens, Sotiria General Hospital, 115 27 Athens, Greece;
| |
Collapse
|
29
|
Wang Q, Cheng S, Wang Y, Li F, Chen J, Du W, Kang H, Wang Z. Global characteristics and trends in research on Candida auris. Front Microbiol 2023; 14:1287003. [PMID: 38125576 PMCID: PMC10731253 DOI: 10.3389/fmicb.2023.1287003] [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: 09/01/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Candida auris, a fungal pathogen first reported in 2009, has shown strong resistance to azole antifungal drugs and has caused severe nosocomial outbreaks. It can also form biofilms, which can colonize patients' skin and transmit to others. Despite numerous reports of C. auris isolation in various countries, many studies have reported contradictory results. Method A bibliometric analysis was conducted using VOSviewer to summarize research trends and provide guidance for future research on controlling C. auris infection. The analysis revealed that the United States and the US CDC were the most influential countries and research institutions, respectively. For the researchers, Jacques F. Meis published the highest amount of related articles, and Anastasia P. Litvintseva's articles with the highest average citation rate. The most cited publications focused on clade classification, accurate identification technologies, nosocomial outbreaks, drug resistance, and biofilm formation. Keyword co-occurrence analysis revealed that the top five highest frequencies were for 'drug resistance,' 'antifungal susceptibility test,' 'infection,' 'Candida auris,' and 'identification.' The high-frequency keywords clustered into four groups: rapid and precise identification, drug resistance research, pathogenicity, and nosocomial transmission epidemiology studies. These clusters represent different study fields and current research hotspots of C. auris. Conclusion The bibliometric analysis identified the most influential country, research institution, and researcher, indicating current research trends and hotspots for controlling C. auris.
Collapse
Affiliation(s)
- Qihui Wang
- Laboratory of Microbiology, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shitong Cheng
- Laboratory of Microbiology, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yinling Wang
- Laboratory of Microbiology, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fushun Li
- Laboratory of Microbiology, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingjing Chen
- Laboratory of Microbiology, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei Du
- National Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hui Kang
- Laboratory of Microbiology, Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongqing Wang
- Department of Information Centre, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
30
|
Lockhart SR, Chowdhary A, Gold JAW. The rapid emergence of antifungal-resistant human-pathogenic fungi. Nat Rev Microbiol 2023; 21:818-832. [PMID: 37648790 PMCID: PMC10859884 DOI: 10.1038/s41579-023-00960-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/01/2023]
Abstract
During recent decades, the emergence of pathogenic fungi has posed an increasing public health threat, particularly given the limited number of antifungal drugs available to treat invasive infections. In this Review, we discuss the global emergence and spread of three emerging antifungal-resistant fungi: Candida auris, driven by global health-care transmission and possibly facilitated by climate change; azole-resistant Aspergillus fumigatus, driven by the selection facilitated by azole fungicide use in agricultural and other settings; and Trichophyton indotineae, driven by the under-regulated use of over-the-counter high-potency corticosteroid-containing antifungal creams. The diversity of the fungi themselves and the drivers of their emergence make it clear that we cannot predict what might emerge next. Therefore, vigilance is critical to monitoring fungal emergence, as well as the rise in overall antifungal resistance.
Collapse
Affiliation(s)
- Shawn R Lockhart
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - 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, Medical Mycology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Jeremy A W Gold
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
31
|
Abstract
Candida auris recently emerged as an urgent public health threat, causing outbreaks of invasive infections in healthcare settings throughout the world. This fungal pathogen persists on the skin of patients and on abiotic surfaces despite antiseptic and decolonization attempts. The heightened capacity for skin colonization and environmental persistence promotes rapid nosocomial spread. Following skin colonization, C. auris can gain entrance to the bloodstream and deeper tissues, often through a wound or an inserted medical device, such as a catheter. C. auris possesses a variety of virulence traits, including the capacity for biofilm formation, production of adhesins and proteases, and evasion of innate immune responses. In this review, we highlight the interactions of C. auris with the host, emphasizing the intersection of laboratory studies and clinical observations.
Collapse
Affiliation(s)
- Mark V. Horton
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medical Microbiology & Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ashley M. Holt
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medical Microbiology & Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Jeniel E. Nett
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medical Microbiology & Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
32
|
Tuor M, LeibundGut-Landmann S. The skin mycobiome and intermicrobial interactions in the cutaneous niche. Curr Opin Microbiol 2023; 76:102381. [PMID: 37703811 DOI: 10.1016/j.mib.2023.102381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
Mammalian microbiomes have coevolved with their host to establish a stable homeostatic relationship. Multifaceted commensal-host and commensal-commensal interactions contribute to the maintenance of the equilibrium with an impact on diverse host physiological processes. Despite constant exposure to physical and chemical insults from the environment, the skin harbors a surprisingly stable microbiome. The fungal compartment of the skin microbiome, the skin mycobiome, is unique in that it is dominated by a single fungus, Malassezia. The lack in diversity suggests that the skin may provide a unique niche for this fungal genus and that Malassezia may efficiently outcompete other fungi from the skin. This opinion article examines aspects in support of this hypothesis, discusses how changes in niche conditions associate with skin mycobiome dysregulation, and highlights an emerging example of Malassezia being displaced from the skin by the emerging fungal pathogen C. auris, thereby generating a predisposing situation for fatal-invasive infection.
Collapse
Affiliation(s)
- Meret Tuor
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zurich, Switzerland.
| |
Collapse
|
33
|
Mulet Bayona JV, Tormo Palop N, Salvador García C, Guna Serrano MDR, Gimeno Cardona C. Candida auris from colonisation to candidemia: A four-year study. Mycoses 2023; 66:882-890. [PMID: 37401661 DOI: 10.1111/myc.13626] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Candida auris has become a worrisome multi-drug resistant healthcare-associated pathogen due to its capacity to colonise patients and surfaces and to cause outbreaks of invasive infections in critically ill patients. OBJECTIVES This study evaluated the outbreak in our setting in a 4-year period, reporting the risk factors for developing candidemia in previously colonised patients, the therapeutic measures for candidemia and the outcome of candidemia and colonisation cases among all C. auris isolates and their susceptibility to antifungals. METHODS Data were retrospectively collected from patients admitted to Consorcio Hospital General Universitario de Valencia (Spain) from September 2017 to September 2021. A retrospective case-control study was designed to identify risk factors for developing C. auris candidemia in previously colonised patients. RESULTS C. auris affected 550 patients, of which 210 (38.2%) had some clinical sample positive. Isolates were uniformly resistant to fluconazole, 20 isolates were resistant to echinocandins (2.8%) and four isolates were resistant to ampfotericin B (0.6%). There were 86 candidemia cases. APACHE II, digestive disease and catheter isolate were proven to be independent risk factors for developing candidemia in previously colonised patients. Thirty-day mortality rate for C. auris candidemia cases was 32.6%, while for colonisation cases was 33.7%. CONCLUSIONS Candidemia was one of the most frequent and severe infections caused by C. auris. The risk factors identified in this study should help to detect patients who are at more risk of developing candidemia, as long as an adequate surveillance of C. auris colonisation is performed.
Collapse
Affiliation(s)
- Juan Vicente Mulet Bayona
- Department of Microbiology and Parasitology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Nuria Tormo Palop
- Department of Microbiology and Parasitology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Carme Salvador García
- Department of Microbiology and Parasitology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | | | - Concepción Gimeno Cardona
- Department of Microbiology and Parasitology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
- Department of Microbiology and Ecology, University of Valencia, Valencia, Spain
| |
Collapse
|
34
|
Carty J, Chowdhary A, Bernstein D, Thangamani S. Tools and techniques to identify, study, and control Candida auris. PLoS Pathog 2023; 19:e1011698. [PMID: 37856418 PMCID: PMC10586630 DOI: 10.1371/journal.ppat.1011698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Candida auris, is an emerging fungal pathogen that can cause life-threatening infections in humans. Unlike many other Candida species that colonize the intestine, C. auris most efficiently colonizes the skin. Such colonization contaminates the patient's environment and can result in rapid nosocomial transmission. In addition, this transmission can lead to outbreaks of systemic infections that have mortality rates between 40% and 60%. C. auris isolates resistant to all known classes of antifungals have been identified and as such, understanding the underlying biochemical mechanisms of how skin colonization initiates and progresses is critical to developing better therapeutic options. With this review, we briefly summarize what is known about horizontal transmission and current tools used to identify, understand, and control C. auris infections.
Collapse
Affiliation(s)
- James Carty
- Department of Biology, Ball State University, Muncie, Indiana, United States of America
| | - 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
| | - Douglas Bernstein
- Department of Biology, Ball State University, Muncie, Indiana, United States of America
| | - Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), Indiana, United States of America
| |
Collapse
|
35
|
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: 42] [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
|
36
|
Biswas B, Rana A, Gupta N, Gupta I, Puria R, Thakur A. A Novel Robust Method Mimicking Human Substratum To Dissect the Heterogeneity of Candida auris Biofilm Formation. Microbiol Spectr 2023; 11:e0089223. [PMID: 37439683 PMCID: PMC10434199 DOI: 10.1128/spectrum.00892-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/21/2023] [Indexed: 07/14/2023] Open
Abstract
Candida auris is a pathogen of urgent threat level as marked by the CDC. The formation of biofilms is an essential property of this fungus to establish infection and escape drug treatment. However, our understanding of pathogenesis through biofilm is hampered by heterogeneity in C. auris biofilms observed in different studies. It is imperative to replicate in vivo conditions for studying C. auris biofilm formation in vitro. Different methods are standardized, but the surface used to form biofilms lacks consistency as well as the architecture of a typical biofilm. Here, we report an in vitro technique to grow C. auris biofilms on gelatin-coated coverslips. Interestingly, C. auris cells grown on gelatin-coated coverslips either on modified synthetic sweat media or RPMI 1640 resulted in similar multilayer biofilm formation with extracellular polymeric substances (EPS). This method is also consistent with the biofilm formation of other Candida species, such as Candida glabrata and Candida albicans. Biofilms of C. glabrata developed through this method show pseudohyphae and EPS. This method can be used to understand the molecular basis of biofilm formation, associated pathogenesis, and drug tolerance. The technique is cost-effective and would thus serve in rightful screening and repurposing drug libraries for designing new therapeutics against the less-studied high-alarm pathogen C. auris. IMPORTANCE Heterogeneity is seen when multidrug-resistant C. auris biofilm is cultured using different reported methods. Biofilm formed on the gelatin surface mimics the condition of a host environment that has multilayers and EPS. This method has feasibility for drug screening and analyzing biofilms through three-dimensional (3D) reconstruction. This in vitro biofilm formation technique is also exploited to study the formation of biofilm of other Candida species. The biofilms of C. glabrata and C. albicans can also be correctly mimicked using gelatin in the biofilm-forming environment. Thus, the novel in vitro method for biofilm formation reported here can be widely used to understand the mechanism of biofilm formation, related virulence properties, and drug tolerance of C. auris and other Candida species. This simple and low-cost technique is highly suitable for screening novel inhibitors and repurposed libraries and to design new therapeutics against Candida species.
Collapse
Affiliation(s)
- Biswambhar Biswas
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Aishwarya Rana
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Nidhi Gupta
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Delhi, India
| | - Rekha Puria
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, India
| | - Anil Thakur
- Laboratory of Protein Translation and Fungal Pathogenesis, Regional Centre for Biotechnology, Faridabad, Haryana, India
| |
Collapse
|
37
|
Hetta HF, Ramadan YN, Al-Kadmy IMS, Ellah NHA, Shbibe L, Battah B. Nanotechnology-Based Strategies to Combat Multidrug-Resistant Candida auris Infections. Pathogens 2023; 12:1033. [PMID: 37623993 PMCID: PMC10458664 DOI: 10.3390/pathogens12081033] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
An emerging multidrug-resistant pathogenic yeast called Candida auris has a high potential to spread quickly among hospitalized patients and immunodeficient patients causing nosocomial outbreaks. It has the potential to cause pandemic outbreaks in about 45 nations with high mortality rates. Additionally, the fungus has become resistant to decontamination techniques and can survive for weeks in a hospital environment. Nanoparticles might be a good substitute to treat illnesses brought on by this newly discovered pathogen. Nanoparticles have become a trend and hot topic in recent years to combat this fatal fungus. This review gives a general insight into the epidemiology of C. auris and infection. It discusses the current conventional therapy and mechanism of resistance development. Furthermore, it focuses on nanoparticles, their different types, and up-to-date trials to evaluate the promising efficacy of nanoparticles with respect to C. auris.
Collapse
Affiliation(s)
- Helal F. Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt;
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt;
| | - Israa M. S. Al-Kadmy
- Branch of Biotechnology, Department of Biology, College of Science, Mustansiriyah University, Baghdad P.O. Box 10244, Iraq;
| | - Noura H. Abd Ellah
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt;
- Department of Pharmaceutics, Faculty of Pharmacy, Badr University in Assiut, Naser City, Assiut 2014101, Egypt
| | - Lama Shbibe
- Faculty of Science, Damascus University, Damascus 97009, Syria;
| | - Basem Battah
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Syrian Private University (SPU), Daraa International Highway, Damascus 36822, Syria
| |
Collapse
|
38
|
Smith DJ, Gold JAW, Benedict K, Wu K, Lyman M, Jordan A, Medina N, Lockhart SR, Sexton DJ, Chow NA, Jackson BR, Litvintseva AP, Toda M, Chiller T. Public Health Research Priorities for Fungal Diseases: A Multidisciplinary Approach to Save Lives. J Fungi (Basel) 2023; 9:820. [PMID: 37623591 PMCID: PMC10455901 DOI: 10.3390/jof9080820] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Fungal infections can cause severe disease and death and impose a substantial economic burden on healthcare systems. Public health research requires a multidisciplinary approach and is essential to help save lives and prevent disability from fungal diseases. In this manuscript, we outline the main public health research priorities for fungal diseases, including the measurement of the fungal disease burden and distribution and the need for improved diagnostics, therapeutics, and vaccines. Characterizing the public health, economic, health system, and individual burden caused by fungal diseases can provide critical insights to promote better prevention and treatment. The development and validation of fungal diagnostic tests that are rapid, accurate, and cost-effective can improve testing practices. Understanding best practices for antifungal prophylaxis can optimize prevention in at-risk populations, while research on antifungal resistance can improve patient outcomes. Investment in vaccines may eliminate certain fungal diseases or lower incidence and mortality. Public health research priorities and approaches may vary by fungal pathogen.
Collapse
Affiliation(s)
- Dallas J. Smith
- Mycotic Diseases Branch, Division of Foodborne, Waterborne and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (J.A.W.G.); (K.B.); (K.W.); (M.L.); (A.J.); (N.M.); (S.R.L.); (D.J.S.); (N.A.C.); (B.R.J.); (A.P.L.); (M.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Tom Chiller
- Mycotic Diseases Branch, Division of Foodborne, Waterborne and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (J.A.W.G.); (K.B.); (K.W.); (M.L.); (A.J.); (N.M.); (S.R.L.); (D.J.S.); (N.A.C.); (B.R.J.); (A.P.L.); (M.T.)
| |
Collapse
|
39
|
Pechacek J, Lionakis MS. Host defense mechanisms against Candida auris. Expert Rev Anti Infect Ther 2023; 21:1087-1096. [PMID: 37753840 DOI: 10.1080/14787210.2023.2264500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/25/2023] [Indexed: 09/28/2023]
Abstract
INTRODUCTION Candida auris is a pathogen of growing public health concern given its rapid spread across the globe, its propensity for long-term skin colonization and healthcare-related outbreaks, its resistance to a variety of antifungal medications, and the high morbidity and mortality associated with invasive disease. Despite that, the host immune response mechanisms that operate during C. auris skin colonization and invasive infection remains poorly understood. AREAS COVERED In this manuscript, we review the available literature in the growing research field pertaining to C. auris host defenses and we discuss what is known about the ability of C. auris to thrive on mammalian skin, the role of lymphoid cell-mediated, IL-17-dependent defenses in controlling cutaneous colonization, and the contribution of myeloid phagocytes in curtailing systemic infection. EXPERT OPINION Understanding the mechanisms by which the host immune system responds to and controls colonization and infection with C. auris and developing a deeper knowledge of tissue-specific host-C. auris interactions and of C. auris immune-evading mechanisms may help devise improved strategies for decolonization, prognostication, prevention, vaccination, and/or directed antifungal treatment in vulnerable patient populations.
Collapse
Affiliation(s)
- Joseph Pechacek
- From the Fungal Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michail S Lionakis
- From the Fungal Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
40
|
Elshaer M, Herrada J, Gamal A, McCormick TS, Ghannoum M. Efficacy of chlorhexidine in advanced performance technology formulation in decolonizing the skin using Candida auris skin colonization mouse model. Am J Infect Control 2023; 51:836-837. [PMID: 36417953 PMCID: PMC10199144 DOI: 10.1016/j.ajic.2022.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
The incidence of Candida auris, an emerging multidrug resistant fungal species, is increasing. The ability of this yeast to colonize the human skin could lead to infections. Identifying agents to reduce the skin fungal burden is critical. Chlorhexidine formulated in a new Advanced Performance Technology formulation (APT-CH) was significantly more effective than untreated controls. Additional studies are warranted.
Collapse
Affiliation(s)
- Mohammed Elshaer
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Dermatology, Case Western Reserve University, Cleveland, OH
| | - Janet Herrada
- Department of Dermatology, Case Western Reserve University, Cleveland, OH
| | - Ahmed Gamal
- Department of Dermatology, Case Western Reserve University, Cleveland, OH
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, OH
| | - Mahmoud Ghannoum
- Department of Dermatology, Case Western Reserve University, Cleveland, OH.
| |
Collapse
|
41
|
Spoladori LFDA, Andriani GM, Castro IMD, Suzukawa HT, Gimenes ACR, Bartolomeu-Gonçalves G, Ishida K, Nakazato G, Pinge-Filho P, Machado RRB, Nakamura CV, Andrade G, Tavares ER, Yamauchi LM, Yamada-Ogatta SF. Synergistic Antifungal Interaction between Pseudomonas aeruginosa LV Strain Metabolites and Biogenic Silver Nanoparticles against Candida auris. Antibiotics (Basel) 2023; 12:antibiotics12050861. [PMID: 37237764 DOI: 10.3390/antibiotics12050861] [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: 03/30/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Candida auris has been found to be a persistent colonizer of human skin and a successful pathogen capable of causing potentially fatal infection, especially in immunocompromised individuals. This fungal species is usually resistant to most antifungal agents and has the ability to form biofilms on different surfaces, representing a significant therapeutic challenge. Herein, the effect of metabolites of Pseudomonas aeruginosa LV strain, alone and combined with biologically synthesized silver nanoparticles (bioAgNP), was evaluated in planktonic and sessile (biofilm) cells of C. auris. First, the minimal inhibitory and fungicidal concentration values of 3.12 and 6.25 μg/mL, respectively, were determined for F4a, a semi-purified bacterial fraction. Fluopsin C and indolin-3-one seem to be the active components of F4a. Like the semi-purified fraction, they showed a time- and dose-dependent fungicidal activity. F4a and bioAgNP caused severe changes in the morphology and ultrastructure of fungal cells. F4a and indolin-3-one combined with bioAgNP exhibited synergistic fungicidal activity against planktonic cells. F4a, alone or combined with bioAgNP, also caused a significant decrease in the number of viable cells within the biofilms. No cytotoxicity to mammalian cells was detected for bacterial metabolites combined with bioAgNP at synergistic concentrations that presented antifungal activity. These results indicate the potential of F4a combined with bioAgNP as a new strategy for controlling C. auris infections.
Collapse
Affiliation(s)
| | - Gabriella Maria Andriani
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Isabela Madeira de Castro
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Helena Tiemi Suzukawa
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Ana Carolina Ramos Gimenes
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Guilherme Bartolomeu-Gonçalves
- Programa de Pós-Graduação em Fisiopatologia Clínica e Laboratorial, Universidade Estadual de Londrina, Londrina CEP 86038-350, Brazil
| | - Kelly Ishida
- Laboratório de Quimioterapia Antifúngica, Universidade de São Paulo, São Paulo CEP 05508-000, Brazil
| | - Gerson Nakazato
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Laboratório de Bacteriologia Básica e Aplicada, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Phileno Pinge-Filho
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Laboratório de Imunopatologia Experimental, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Rayanne Regina Beltrame Machado
- Laboratório de Inovação Tecnológica no Desenvolvimento de Fármacos e Cosméticos, Universidade Estadual de Maringá, Maringá CEP 87020-900, Brazil
| | - Celso Vataru Nakamura
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Laboratório de Inovação Tecnológica no Desenvolvimento de Fármacos e Cosméticos, Universidade Estadual de Maringá, Maringá CEP 87020-900, Brazil
| | - Galdino Andrade
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Laboratório de Ecologia Microbiana, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Eliandro Reis Tavares
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Lucy Megumi Yamauchi
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
| | - Sueli Fumie Yamada-Ogatta
- Programa de Pós-Graduação em Microbiologia, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina CEP 86057-970, Brazil
- Programa de Pós-Graduação em Fisiopatologia Clínica e Laboratorial, Universidade Estadual de Londrina, Londrina CEP 86038-350, Brazil
| |
Collapse
|
42
|
Proctor DM, Drummond RA, Lionakis MS, Segre JA. One population, multiple lifestyles: Commensalism and pathogenesis in the human mycobiome. Cell Host Microbe 2023; 31:539-553. [PMID: 37054674 PMCID: PMC10155287 DOI: 10.1016/j.chom.2023.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/12/2023] [Accepted: 02/23/2023] [Indexed: 04/15/2023]
Abstract
Candida auris and Candida albicans can result in invasive fungal diseases. And yet, these species can stably and asymptomatically colonize human skin and gastrointestinal tracts. To consider these disparate microbial lifestyles, we first review factors shown to influence the underlying microbiome. Structured by the damage response framework, we then consider the molecular mechanisms deployed by C. albicans to switch between commensal and pathogenic lifestyles. Next, we explore this framework with C. auris to highlight how host physiology, immunity, and/or antibiotic receipt are associated with progression from colonization to infection. While treatment with antibiotics increases the risk that an individual will succumb to invasive candidiasis, the underlying mechanisms remain unclear. Here, we describe several hypotheses that may explain this phenomenon. We conclude by highlighting future directions integrating genomics with immunology to advance our understanding of invasive candidiasis and human fungal disease.
Collapse
Affiliation(s)
- Diana M Proctor
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Rebecca A Drummond
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham B15 2TT, UK
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Julia A Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Akinbobola AB, Kean R, Hanifi SMA, Quilliam RS. Environmental reservoirs of the drug-resistant pathogenic yeast Candida auris. PLoS Pathog 2023; 19:e1011268. [PMID: 37053164 PMCID: PMC10101498 DOI: 10.1371/journal.ppat.1011268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Candia auris is an emerging human pathogenic yeast; yet, despite phenotypic attributes and genomic evidence suggesting that it probably emerged from a natural reservoir, we know nothing about the environmental phase of its life cycle and the transmission pathways associated with it. The thermotolerant characteristics of C. auris have been hypothesised to be an environmental adaptation to increasing temperatures due to global warming (which may have facilitated its ability to tolerate the mammalian thermal barrier that is considered a protective strategy for humans against colonisation by environmental fungi with pathogenic potential). Thus, C. auris may be the first human pathogenic fungus to have emerged as a result of climate change. In addition, the release of antifungal chemicals, such as azoles, into the environment (from both pharmaceutical and agricultural sources) is likely to be responsible for the environmental enrichment of resistant strains of C. auris; however, the survival and dissemination of C. auris in the natural environment is poorly understood. In this paper, we critically review the possible pathways through which C. auris can be introduced into the environment and evaluate the environmental characteristics that can influence its persistence and transmission in natural environments. Identifying potential environmental niches and reservoirs of C. auris and understanding its emergence against a backdrop of climate change and environmental pollution will be crucial for the development of effective epidemiological and environmental management responses.
Collapse
Affiliation(s)
- Ayorinde B Akinbobola
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, United Kingdom
| | - Ryan Kean
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Syed Manzoor Ahmed Hanifi
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Health System and Population Studies Division, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, Bangladesh
| | - Richard S Quilliam
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, United Kingdom
| |
Collapse
|
44
|
Alvarez-Moreno CA, Morales-López S, Rodriguez GJ, Rodriguez JY, Robert E, Picot C, Ceballos-Garzon A, Parra-Giraldo CM, Le Pape P. The Mortality Attributable to Candidemia in C. auris Is Higher than That in Other Candida Species: Myth or Reality? J Fungi (Basel) 2023; 9:jof9040430. [PMID: 37108885 PMCID: PMC10143486 DOI: 10.3390/jof9040430] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Candida auris has become a major health threat due to its transmissibility, multidrug resistance and severe outcomes. In a case-control design, 74 hospitalised patients with candidemia were enrolled. In total, 22 cases (29.7%) and 52 controls (C. albicans, 21.6%; C. parapsilosis, 21.6%; C. tropicalis, 21.6%; C. glabrata, 1.4%) were included and analysed in this study. Risk factors, clinical and microbiological characteristics and outcomes of patients with C. auris and non-auris Candida species (NACS) candidemia were compared. Previous fluconazole exposure was significantly higher in C. auris candidemia patients (OR 3.3; 1.15–9.5). Most C. auris isolates were resistant to fluconazole (86.3%) and amphotericin B (59%) whilst NACS isolates were generally susceptible. No isolates resistant to echinocandins were detected. The average time to start antifungal therapy was 3.6 days. Sixty-three (85.1%) patients received adequate antifungal therapy, without significant differences between the two groups. The crude mortality at 30 and 90 days of candidemia was up to 37.8% and 40.5%, respectively. However, there was no difference in mortality both at 30 and 90 days between the group with candidemia by C. auris (31.8%) and by NACS (42.3%) (OR 0.6; 95% IC 0.24–1.97) and 36.4% and 42.3% (0.77; 0.27–2.1), respectively. In this study, mortality due to candidemia between C. auris and NACS was similar. Appropriate antifungal therapy in both groups may have contributed to finding no differences in outcomes.
Collapse
Affiliation(s)
- Carlos A. Alvarez-Moreno
- Facultad de Medicina, Universidad Nacional de Colombia, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
- Correspondence: ; Tel.: +57-31-4330-2367
| | - Soraya Morales-López
- Grupo CINBIOS, Programa de Microbiología, Universidad Popular del Cesar, Valledupar 200004, Colombia
| | - Gerson J. Rodriguez
- Centro de Investigaciones Microbiológicas del Cesar (CIMCE), Valledupar 200002, Colombia
| | - Jose Y. Rodriguez
- Centro de Investigaciones Microbiológicas del Cesar (CIMCE), Valledupar 200002, Colombia
| | - Estelle Robert
- Cibles et Médicaments des Infections et de l’Immunité, Nantes Université, CHU de Nantes, IICiMed, 10 UR1155, 44000 Nantes, France
| | - Carine Picot
- Cibles et Médicaments des Infections et de l’Immunité, Nantes Université, CHU de Nantes, IICiMed, 10 UR1155, 44000 Nantes, France
| | - Andrés Ceballos-Garzon
- Cibles et Médicaments des Infections et de l’Immunité, Nantes Université, CHU de Nantes, IICiMed, 10 UR1155, 44000 Nantes, France
- Unidad de Investigacion en Proteómica y Micosis Humanas, Grupo de investigacion en Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Claudia M. Parra-Giraldo
- Unidad de Investigacion en Proteómica y Micosis Humanas, Grupo de investigacion en Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Patrice Le Pape
- Cibles et Médicaments des Infections et de l’Immunité, Nantes Université, CHU de Nantes, IICiMed, 10 UR1155, 44000 Nantes, France
| |
Collapse
|
45
|
Khari A, Biswas B, Gangwar G, Thakur A, Puria R. Candida auris biofilm: a review on model to mechanism conservation. Expert Rev Anti Infect Ther 2023; 21:295-308. [PMID: 36755419 DOI: 10.1080/14787210.2023.2179036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
INTRODUCTION Candida auris is included in the fungal infection category 'critical' by WHO because of associated high drug tolerance and spread at an alarming rate which if remains untouched may result in serious outbreaks. Since its discovery in 2009, several assiduous efforts by mycologists across the world have deciphered its biology including growth physiology, drug tolerance, biofilm formation, etc. The differential response of various strains from different clades poses a hurdle in drawing a final conclusion. AREAS COVERED This review provides brief insights into the understanding of C. auris biofilm. It includes information on various models developed to understand the biofilms and conservation of different signaling pathways. Significant development has been made in the recent past with the generation of relevant in vivo and ex vivo models. The role of signaling pathways in the development of biofilm is largely unknown. EXPERT OPINION The selection of an appropriate model system is a must for the accuracy and reproducibility of results. The conservation of major signaling pathways in C. auris with respect to C. albicans and S. cerevisiae highlights that initial inputs acquired from orthologs will be valuable in getting insights into the mechanism of biofilm formation and associated pathogenesis.
Collapse
Affiliation(s)
- Arsha Khari
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | | | | | - Anil Thakur
- Regional Centre for Biotechnology, Faridabad, India
| | - Rekha Puria
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| |
Collapse
|
46
|
Chen X, Jia X, Bing J, Zhang H, Hong N, Liu Y, Xi H, Wang W, Liu Z, Zhang Q, Li L, Kang M, Xiao Y, Yang B, Lin Y, Xu H, Fan X, Huang J, Gong J, Xu J, Xie X, Yang W, Zhang G, Zhang J, Kang W, Wang H, Hou X, Xiao M, Xu Y. Clonal Dissemination of Antifungal-Resistant Candida haemulonii, China. Emerg Infect Dis 2023; 29:576-584. [PMID: 36823029 PMCID: PMC9973686 DOI: 10.3201/eid2903.221082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Candida haemulonii, a relative of C. auris, frequently shows antifungal resistance and is transmissible. However, molecular tools for genotyping and investigating outbreaks are not yet established. We performed genome-based population analysis on 94 C. haemulonii strains, including 58 isolates from China and 36 other published strains. Phylogenetic analysis revealed that C. haemulonii can be divided into 4 clades. Clade 1 comprised strains from China and other global strains; clades 2-4 contained only isolates from China, were more recently evolved, and showed higher antifungal resistance. Four regional epidemic clusters (A, B, C, and D) were identified in China, each comprising ≥5 cases (largest intracluster pairwise single-nucleotide polymorphism differences <50 bp). Cluster A was identified in 2 hospitals located in the same city, suggesting potential intracity transmissions. Cluster D was resistant to 3 classes of antifungals. The emergence of more resistant phylogenetic clades and regional dissemination of antifungal-resistant C. haemulonii warrants further monitoring.
Collapse
|
47
|
Swaney MH, Nelsen A, Sandstrom S, Kalan LR. Sweat and Sebum Preferences of the Human Skin Microbiota. Microbiol Spectr 2023; 11:e0418022. [PMID: 36602383 PMCID: PMC9927561 DOI: 10.1128/spectrum.04180-22] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
The microorganisms inhabiting human skin must overcome numerous challenges that typically impede microbial growth, including low pH, osmotic pressure, and low nutrient availability. Yet the skin microbiota thrive on the skin and have adapted to these stressful conditions. The limited nutrients available for microbial use in this unique niche include those from host-derived sweat, sebum, and corneocytes. Here, we have developed physiologically relevant, synthetic skin-like growth media composed of compounds present in sweat and sebum. We find that skin-associated bacterial species exhibit unique growth profiles at different concentrations of artificial sweat and sebum. Most strains evaluated demonstrate a preference for high sweat concentrations, while the sebum preference is highly variable, suggesting that the capacity for sebum utilization may be a driver of the skin microbial community structure. In particular, the prominent skin commensal Staphylococcus epidermidis exhibits the strongest preference for sweat while growing equally well across sebum concentrations. Conversely, the growth of Corynebacterium kefirresidentii, another dominant skin microbiome member, is dependent on increasing concentrations of both sweat and sebum but only when sebum is available, suggesting a lipid requirement of this species. Furthermore, we observe that strains with similar growth profiles in the artificial media cluster by phylum, suggesting that phylogeny is a key factor in sweat and sebum use. Importantly, these findings provide an experimental rationale for why different skin microenvironments harbor distinct microbiome communities. In all, our study further emphasizes the importance of studying microorganisms in an ecologically relevant context, which is critical for our understanding of their physiology, ecology, and function on the skin. IMPORTANCE The human skin microbiome is adapted to survive and thrive in the harsh environment of the skin, which is low in nutrient availability. To study skin microorganisms in a system that mimics the natural skin environment, we developed and tested a physiologically relevant, synthetic skin-like growth medium that is composed of compounds found in the human skin secretions sweat and sebum. We find that most skin-associated bacterial species tested prefer high concentrations of artificial sweat but that artificial sebum concentration preference varies from species to species, suggesting that sebum utilization may be an important contributor to skin microbiome composition. This study demonstrates the utility of a skin-like growth medium, which can be applied to diverse microbiological systems, and underscores the importance of studying microorganisms in an ecologically relevant context.
Collapse
Affiliation(s)
- Mary Hannah Swaney
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Amanda Nelsen
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Shelby Sandstrom
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Lindsay R. Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
- M. G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
48
|
Junqueira JC, Mylonakis E. Editorial: Candida biofilms. Front Microbiol 2023; 13:1128600. [PMID: 36687614 PMCID: PMC9846752 DOI: 10.3389/fmicb.2022.1128600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil,*Correspondence: Juliana Campos Junqueira ✉
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States,Eleftherios Mylonakis ✉
| |
Collapse
|
49
|
Extracellular Vesicles Contribute to Mixed-Fungal Species Competition during Biofilm Initiation. mBio 2022; 13:e0298822. [PMID: 36377868 PMCID: PMC9765065 DOI: 10.1128/mbio.02988-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles commonly modulate interactions among cellular communities. Recent studies demonstrate that biofilm maturation features, including matrix production, drug resistance, and dispersion, require the delivery of a core protein and carbohydrate vesicle cargo in Candida species. The function of the vesicle cargo for these advanced-phase biofilm characteristics appears to be conserved across Candida species. Mixed-species interactions in mature biofilms indicate that vesicle cargo serves a cooperative role in preserving the community. Here, we define the function of biofilm-associated vesicles for biofilm initiation both within and among five species across the Candida genus. We found similar vesicle cargo functions for several conserved proteins across species, based on the behavior of mutants. Repletion of the adhesion environment with wild-type vesicles returned the community phenotype toward reference levels in intraspecies experiments. However, cross-species vesicle complementation did not restore the wild-type biology and in fact drove the phenotype in the opposite direction for most cross-species interactions. Further study of mixed-species biofilm adhesion and exogenous wild-type vesicle administration similarly demonstrated competitive interactions. Our studies indicate that similar vesicle cargoes contribute to biofilm initiation. However, vesicles from disparate species serve an interference competitive role in mixed-Candida species scenarios. IMPORTANCE Candida species commonly form mixed-species biofilms with other Candida species and bacteria. In the established biofilm state, vesicle cargo delivers public goods to support the mature community. At biofilm initiation, however, vesicles play a negative role in cross-species interactions, presumably to allow species to gain a survival advantage. These observations and recent reports reveal that vesicle cargo has both cooperative and competitive roles among Candida species, depending on the needs of the community biofilm formation.
Collapse
|
50
|
Nguyen UT, Kalan LR. Forgotten fungi: the importance of the skin mycobiome. Curr Opin Microbiol 2022; 70:102235. [PMID: 36372041 PMCID: PMC10044452 DOI: 10.1016/j.mib.2022.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/21/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022]
Abstract
The mosaic ecosystems of microbes that live on our skin encompass not only bacteria but also fungi, microeukaryotes, and viruses. As the second most prevalent group, unique fungal communities are found across the dry, moist, and oily microenvironments of human skin, and alterations of these communities are largely driven by changes in skin physiology throughout an individual's lifespan. Fungi have also been associated with infection and dermatological disorders, resulting from the disrupted balance between fungal-bacterial networks on the skin. Mechanisms of colonization resistance toward fungi in the skin microbiome of animals have advanced our understanding in conservation strategies, yet in the human skin, the fungal microbiome (mycobiome) remains vastly unexplored. Here, we review recent studies on the role of fungi in the skin microbiome, emphasizing how fungal-bacterial interactions at the skin surface play an important ecological function in vertebrate hosts.
Collapse
Affiliation(s)
- Uyen Thy Nguyen
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, United States; Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States.
| | - Lindsay R Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States; Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|