1
|
Sørensen S, Kvich L, Xu Y, Thomsen TR, Bjarnsholt T, Thaarup I. Development of a tri-species wound model for studying fungal-bacterial interactions and antimicrobial therapies. Biofilm 2025; 9:100256. [PMID: 39927095 PMCID: PMC11804781 DOI: 10.1016/j.bioflm.2025.100256] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
Chronic wounds are increasing in numbers and biofilm-producing bacteria are highly prevalent in these wounds and often create resilient polymicrobial infections. Moreover, estimates suggest that up to 23 % of wounds contain fungi, particularly Candida albicans. Currently, inter-kingdom chronic wound models are scarce; thus, this study presents one of the few in vitro models that incorporate both bacterial and fungal species in a wound-relevant environment, addressing a critical gap in current biofilm research. The newly developed model contained the commonly isolated wound bacteria Pseudomonas aeruginosa and Staphylococcus aureus, and the fungus Candida albicans. Inter-species interactions were investigated through selective plate counting and pH and oxygen measurements, as well as confocal microscopy. Investigations were carried out before and after exposure to commonly used clinical antimicrobial treatments, including silver-infused bandages. When grown in a tri-species consortium, P. aeruginosa and S. aureus exhibited a higher tolerance towards silver-infused bandages than when they were grown individually. This suggests that C. albicans plays a protective role for the bacteria. In addition, the treatment also caused a shift in species ratios, moving from a P. aeruginosa-dominated consortium to a S. aureus-dominated consortium. Moreover, confocal microscopy revealed a change in biofilm architecture when comparing single-species models to tri-species models. Finally, we observed that silver-infused bandages increased the pH in the tri-species model as well as partially restoring the oxygenation within the wound model. In conclusion, our novel model exemplifies how inter-kingdom interactions in fungal-bacterial infections can complicate both the microenvironment and treatment efficacy.
Collapse
Affiliation(s)
- Stine Sørensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Kvich
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Yijuan Xu
- SEGES Innovation P/S, Aarhus, Denmark
| | - Trine R. Thomsen
- Department of Chemistry and Biotechnology, Aalborg University, Aalborg, Denmark
- Danish Technology Institute, Aarhus, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ida Thaarup
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Seal S, Basu DN, Ghosh K, Ramachandran A, Kutum R, Shelke T, Gupta I, Khan I. Pathogen growth and virulence dynamics drive the host evolution against coinfections. Proc Natl Acad Sci U S A 2025; 122:e2412124122. [PMID: 40267133 PMCID: PMC12054814 DOI: 10.1073/pnas.2412124122] [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: 06/24/2024] [Accepted: 03/22/2025] [Indexed: 04/25/2025] Open
Abstract
The occurrence of coinfections, where hosts are simultaneously infected by multiple pathogens, is widespread in nature and has significant negative impacts on global health. In humans, over one-sixth of the world's population is affected by coinfections, contributing to several diseases. However, despite the broad ecological relevance and impact on global health, most biomedical research has focused on understanding interactions between a single host and a single pathogen. The extent to which coinfections could impact host adaptation and immune system evolution, particularly in comparison to infections by single pathogens, thus remains largely unknown. Also, what roles do individual pathogen species play in this evolutionary process? To address these questions, in this study, we combined theoretical modeling and experimental validation in a model insect Tribolium castaneum evolving against two coinfecting bacterial pathogens with contrasting growth (e.g., fast- vs slow-growing) and virulence (fast- vs slow-killing) dynamics. Our findings show that fast-growing pathogens causing rapid mortality surges (i.e., fast-acting) can effectively limit the host's adaptive success against coinfections. While hosts rapidly evolved better survival against slow-growing bacteria causing long-lasting infections, adaptation against coinfections was significantly delayed and resembled the slow rate of adaptation against fast-acting pathogens. Finally, RNAseq analyses revealed that the observed delay in adaptation was associated with the limited scopes for suitable immune modulations against fast-acting pathogens. They might also be costly and pleiotropic (e.g., phenoloxidase activity), posing challenges for further immunomodulation and slowing adaptation. Our study thus highlights how individual pathogens' growth and virulence dynamics critically regulate adaptive responses against coinfections.
Collapse
Affiliation(s)
- Srijan Seal
- Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana131029, India
| | - Dipendra Nath Basu
- Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana131029, India
| | - Kripanjali Ghosh
- Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana131029, India
| | - Aryan Ramachandran
- Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana131029, India
| | - Rintu Kutum
- Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana131029, India
| | - Triveni Shelke
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi110016, India
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi110016, India
| | - Imroze Khan
- Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana131029, India
| |
Collapse
|
3
|
Sampaio C, Pessan JP, Nunes GP, Magno MB, Maia LC, Exterkate R, Deng D, Monteiro DR. Are the counts of Streptococcus mutans and Staphylococcus aureus changed in complete denture wearers carrying denture stomatitis? A systematic review with meta-analyses. J Prosthet Dent 2025; 133:427-437. [PMID: 37080861 DOI: 10.1016/j.prosdent.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 04/22/2023]
Abstract
STATEMENT OF PROBLEM Despite the importance of Candida spp. on the etiology of denture stomatitis (DS), information on the role of the bacterial component is still scarce. PURPOSE The purpose of this systematic review was to evaluate whether the counts of Staphylococcus aureus and Streptococcus mutans were changed in complete denture wearers diagnosed with Candida-associated DS. MATERIAL AND METHODS The literature search was performed in 8 databases and by hand searching. The risk of bias was assessed according to the Newcastle-Ottawa qualifier. Meta-analyses were performed considering the microorganism evaluated (S. aureus or S. mutans) and the collection area (mucosa or dentures). The certainty of evidence was assessed according to the grading of recommendations assessment, development and evaluations (GRADE) criteria. RESULTS Participants with DS presented higher counts of S. aureus in the mucosa compared with those from the control group (OR, 3.16 [1.62, 6.15]; P<.001). No significant difference between the groups was observed for samples collected from dentures (OR, 0.73 [0.50, 1.07]; P=.110). Conversely, participants without DS presented higher counts of S. mutans both in the mucosa (OR, 0.19 [0.06, 0.63]; P=.006) and dentures (OR, 0.64 [0.41, 1.0]; P=.050). CONCLUSIONS Microbial counts in participants with DS changed as a function of the type of microorganism and collection site. The certainty of evidence ranged from very low to low. The findings reinforce the fact that bacteria also play a relevant role in DS and should be more extensively studied. Such information may be useful to guide further therapies to prevent or control DS.
Collapse
Affiliation(s)
- Caio Sampaio
- PhD, Department of Preventive and Restorative Dentistry, School of Dentistry, São Paulo State University (Unesp), Araçatuba, SP, Brazil
| | - Juliano P Pessan
- Associate Professor, Department of Preventive and Restorative Dentistry, School of Dentistry, São Paulo State University (Unesp), Araçatuba, SP, Brazil
| | - Gabriel P Nunes
- PhD student, Department of Preventive and Restorative Dentistry, School of Dentistry, São Paulo State University (Unesp), Araçatuba, SP, Brazil
| | - Marcela B Magno
- Postdoctoral Fellow, Department of Pediatric Dentistry, School of Dentistry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lucianne C Maia
- Professor, Department of Pediatric Dentistry, School of Dentistry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Rob Exterkate
- Research Staff Member, Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Dongmei Deng
- Associate Professor, Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Douglas R Monteiro
- Professor, Postgraduate Program in Health Sciences, School of Dentistry, University of Western São Paulo (UNOESTE), Presidente Prudente, SP, Brazil.
| |
Collapse
|
4
|
Hurley J. Structural Equation Modelling as a Proof-of-Concept Tool for Mediation Mechanisms Between Topical Antibiotic Prophylaxis and Six Types of Blood Stream Infection Among ICU Patients. Antibiotics (Basel) 2024; 13:1096. [PMID: 39596789 PMCID: PMC11591272 DOI: 10.3390/antibiotics13111096] [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: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Whether exposing the microbiome to antibiotics decreases or increases the risk of blood stream infection with Pseudomonas aeruginosa, Staphylococcus aureus, Acinetobacter, and Candida among ICU patients, and how this altered risk might be mediated, are critical research questions. Addressing these questions through the direct study of specific constituents within the microbiome would be difficult. An alternative tool for addressing these research questions is structural equation modelling (SEM). SEM enables competing theoretical causation networks to be tested 'en bloc' by confrontation with data derived from the literature. These causation models have three conceptual steps: exposure to specific antimicrobials are the key drivers, clinically relevant infection end points are the measurable observables, and the activity of key microbiome constituents on microbial invasion serve as mediators. These mediators, whether serving to promote, to impede, or neither, are typically unobservable and appear as latent variables in each model. SEM methods enable comparisons through confronting the three competing models, each versus clinically derived data with the various exposures, such as topical or parenteral antibiotic prophylaxis, factorized in each model. Candida colonization, represented as a latent variable, and concurrency are consistent promoters of all types of blood stream infection, and emerge as harmful mediators.
Collapse
Affiliation(s)
- James Hurley
- Melbourne Medical School, University of Melbourne, Parkville, VIC 3052, Australia;
- Ballarat Health Services, Grampians Health, Ballarat, VIC 3350, Australia;
- Ballarat Clinical School, Deakin University, Ballarat, VIC 3350, Australia
| |
Collapse
|
5
|
Arévalo-Jaimes BV, Torrents E. Candida albicans enhances Staphylococcus aureus virulence by progressive generation of new phenotypes. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100316. [PMID: 39649408 PMCID: PMC11621768 DOI: 10.1016/j.crmicr.2024.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
Candida albicans and Staphylococcus aureus have been co-isolated from several biofilm-associated diseases, including those related to medical devices. This association confers advantages to both microorganisms, resulting in detrimental effects on the host. To elucidate this phenomenon, the present study investigated colony changes derived from non-physical interactions between C. albicans and S. aureus. We performed proximity assays by confronting colonies of the yeast and the bacteria on agar plates at six different distances for 9-10 days. We found that colony variants of S. aureus originated progressively after prolonged exposure to C. albicans proximity, specifically in response to pH neutralization of the media by the fungi. The new phenotypes of S. aureus were more virulent in a Galleria mellonella larvae model compared to colonies grown without C. albicans influence. This event was associated with an upregulation of RNA III and agrA expression, suggesting a role for α-toxin. Our findings indicate that C. albicans enhances S. aureus virulence by inducing the formation of more aggressive colonies. This highlights the importance of understanding the intricate connection between environmental responses, virulence and, fitness in S. aureus pathogenesis.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac Street 10, 08037, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal Street 647, 08028, Barcelona, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac Street 10, 08037, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal Street 647, 08028, Barcelona, Spain
| |
Collapse
|
6
|
Kapitan M, Niemiec MJ, Millet N, Brandt P, Chowdhury MEK, Czapka A, Abdissa K, Hoffmann F, Lange A, Veleba M, Nietzsche S, Mosig AS, Löffler B, Marquet M, Makarewicz O, Kline KA, Vylkova S, Swidergall M, Jacobsen ID. Synergistic cross-kingdom host cell damage between Candida albicans and Enterococcus faecalis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612452. [PMID: 39314435 PMCID: PMC11419042 DOI: 10.1101/2024.09.11.612452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The fungus Candida albicans and the Gram-positive bacterium Enterococcus faecalis share mucosal niches in the human body. As opportunistic pathogens, both are found to expand population size during dysbiosis, and can cause severe systemic infections in susceptible individuals. Here, we show that the presence of C. albicans results in increased host cell damage by E. faecalis . Furthermore, E. faecalis aggravates oropharyngeal candidiasis in mice. Increased damage is mediated by enterococcal cytolysin, and involves both physical interaction and altered glucose availability. Physical interaction promotes accumulation of bacteria on host cells, facilitating contact of cytolysin with host cells. Glucose depletion by the metabolic activity of the fungus sensitized host cells to cytolysin. This work illustrates how a complex interplay between fungi and bacteria can result in detrimental consequences for the host.
Collapse
|
7
|
Predoi D, Iconaru SL, Ciobanu SC, Ţălu Ş, Predoi SA, Buton N, Ramos GQ, da Fonseca Filho HD, Matos RS. Synthesis, characterization, and antifungal properties of chrome-doped hydroxyapatite thin films. MATERIALS CHEMISTRY AND PHYSICS 2024; 324:129690. [DOI: 10.1016/j.matchemphys.2024.129690] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
|
8
|
Pasman R, Zhang J, Zaat SAJ, Brul S, Krom BP. A customizable and defined medium supporting culturing of Candida albicans, Staphylococcus aureus, and human oral epithelial cells. Appl Environ Microbiol 2024; 90:e0036024. [PMID: 39072650 PMCID: PMC11337806 DOI: 10.1128/aem.00360-24] [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: 02/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Candida albicans, an opportunistic oral pathogen, synergizes with Staphylococcus aureus, allowing bacteria to co-invade and systemically disseminate within the host. Studying human-microbe interactions creates the need for a universal culture medium that supports fungal, bacterial, and human cell culturing, while allowing sensitive analytical approaches such as OMICs and chromatography techniques. In this study, we established a fully defined, customizable adaptation of Dulbecco's modified Eagle medium (DMEM), allowing multi-kingdom culturing of S. aureus, C. albicans, and human oral cell lines, whereas minimal version of DMEM (mDMEM) did not support growth of S. aureus, and neither did supplementation with dextrose, MEM non-essential amino acids, pyruvate, and Glutamax. This new medium composition, designated as "mDMEM-DMP," promoted growth of all tested S. aureus strains. Addition of 25 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) further improved growth, while higher concentrations did not improve growth any further. Higher concentrations of HEPES did result in prolonged stabilization of medium pH. mDMEM-DMP promoted (hyphal) C. albicans monoculturing and co-culturing on both solid and semi-solid surfaces. In contrast to S. aureus, addition of HEPES reduced C. albicans maximum culture optical density (OD). Finally, only buffered mDMEM-DMP (100 mM HEPES) was successful in maintaining the metabolic activity of human oral Ca9-22 and HO1N1 cell lines for 24 hours. Altogether, our findings show that mDMEM-DMP is a versatile and potent culture medium for both microbial and human cell culturing, providing a customizable platform to study human as well as microbial molecular physiology and putative interactions. IMPORTANCE Interaction between microbes and the host are in the center of interest both in disease and in health. In order to study the interactions between microbes of different kingdoms and the host, alternative media are required. Synthetic media are useful as they allow addition of specific components. In addition, well-defined media are required if high-resolution analyses such as metabolomics and proteomics are desired. We describe the development of a synthetic medium to study the interactions between C. albicans, S. aureus, and human oral epithelial cells. Our findings show that mDMEM-DMP is a versatile and potent culture medium for both microbial and human cell culturing, providing a customizable platform to study human as well as microbial molecular physiology and putative interactions.
Collapse
Affiliation(s)
- Raymond Pasman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Jianbo Zhang
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Sebastian A. J. Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Stanley Brul
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Free University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Jaswal K, Todd OA, Flores Audelo RC, Santus W, Paul S, Singh M, Miao J, Underhill DM, Peters BM, Behnsen J. Commensal Yeast Promotes Salmonella Typhimurium Virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606421. [PMID: 39211098 PMCID: PMC11360897 DOI: 10.1101/2024.08.08.606421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Enteric pathogens engage in complex interactions with the host and the resident microbiota to establish gut colonization. Although mechanistic interactions between enteric pathogens and bacterial commensals have been extensively studied, whether and how commensal fungi affect pathogenesis of enteric infections remains largely unknown. Here we show that colonization with the common human gut commensal fungus Candida albicans worsened infections with the enteric pathogen Salmonella enterica serovar Typhimurium. Presence of C. albicans in the mouse gut increased Salmonella cecum colonization and systemic dissemination. We investigated the underlying mechanism and found that Salmonella binds to C. albicans via Type 1 fimbriae and uses its Type 3 Secretion System (T3SS) to deliver effector proteins into C. albicans . A specific effector, SopB, was sufficient to manipulate C. albicans metabolism, triggering increased arginine biosynthesis in C. albicans and the release of millimolar amounts of arginine into the extracellular environment. The released arginine, in turn, induced T3SS expression in Salmonella , increasing its invasion of epithelial cells. C. albicans deficient in arginine production was unable to increase Salmonella virulence in vitro or in vivo . In addition to modulating pathogen invasion, arginine also directly influenced the host response to infection. Arginine-producing C. albicans dampened the inflammatory response during Salmonella infection, whereas C. albicans deficient in arginine production did not. Arginine supplementation in the absence of C. albicans increased the systemic spread of Salmonella and decreased the inflammatory response, phenocopying the presence of C. albicans . In summary, we identified C. albicans colonization as a susceptibility factor for disseminated Salmonella infection, and arginine as a central metabolite in the cross-kingdom interaction between fungi, bacteria, and host.
Collapse
|
10
|
Elnagar RM. Cross interaction between bacterial and fungal microbiota and their relevance to human health and disease: mechanistic pathways and prospective therapy. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:309-320. [PMID: 39364131 PMCID: PMC11444862 DOI: 10.12938/bmfh.2024-031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/27/2024] [Indexed: 10/05/2024]
Abstract
Diverse bacterial and fungal microbiota communities inhabit the human body, and their presence is essential for maintaining host homeostasis. The oral cavity, lung, gut, and vagina are just a few of the bodily cavities where these microorganisms communicate with one another, either directly or indirectly. The effects of this interaction can be either useful or detrimental to the host. When the healthy microbial diversity is disturbed, for instance, as a result of prolonged treatment with broad spectrum antibiotics, this allows the growth of specific microbes at the expense of others and alters their pathogenicity, causing a switch of commensal germs into pathogenic germs, which could promote tissue invasion and damage, as occurs in immunocompromised patients. Consequently, antimicrobials that specifically target pathogens may help in minimizing secondary issues that result from the disruption of useful bacterial/fungal interactions (BFIs). The interface between Candida albicans and Aspergillus fumigatus with bacteria at various body sites is emphasized in the majority of the medically important BFIs that have been reported thus far. This interface either supports or inhibits growth, or it enhances or blocks the generation of virulence factors. The aim of this review is to draw attention to the link between the bacterial and fungal microbiota and how they contribute to both normal homeostasis and disease development. Additionally, recent research that has studied microbiota as novel antimicrobials is summarized.
Collapse
Affiliation(s)
- Rasha Mokhtar Elnagar
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
11
|
Paul S, Todd OA, Eichelberger KR, Tkaczyk C, Sellman BR, Noverr MC, Cassat JE, Fidel PL, Peters BM. A fungal metabolic regulator underlies infectious synergism during Candida albicans-Staphylococcus aureus intra-abdominal co-infection. Nat Commun 2024; 15:5746. [PMID: 38982056 PMCID: PMC11233573 DOI: 10.1038/s41467-024-50058-w] [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: 02/16/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.
Collapse
Affiliation(s)
- Saikat Paul
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Olivia A Todd
- Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine Tkaczyk
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, USA
| | - Bret R Sellman
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, USA
| | - Mairi C Noverr
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul L Fidel
- Department of Oral and Craniofacial Biology, Louisiana State University Health - School of Dentistry, New Orleans, LA, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
12
|
Xu Z, Li Y, Xu A, Xue L, Soteyome T, Yuan L, Ma Q, Seneviratne G, Hong W, Mao Y, Kjellerup BV, Liu J. Differential alteration in Lactiplantibacillus plantarum subsp. plantarum quorum-sensing systems and reduced Candida albicans yeast survival and virulence gene expression in dual-species interaction. Microbiol Spectr 2024; 12:e0035324. [PMID: 38717160 PMCID: PMC11237386 DOI: 10.1128/spectrum.00353-24] [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: 02/08/2024] [Accepted: 04/15/2024] [Indexed: 06/06/2024] Open
Abstract
Candida albicans (C. albicans) and Lactiplantibacillus plantarum subsp. plantarum (L. plantarum) are frequently identified in various niches, but their dual-species interaction, especially with C. albicans in yeast form, remains unclear. This study aimed to investigate the dual-species interaction of L. plantarum and C. albicans, including proliferation, morphology, and transcriptomes examined by selective agar plate counting, microscopy, and polymicrobial RNA-seq, respectively. Maintaining a stable and unchanged growth rate, L. plantarum inhibited C. albicans yeast cell proliferation but not hyphal growth. Combining optical microscopy and atomic force microscopy, cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed during dual-species interaction. Reduced C. albicans yeast cell proliferation in mixed culture was partially due to L. plantarum cell-free culture supernatant but not the acidic environment. Upon polymicrobial transcriptomics analysis, interesting changes were identified in both L. plantarum and C. albicans gene expression. First, two L. plantarum quorum-sensing systems showed contrary changes, with the activation of lamBDCA and repression of luxS. Second, the upregulation of stress response-related genes and downregulation of cell cycle, cell survival, and cell integrity-related pathways were identified in C. albicans, possibly connected to the stress posed by L. plantarum and the reduced yeast cell proliferation. Third, a large scale of pathogenesis and virulence factors were downregulated in C. albicans, indicating the potential interruption of pathogenic activities by L. plantarum. Fourth, partial metabolism and transport pathways were changed in L. plantarum and C. albicans. The information in this study might aid in understanding the behavior of L. plantarum and C. albicans in dual-species interaction.IMPORTANCEThe anti-Candida albicans activity of Lactiplantibacillus plantarum has been explored in the past decades. However, the importance of C. albicans yeast form and the effect of C. albicans on L. plantarum had also been omitted. In this study, the dual-species interaction of L. plantarum and C. albicans was investigated with a focus on the transcriptomes. Cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed. Upon polymicrobial transcriptomics analysis, interesting changes were identified, including contrary changes in two L. plantarum quorum-sensing systems and reduced cell survival-related pathways and pathogenesis determinants in C. albicans.
Collapse
Affiliation(s)
- Zhenbo Xu
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yaqin Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou, China
| | - Liang Xue
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, China, Guangzhou, Guangdong
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qin Ma
- Key Laboratory of Functional Foods, Ministry of Agriculture, Guangdong Key Laboratory of Agricultural Products Processing, Sericultural and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | | | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuzhu Mao
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Birthe V. Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Junyan Liu
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, College of Light Industry and Food Science, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
13
|
Bényei ÉB, Nazeer RR, Askenasy I, Mancini L, Ho PM, Sivarajan GAC, Swain JEV, Welch M. The past, present and future of polymicrobial infection research: Modelling, eavesdropping, terraforming and other stories. Adv Microb Physiol 2024; 85:259-323. [PMID: 39059822 DOI: 10.1016/bs.ampbs.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Over the last two centuries, great advances have been made in microbiology as a discipline. Much of this progress has come about as a consequence of studying the growth and physiology of individual microbial species in well-defined laboratory media; so-called "axenic growth". However, in the real world, microbes rarely live in such "splendid isolation" (to paraphrase Foster) and more often-than-not, share the niche with a plethora of co-habitants. The resulting interactions between species (and even between kingdoms) are only very poorly understood, both on a theoretical and experimental level. Nevertheless, the last few years have seen significant progress, and in this review, we assess the importance of polymicrobial infections, and show how improved experimental traction is advancing our understanding of these. A particular focus is on developments that are allowing us to capture the key features of polymicrobial infection scenarios, especially as those associated with the human airways (both healthy and diseased).
Collapse
Affiliation(s)
| | | | - Isabel Askenasy
- Department of Biochemistry, Tennis Court Road, Cambridge, United Kingdom
| | - Leonardo Mancini
- Department of Biochemistry, Tennis Court Road, Cambridge, United Kingdom
| | - Pok-Man Ho
- Department of Biochemistry, Tennis Court Road, Cambridge, United Kingdom
| | | | - Jemima E V Swain
- Department of Biochemistry, Tennis Court Road, Cambridge, United Kingdom
| | - Martin Welch
- Department of Biochemistry, Tennis Court Road, Cambridge, United Kingdom.
| |
Collapse
|
14
|
Ferngren G, Yu D, Unalan-Altintop T, Dinnétz P, Özenci V. Epidemiological patterns of candidaemia: A comprehensive analysis over a decade. Mycoses 2024; 67:e13729. [PMID: 38682399 DOI: 10.1111/myc.13729] [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: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND The prevalence of fungal bloodstream infections (BSI), especially candidaemia, has been increasing globally during the last decades. Fungal diagnosis is still challenging due to the slow growth of fungal microorganisms and need for special expertise. Fungal polymicrobial infections further complicate the diagnosis and extend the time required. Epidemiological data are vital to generate effective empirical treatment strategies. OBJECTIVES The overall aim of this project is to describe the epidemiology of monomicrobial candidaemia and polymicrobial BSI, both with mixed fungaemia and with mixed Candida/bacterial BSIs. METHODS We conducted a single-centre retrospective epidemiological study that encompasses 950,161 blood cultures during the years 2010 to 2020. The epidemiology of monomicrobial and polymicrobial candidaemia episodes were investigated from the electronic records. RESULTS We found that 1334 candidaemia episodes were identified belonging to 1144 individual patients during 2010 to 2020. Candida albicans was the most prevalent species detected in candidaemia patients, representing 57.7% of these episodes. Nakaseomyces (Candida) glabrata and Candida parapsilosis complex showed an increasing trend compared to previous studies, whereas Candida albicans demonstrated a decrease. 19.8% of these episodes were polymicrobial and 17% presented with mixed Candida/bacterial BSIs while 2.8% were mixed fungaemia. C. albicans and N. glabrata were the most common combination (51.4%) in mixed fungaemia episodes. Enterococcus and Lactobacillus spp. were the most common bacteria isolated in mixed Candida/bacterial BSIs. CONCLUSIONS Polymicrobial growth with candidaemia is common, mostly being mixed Candida/bacterial BSIs. C. albicans was detected in more than half of all the candidaemia patients however showed a decreasing trend in time, whereas an increase is noteworthy in C. parapsilosis complex and N. glabrata.
Collapse
Affiliation(s)
- Gordon Ferngren
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David Yu
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tugce Unalan-Altintop
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Microbiology, Hacettepe University Medical School, Ankara, Turkey
| | - Patrik Dinnétz
- School of Natural Sciences, Technology and Environmental Studies, Södertörn University, Stockholm, Sweden
| | - Volkan Özenci
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
15
|
Paul S, Todd OA, Eichelberger KR, Tkaczyk C, Sellman BR, Noverr MC, Cassat JE, Fidel PL, Peters BM. A fungal metabolic regulator underlies infectious synergism during Candida albicans - Staphylococcus aureus intra-abdominal co-infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580531. [PMID: 38405692 PMCID: PMC10888754 DOI: 10.1101/2024.02.15.580531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) uncovered synergistic lethality that was driven by Candida -induced upregulation of functional S. aureus ⍺-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken and revealed that zcf13 Δ/Δ failed to drive augmented ⍺-toxin or lethal synergism during co-infection. Using a combination of transcriptional and phenotypic profiling approaches, ZCF13 was shown to regulate genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments revealed that ribose inhibited the staphylococcal agr quorum sensing system and concomitantly repressed toxicity. Unlike wild-type C. albicans , zcf13 Δ/Δ was unable to effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13 Δ/Δ mutant fully restored pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.
Collapse
|
16
|
Timme S, Wendler S, Klassert TE, Saraiva JP, da Rocha UN, Wittchen M, Schramm S, Ehricht R, Monecke S, Edel B, Rödel J, Löffler B, Ramirez MS, Slevogt H, Figge MT, Tuchscherr L. Competitive inhibition and mutualistic growth in co-infections: deciphering Staphylococcus aureus-Acinetobacter baumannii interaction dynamics. ISME COMMUNICATIONS 2024; 4:ycae077. [PMID: 38962494 PMCID: PMC11221087 DOI: 10.1093/ismeco/ycae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
Staphylococcus aureus (Sa) and Acinetobacter baumannii (Ab) are frequently co-isolated from polymicrobial infections that are severe and refractory to therapy. Here, we apply a combination of wet-lab experiments and in silico modeling to unveil the intricate nature of the Ab/Sa interaction using both, representative laboratory strains and strains co-isolated from clinical samples. This comprehensive methodology allowed uncovering Sa's capability to exert a partial interference on Ab by the expression of phenol-soluble modulins. In addition, we observed a cross-feeding mechanism by which Sa supports the growth of Ab by providing acetoin as an alternative carbon source. This study is the first to dissect the Ab/Sa interaction dynamics wherein competitive and cooperative strategies can intertwine. Through our findings, we illuminate the ecological mechanisms supporting their coexistence in the context of polymicrobial infections. Our research not only enriches our understanding but also opens doors to potential therapeutic avenues in managing these challenging infections.
Collapse
Affiliation(s)
- Sandra Timme
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Friedrich Schiller University Jena, Leibniz Centre for Photonics in Infection Research (LPI), D-07743 Jena, Germany
| | - Sindy Wendler
- Institute of Medical Microbiology, Jena University Hospital, D-07740 Jena, Germany
| | - Tilman E Klassert
- Respiratory Infection Dynamics, Helmholtz Centre for Infection Research – HZI, D-38124 Braunschweig, Germany
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, German Center for Lung Research (DZL), BREATH, D-30625 Hannover, Germany
| | - Joao Pedro Saraiva
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, D-04318 Leipzig, Germany
| | - Ulisses Nunes da Rocha
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, D-04318 Leipzig, Germany
| | - Manuel Wittchen
- Center for Biotechnology, Bielefeld University, D-33501 Bielefeld, Germany
| | - Sareda Schramm
- Department of Biological Science, Center for Applied Biotechnology Studies, California State University, 800 N State College Blvd, Fullerton, CA 92831, United States
| | - Ralf Ehricht
- Leibniz Institute of Photonic Technology, Leibniz Centre for Photonics in Infection Research (LPI), D-07745 Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Leibniz Centre for Photonics in Infection Research (LPI) , D-07743 Jena, Germany
| | - Stefan Monecke
- Leibniz Institute of Photonic Technology, Leibniz Centre for Photonics in Infection Research (LPI), D-07745 Jena, Germany
- Institute for Medical Microbiology and Virology, Dresden University Hospital, Dresden, Germany
| | - Birgit Edel
- Institute of Medical Microbiology, Jena University Hospital, D-07740 Jena, Germany
| | - Jürgen Rödel
- Institute of Medical Microbiology, Jena University Hospital, D-07740 Jena, Germany
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, D-07740 Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, D-07743 Jena, Germany
| | - Maria Soledad Ramirez
- Department of Biological Science, Center for Applied Biotechnology Studies, California State University, 800 N State College Blvd, Fullerton, CA 92831, United States
| | - Hortense Slevogt
- Respiratory Infection Dynamics, Helmholtz Centre for Infection Research – HZI, D-38124 Braunschweig, Germany
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, German Center for Lung Research (DZL), BREATH, D-30625 Hannover, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Friedrich Schiller University Jena, Leibniz Centre for Photonics in Infection Research (LPI), D-07743 Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, D-07743 Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, D-07743 Jena, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, D-07740 Jena, Germany
| |
Collapse
|
17
|
Baxter KJ, Sargison FA, Fitzgerald JR, McConnell G, Hoskisson PA. Time-lapse mesoscopy of Candida albicans and Staphylococcus aureus dual-species biofilms reveals a structural role for the hyphae of C. albicans in biofilm formation. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001426. [PMID: 38261525 PMCID: PMC10866020 DOI: 10.1099/mic.0.001426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
Polymicrobial infection with Candida albicans and Staphylococcus aureus may result in a concomitant increase in virulence and resistance to antimicrobial drugs. This enhanced pathogenicity phenotype is mediated by numerous factors, including metabolic processes and direct interaction of S. aureus with C. albicans hyphae. The overall structure of biofilms is known to contribute to their recalcitrance to treatment, although the dynamics of direct interaction between species and how it contributes to pathogenicity is poorly understood. To address this, a novel time-lapse mesoscopic optical imaging method was developed to enable the formation of C. albicans/S. aureus whole dual-species biofilms to be followed. It was found that yeast-form or hyphal-form C. albicans in the biofilm founder population profoundly affects the structure of the biofilm as it matures. Different sub-populations of C. albicans and S. aureus arise within each biofilm as a result of the different C. albicans morphotypes, resulting in distinct sub-regions. These data reveal that C. albicans cell morphology is pivotal in the development of global biofilm architecture and the emergence of colony macrostructures and may temporally influence synergy in infection.
Collapse
Affiliation(s)
- Katherine J. Baxter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Fiona A. Sargison
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
18
|
Eichelberger KR, Paul S, Peters BM, Cassat JE. Candida-bacterial cross-kingdom interactions. Trends Microbiol 2023; 31:1287-1299. [PMID: 37640601 PMCID: PMC10843858 DOI: 10.1016/j.tim.2023.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
While the fungus Candida albicans is a common colonizer of healthy humans, it is also responsible for mucosal infections and severe invasive disease. Understanding the mechanisms that allow C. albicans to exist as both a benign commensal and as an invasive pathogen have been the focus of numerous studies, and recent findings indicate an important role for cross-kingdom interactions on C. albicans biology. This review highlights how C. albicans-bacteria interactions influence healthy polymicrobial community structure, host immune responses, microbial pathogenesis, and how dysbiosis may lead to C. albicans infection. Finally, we discuss how cross-kingdom interactions represent an opportunity to identify new antivirulence compounds that target fungal infections.
Collapse
Affiliation(s)
- Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Saikat Paul
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
19
|
Ramage G, Borghi E, Rodrigues CF, Kean R, Williams C, Lopez-Ribot J. Our current clinical understanding of Candida biofilms: where are we two decades on? APMIS 2023; 131:636-653. [PMID: 36932821 DOI: 10.1111/apm.13310] [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: 02/27/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023]
Abstract
Clinically we have been aware of the concept of Candida biofilms for many decades, though perhaps without the formal designation. Just over 20 years ago the subject emerged on the back of progress made from the bacterial biofilms, and academic progress pace has continued to mirror the bacterial biofilm community, albeit at a decreased volume. It is apparent that Candida species have a considerable capacity to colonize surfaces and interfaces and form tenacious biofilm structures, either alone or in mixed species communities. From the oral cavity, to the respiratory and genitourinary tracts, wounds, or in and around a plethora of biomedical devices, the scope of these infections is vast. These are highly tolerant to antifungal therapies that has a measurable impact on clinical management. This review aims to provide a comprehensive overight of our current clinical understanding of where these biofilms cause infections, and we discuss existing and emerging antifungal therapies and strategies.
Collapse
Affiliation(s)
- Gordon Ramage
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
| | - Elisa Borghi
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Department of Health Sciences, San Paolo Medical School, Università Degli Studi di Milano, Milan, Italy
| | - Célia Fortuna Rodrigues
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- LEPABE-Department of Chemical Engineering, Faculty of Engineering, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
| | - Ryan Kean
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Department of Biological Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Craig Williams
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Microbiology Department, Morecambe Bay NHS Trust, Lancaster, UK
| | - Jose Lopez-Ribot
- Department of Biology and the South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
20
|
Wang F, Wang Z, Tang J. The interactions of Candida albicans with gut bacteria: a new strategy to prevent and treat invasive intestinal candidiasis. Gut Pathog 2023; 15:30. [PMID: 37370138 DOI: 10.1186/s13099-023-00559-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The gut microbiota plays an important role in human health, as it can affect host immunity and susceptibility to infectious diseases. Invasive intestinal candidiasis is strongly associated with gut microbiota homeostasis. However, the nature of the interaction between Candida albicans and gut bacteria remains unclear. OBJECTIVE This review aimed to determine the nature of interaction and the effects of gut bacteria on C. albicans so as to comprehend an approach to reducing intestinal invasive infection by C. albicans. METHODS This review examined 11 common gut bacteria's interactions with C. albicans, including Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Enterococcus faecalis, Staphylococcus aureus, Salmonella spp., Helicobacter pylori, Lactobacillus spp., Bacteroides spp., Clostridium difficile, and Streptococcus spp. RESULTS Most of the studied bacteria demonstrated both synergistic and antagonistic effects with C. albicans, and just a few bacteria such as P. aeruginosa, Salmonella spp., and Lactobacillus spp. demonstrated only antagonism against C. albicans. CONCLUSIONS Based on the nature of interactions reported so far by the literature between gut bacteria and C. albicans, it is expected to provide new ideas for the prevention and treatment of invasive intestinal candidiasis.
Collapse
Affiliation(s)
- Fei Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200240, China
| | - Zetian Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200240, China.
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200240, China.
| |
Collapse
|
21
|
Chen YH, Yeung F, Lacey KA, Zaldana K, Lin JD, Bee GCW, McCauley C, Barre RS, Liang SH, Hansen CB, Downie AE, Tio K, Weiser JN, Torres VJ, Bennett RJ, Loke P, Graham AL, Cadwell K. Rewilding of laboratory mice enhances granulopoiesis and immunity through intestinal fungal colonization. Sci Immunol 2023; 8:eadd6910. [PMID: 37352372 PMCID: PMC10350741 DOI: 10.1126/sciimmunol.add6910] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
The paucity of blood granulocyte populations such as neutrophils in laboratory mice is a notable difference between this model organism and humans, but the cause of this species-specific difference is unclear. We previously demonstrated that laboratory mice released into a seminatural environment, referred to as rewilding, display an increase in blood granulocytes that is associated with expansion of fungi in the gut microbiota. Here, we find that tonic signals from fungal colonization induce sustained granulopoiesis through a mechanism distinct from emergency granulopoiesis, leading to a prolonged expansion of circulating neutrophils that promotes immunity. Fungal colonization after either rewilding or oral inoculation of laboratory mice with Candida albicans induced persistent expansion of myeloid progenitors in the bone marrow. This increase in granulopoiesis conferred greater long-term protection from bloodstream infection by gram-positive bacteria than by the trained immune response evoked by transient exposure to the fungal cell wall component β-glucan. Consequently, introducing fungi into laboratory mice may restore aspects of leukocyte development and provide a better model for humans and free-living mammals that are constantly exposed to environmental fungi.
Collapse
Affiliation(s)
- Ying-Han Chen
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Frank Yeung
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kimberly Zaldana
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, Taiwan
| | - Gavyn Chern Wei Bee
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Caroline McCauley
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ramya S. Barre
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Shen-Huan Liang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Christina B. Hansen
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Alexander E Downie
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Kyle Tio
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Jeffrey N. Weiser
- Antimicrobial-Resistant Pathogens Program
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Victor J Torres
- Antimicrobial-Resistant Pathogens Program
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Richard J. Bennett
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
22
|
MacAlpine J, Robbins N, Cowen LE. Bacterial-fungal interactions and their impact on microbial pathogenesis. Mol Ecol 2023; 32:2565-2581. [PMID: 35231147 PMCID: PMC11032213 DOI: 10.1111/mec.16411] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
Microbial communities of the human microbiota exhibit diverse effects on human health and disease. Microbial homeostasis is important for normal physiological functions and changes to the microbiota are associated with many human diseases including diabetes, cancer, and colitis. In addition, there are many microorganisms that are either commensal or acquired from environmental reservoirs that can cause diverse pathologies. Importantly, the balance between health and disease is intricately connected to how members of the microbiota interact and affect one another's growth and pathogenicity. However, the mechanisms that govern these interactions are only beginning to be understood. In this review, we outline bacterial-fungal interactions in the human body, including examining the mechanisms by which bacteria govern fungal growth and virulence, as well as how fungi regulate bacterial pathogenesis. We summarize advances in the understanding of chemical, physical, and protein-based interactions, and their role in exacerbating or impeding human disease. We focus on the three fungal species responsible for the majority of systemic fungal infections in humans: Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. We conclude by summarizing recent studies that have mined microbes for novel antimicrobials and antivirulence factors, highlighting the potential of the human microbiota as a rich resource for small molecule discovery.
Collapse
Affiliation(s)
- Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
23
|
Manoharadas S, Ahmad N, Altaf M, Alrefaei AF, Al-Rayes BF. An Enzybiotic Cocktail Effectively Disrupts Preformed Dual Biofilm of Staphylococcus aureus and Enterococcus faecalis. Pharmaceuticals (Basel) 2023; 16:ph16040564. [PMID: 37111322 PMCID: PMC10145859 DOI: 10.3390/ph16040564] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/05/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Multidrug-resistant bacterial infections are on the rise around the world. Chronic infections caused by these pathogens through biofilm mediation often complicate the situation. In natural settings, biofilms are often formed with different species of bacteria existing synergistically or antagonistically. Biofilms on diabetic foot ulcers are formed predominantly by two opportunistic pathogens, Staphylococcus aureus and Enterococcus faecalis. Bacteriophages and phage-based proteins, including endolysins, have been found to be active against biofilms. In this study, we evaluated the activity of two engineered enzybiotics either by themselves or as a combination against a dual biofilm formed by S. aureus and E. faecalis in an inert glass surface. An additive effect in rapidly disrupting the preformed dual biofilm was observed with the cocktail of proteins, in comparison with mono treatment. The cocktail-treated biofilms were dispersed by more than 90% within 3 h of treatment. Apart from biofilm disruption, bacterial cells embedded in the biofilm matrix were also effectively reduced by more than 90% within 3 h of treatment. This is the first instance where a cocktail of engineered enzybiotics has been effectively used to impede the structural integrity of a dual biofilm.
Collapse
Affiliation(s)
- Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
- Central Laboratory RM 63AA, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
| | - Naushad Ahmad
- Central Laboratory RM 63AA, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
| | - Mohammad Altaf
- Central Laboratory RM 63AA, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
| | - Abdulwahed Fahad Alrefaei
- Department of Zoology, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
| | - Basel F Al-Rayes
- Central Laboratory RM 63AA, College of Science, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia
| |
Collapse
|
24
|
Khan F, Tabassum N, Jeong GJ, Jung WK, Kim YM. Inhibition of Mixed Biofilms of Candida albicans and Staphylococcus aureus by β-Caryophyllene-Gold Nanoparticles. Antibiotics (Basel) 2023; 12:antibiotics12040726. [PMID: 37107087 PMCID: PMC10134979 DOI: 10.3390/antibiotics12040726] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Polymicrobial biofilms, consisting of fungal and bacterial pathogens, often contribute to the failure of antimicrobial treatment. The growing resistance of pathogenic polymicrobial biofilms to antibiotics has led to the development of alternative strategies to combat polymicrobial diseases. To this end, nanoparticles synthesized using natural molecules have received significant attention for disease treatment. Here, gold nanoparticles (AuNPs) were synthesized using β-caryophyllene, a bioactive compound isolated from various plant species. The shape, size, and zeta potential of the synthesized β-c-AuNPs were found to be non-spherical, 17.6 ± 1.2 nm, and -31.76 ± 0.73 mV, respectively. A mixed biofilm of Candida albicans and Staphylococcus aureus was used to test the efficacy of the synthesized β-c-AuNPs. The results revealed a concentration-dependent inhibition of the initial stages of formation of single-species as well as mixed biofilms. Furthermore, β-c-AuNPs also eliminated mature biofilms. Therefore, using β-c-AuNPs to inhibit biofilm and eradicate bacterial-fungal mixed biofilms represents a promising therapeutic approach for controlling polymicrobial infections.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
25
|
Rather IA, Wani MY, Kamli MR, Sabir JSM, Hakeem KR, Firoz A, Park YH, Hor YY. Limosilactobacillus fermentum KAU0021 Abrogates Mono- and Polymicrobial Biofilms Formed by Candida albicans and Staphylococcus aureus. Pharmaceutics 2023; 15:pharmaceutics15041079. [PMID: 37111565 PMCID: PMC10145238 DOI: 10.3390/pharmaceutics15041079] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/28/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Candida albicans and Staphylococcus aureus, representing two different kingdoms, are the most frequently isolated pathogens from invasive infections. Their pathogenic attributes, combined with drug resistance, make them a major threat and a challenge to successful treatments, mainly when involved in polymicrobial biofilm-associated infections. In the present study, we investigated the antimicrobial potential of Lactobacillus metabolite extracts (LMEs) purified from cell-free supernatant of four Lactobacillus strains (KAU007, KAU0010, KAU0021, and Pro-65). Furthermore, LME obtained from the strain KAU0021 (LMEKAU0021), being the most effective, was analyzed for its anti-biofilm property against mono- and polymicrobial biofilms formed by C. albicans and S. aureus. The impact of LMEKAU0021 on membrane integrity in single and mixed culture conditions was also evaluated using propidium iodide. The MIC values recorded for LMEKAU0021 was 406 µg/mL, 203 µg/mL, and 406 µg/mL against planktonic cells of C. albicans SC5314, S. aureus and polymicrobial culture, respectively. The LMEKAU0021 at sub-MIC values potentially abrogates both biofilm formation as well as 24 h mature mono- and polymicrobial biofilms. These results were further validated using different microscopy and viability assays. For insight mechanism, LMEKAU0021 displayed a strong impact on cell membrane integrity of both pathogens in single and mixed conditions. A hemolytic assay using horse blood cells at different concentrations of LMEKAU0021 confirmed the safety of this extract. The results from this study correlate the antimicrobial and anti-biofilm properties of lactobacilli against bacterial and fungal pathogens in different conditions. Further in vitro and in vivo studies determining these effects will support the aim of discovering an alternative strategy for combating serious polymicrobial infections caused by C. albicans and S. aureus.
Collapse
|
26
|
Grazul M, Balcerczak E, Sienkiewicz M. Analysis of the Presence of the Virulence and Regulation Genes from Staphylococcus aureus ( S. aureus) in Coagulase Negative Staphylococci and the Influence of the Staphylococcal Cross-Talk on Their Functions. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:5155. [PMID: 36982064 PMCID: PMC10049693 DOI: 10.3390/ijerph20065155] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 06/18/2023]
Abstract
Coagulase-negative staphylococci (CoNS) are increasingly becoming a public health issue worldwide due to their growing resistance to antibiotics and common involvement in complications related to invasive surgical procedures, and nosocomial and urinary tract infections. Their behavior either as a commensal or a pathogen is a result of strict regulation of colonization and virulence factors. Although functionality of virulence factors and processes involved in their regulation are quite well understood in S. aureus, little is known about them in CoNS species. Therefore, the aim of our studies was to check if clinical CoNS strains may contain virulence factors and genes involved in resistance to methicillin, that are homologous to S. aureus. Moreover, we checked the presence of elements responsible for regulation of genes that encode virulence factors typical for S. aureus in tested isolates. We also investigated whether the regulation factors produced by one CoNS isolate can affect virulence activity of other strains by co-incubation of tested isolates with supernatant from other isolates. Our studies confirmed the presence of virulence factor and regulatory genes attributed to S. aureus in CoNS isolates and indicated that one strain with an active agr gene is able to affect biofilm formation and δ-toxin activity of strains with inactive agr genes. The cognition of prevalence and regulation of virulence factors as well as antibiotic resistance of CoNS isolates is important for better control and treatment of CoNS infections.
Collapse
Affiliation(s)
- Magdalena Grazul
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| |
Collapse
|
27
|
Hurley JC. Structural equation modelling the impact of antimicrobials on the human microbiome. Colonization resistance versus colonization susceptibility as case studies. J Antimicrob Chemother 2023; 78:328-337. [PMID: 36512373 DOI: 10.1093/jac/dkac408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The impact of antimicrobials on the human microbiome and its relationship to human health are of great interest. How antimicrobial exposure might drive change within specific constituents of the microbiome to effect clinically relevant endpoints is difficult to study. Clinical investigation of each step within a network of causation would be challenging if done 'step-by-step'. An analytic tool of great potential to clinical microbiome research is structural equation modelling (SEM), which has a long history of applications to research questions arising within subject areas as diverse as psychology and econometrics. SEM enables postulated models based on a network of causation to be tested en bloc by confrontation with data derived from the literature. Case studies for the potential application of SEM techniques are colonization resistance (CR) and its counterpart, colonization susceptibility (CS), wherein specific microbes within the microbiome are postulated to either impede (CR) or facilitate (CS) invasive infection with pathogenic bacteria. These postulated networks have three causation steps: exposure to specific antimicrobials are key drivers, clinically relevant infection endpoints are the measurable observables and the activity of key microbiome constituents mediating CR or CS, which may be unobservable, appear as latent variables in the model. SEM methods have potential application towards evaluating the activity of specific antimicrobial agents within postulated networks of causation using clinically derived data.
Collapse
Affiliation(s)
- James C Hurley
- Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.,Division of Internal Medicine, Ballarat Health Services, Ballarat, Victoria, Australia
| |
Collapse
|
28
|
There Is More to Wounds than Bacteria: Fungal Biofilms in Chronic Wounds. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023. [DOI: 10.1007/s40588-022-00187-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Abstract
Purpose of Review
The management of chronic wounds, a debilitating condition, presents a considerable challenge to healthcare professionals and a significant burden on services. When these wounds are exposed to the external environment, they are susceptible to microbial infection, which further complicates their management and worsens clinical outcomes.
Recent Findings
Bacteria typically exist in wounds as part of a biofilm, which is often polymicrobial in nature, alongside bacteria and fungi that are described as being more virulent and tolerant towards antimicrobials and antiseptics. Despite advancing knowledge in polymicrobial biofilm wound infections with respect to bacteria, the role of fungi is largely ignored, and their influence in chronicity and clinical management is not fully appreciated or understood.
Summary
The purpose of this review is to explore the significance of fungi within chronic wound environments and, in doing so, understand the importance of interkingdom interactions in wound management.
Collapse
|
29
|
Liu CC, Lin MH. Hitchhiking motility of Staphylococcus aureus involves the interaction between its wall teichoic acids and lipopolysaccharide of Pseudomonas aeruginosa. Front Microbiol 2023; 13:1068251. [PMID: 36687638 PMCID: PMC9849799 DOI: 10.3389/fmicb.2022.1068251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus, which lacks pili and flagella, is nonmotile. However, it hitchhikes motile bacteria, such as Pseudomonas aeruginosa, to migrate in the environment. This study demonstrated that the hitchhiking motility of S. aureus SA113 was reduced after the tagO, which encodes an enzyme for wall teichoic acids (WTA) synthesis, was deleted. The hitchhiking motility was restored after the mutation was complemented by transforming a plasmid expressing TagO into the mutant. We also showed that adding purified lipopolysaccharide (LPS) to a culture that contains S. aureus SA113 and P. aeruginosa PAO1, reduced the movement of S. aureus, showing that WTA and LPS are involved in the hitchhiking motility of S. aureus. This study also found that P. aeruginosa promoted the movement of S. aureus in the digestive tract of Caenorhabditis elegans and in mice. In conclusion, this study reveals how S. aureus hitchhikes P. aeruginosa for translocation in an ecosystem. The results from this study improve our understanding on how a nonmotile pathogen moves in the environment and spreads in animals.
Collapse
Affiliation(s)
- Chao-Chin Liu
- 1Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Mei-Hui Lin
- 1Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan,2Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan,3Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan,*Correspondence: Mei-Hui Lin, ✉
| |
Collapse
|
30
|
Jaswal K, Todd OA, Behnsen J. Neglected gut microbiome: interactions of the non-bacterial gut microbiota with enteric pathogens. Gut Microbes 2023; 15:2226916. [PMID: 37365731 PMCID: PMC10305517 DOI: 10.1080/19490976.2023.2226916] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
A diverse array of commensal microorganisms inhabits the human intestinal tract. The most abundant and most studied members of this microbial community are undoubtedly bacteria. Their important role in gut physiology, defense against pathogens, and immune system education has been well documented over the last decades. However, the gut microbiome is not restricted to bacteria. It encompasses the entire breadth of microbial life: viruses, archaea, fungi, protists, and parasitic worms can also be found in the gut. While less studied than bacteria, their divergent but important roles during health and disease have become increasingly more appreciated. This review focuses on these understudied members of the gut microbiome. We will detail the composition and development of these microbial communities and will specifically highlight their functional interactions with enteric pathogens, such as species of the family Enterobacteriaceae. The interactions can be direct through physical interactions, or indirect through secreted metabolites or modulation of the immune response. We will present general concepts and specific examples of how non-bacterial gut communities modulate bacterial pathogenesis and present an outlook for future gut microbiome research that includes these communities.
Collapse
Affiliation(s)
- Kanchan Jaswal
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Olivia A Todd
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Judith Behnsen
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
31
|
Hamion G, Aucher W, Tardif C, Miranda J, Rouger C, Imbert C, Girardot M. Valorization of Invasive Plant Extracts against the Bispecies Biofilm Staphylococcus aureus- Candida albicans by a Bioguided Molecular Networking Screening. Antibiotics (Basel) 2022; 11:antibiotics11111595. [PMID: 36421241 PMCID: PMC9686625 DOI: 10.3390/antibiotics11111595] [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: 10/21/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Invasive plants efficiently colonize non-native territories, suggesting a great production of bioactive metabolites which could be effective antibiofilm weapons. Our study aimed to look for original molecules able to inhibit bispecies biofilm formed by S. aureus and C. albicans. Extracts from five invasive macrophytes (Ludwigia peploides, Ludwigia grandiflora, Myriophyllum aquaticum, Lagarosiphon major and Egeria densa) were prepared and tested in vitro against 24 h old bispecies biofilms using a crystal violet staining (CVS) assay. The activities of the extracts reducing the biofilm total biomass by 50% or more were comparatively analyzed against each microbial species forming the biofilm by flow cytometry (FCM) and scanning electron microscopy. Extracts active against both species were fractionated. Obtained fractions were analyzed by UHPLC-MS/MS and evaluated by the CVS assay. Chemical and biological data were combined into a bioactivity-based molecular networking (BBMN) to identify active compounds. The aerial stem extract of L. grandiflora showed the highest antibiofilm activity (>50% inhibition at 50 µg∙mL−1). The biological, chemical and BBMN investigations of its fractions highlighted nine ions correlated with the antibiofilm activity. The most correlated compound, identified as betulinic acid (BA), inhibited bispecies biofilms regardless of the three tested couples of strains (ATCC strains: >40% inhibition, clinical isolates: ≈27% inhibition), confirming its antibiofilm interest.
Collapse
Affiliation(s)
- Guillaume Hamion
- Laboratoire EBI, University of Poitiers, UMR CNRS 7267, F-86000 Poitiers, France
- Correspondence:
| | - Willy Aucher
- Laboratoire EBI, University of Poitiers, UMR CNRS 7267, F-86000 Poitiers, France
| | - Charles Tardif
- University of Bordeaux, UMR INRAE 1366, Bordeaux INP, OENO, ISVV, F-33140 Villenave d’Ornon, France
- Bordeaux Sciences Agro, UMR INRAE 1366, Bordeaux INP, OENO, ISVV, F-33170 Gradignan, France
| | - Julie Miranda
- University of Bordeaux, UMR INRAE 1366, Bordeaux INP, OENO, ISVV, F-33140 Villenave d’Ornon, France
- Bordeaux Sciences Agro, UMR INRAE 1366, Bordeaux INP, OENO, ISVV, F-33170 Gradignan, France
| | - Caroline Rouger
- University of Bordeaux, UMR INRAE 1366, Bordeaux INP, OENO, ISVV, F-33140 Villenave d’Ornon, France
- Bordeaux Sciences Agro, UMR INRAE 1366, Bordeaux INP, OENO, ISVV, F-33170 Gradignan, France
- Bordeaux Metabolome, MetaboHUB, PHENOME-EMPHASIS, Centre INRAE de Nouvelle Aquitaine-Bordeaux, F-33140 Villenave d’Ornon, France
| | - Christine Imbert
- Laboratoire EBI, University of Poitiers, UMR CNRS 7267, F-86000 Poitiers, France
| | - Marion Girardot
- Laboratoire EBI, University of Poitiers, UMR CNRS 7267, F-86000 Poitiers, France
| |
Collapse
|
32
|
Essential Fitness Repertoire of Staphylococcus aureus during Co-infection with Acinetobacter baumannii In Vivo. mSystems 2022; 7:e0033822. [PMID: 36040021 PMCID: PMC9600432 DOI: 10.1128/msystems.00338-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus represents a major human pathogen that is frequently involved in polymicrobial infections. However, the prevalence and role of co-infectious microbes on the pathogenesis and fitness essentiality of S. aureus in vivo remain largely unknown. In this study, we firstly performed a retrospective surveillance of 760 clinical samples and revealed a notable predominance of co-infection with S. aureus and Acinetobacter baumannii. The high-density S. aureus transposon mutant library coupled to transposon insertion sequencing (Tn-Seq) further identified a core set of genes enriched in metabolism of inorganic ions, amino acids, and carbohydrates, which are essential for infection and tissue colonization of S. aureus in the murine systemic infection model. Notably, we revealed a differential requirement of fitness factors for S. aureus in tissue-specific (liver and kidney) and infection-type-specific manner (mono- and co-infection). Co-infection with A. baumannii dramatically altered the fitness requirements of S. aureus in vivo; 49% of the mono-infection fitness genes in S. aureus strain Newman were converted to non-essential, and the functionality of ATP-binding cassette (ABC) transporters was significantly elicited during co-infection. Furthermore, the number of genes essential during co-infection (503) outnumbers the genes essential during mono-infection (362). In addition, the roles of 3 infection-type-specific genes in S. aureus during mono-infection or co-infection with A. baumannii were validated with competitive experiments in vivo. Our data indicated a high incidence and clinical relevance of S. aureus and A. baumannii co-infection, and provided novel insights into establishing antimicrobial regimens to control co-infections. IMPORTANCE Polymicrobial infections are widespread in clinical settings, which potentially correlate with increased infection severity and poor clinical outcomes. Staphylococcus aureus is a formidable human pathogen that causes a variety of diseases in polymicrobial nature. Co-infection and interaction of S. aureus have been described with limited pathogens, mainly including Pseudomonas aeruginosa, Candida albicans, and influenza A virus. Thus far, the prevalence and role of co-infectious microbes on the pathogenesis and fitness essentiality of S. aureus in vivo remain largely unknown. Understanding the polymicrobial composition and interaction, from a community and genome-wide perspective, is thus crucial to shed light on S. aureus pathogenesis strategy. Here, our findings demonstrated, for the first time, that a high incidence rate and clinical relevance of co-infection was caused by S. aureus and Acinetobacter baumannii, illustrating the importance of polymicrobial nature in investigating S. aureus pathogenesis. The infection-type-specific genes likely serve as potential therapeutic targets to control S. aureus infections, either in mono- or co-infection situation, providing novel insights into the development of antimicrobial regimens to control co-infections.
Collapse
|
33
|
Ahle CM, Stødkilde K, Poehlein A, Bömeke M, Streit WR, Wenck H, Reuter JH, Hüpeden J, Brüggemann H. Interference and co-existence of staphylococci and Cutibacterium acnes within the healthy human skin microbiome. Commun Biol 2022; 5:923. [PMID: 36071129 PMCID: PMC9452508 DOI: 10.1038/s42003-022-03897-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/25/2022] [Indexed: 12/12/2022] Open
Abstract
Human skin is populated by trillions of microbes collectively called the skin microbiome. Staphylococcus epidermidis and Cutibacterium acnes are among the most abundant members of this ecosystem, with described roles in skin health and disease. However, knowledge regarding the health beneficial effects of these ubiquitous skin residents is still limited. Here, we profiled the staphylococcal and C. acnes landscape across four different skin sites of 30 individuals (120 skin samples) using amplicon-based next-generation sequencing. Relative abundance profiles obtained indicated the existence of phylotype-specific co-existence and exclusion scenarios. Co-culture experiments with 557 staphylococcal strains identified 30 strains exhibiting anti-C. acnes activities. Notably, staphylococcal strains were found to selectively exclude acne-associated C. acnes and co-exist with healthy skin-associated phylotypes, through regulation of the antimicrobial activity. Overall, these findings highlight the importance of skin-resident staphylococci and suggest that selective microbial interference is a contributor to healthy skin homeostasis. The dynamic interaction between the common resident skin microbes Staphylococcus epidermidis and Cutibacterium acnes is uncovered, showing that S. epidermidis can selectively exclude acne-associated C. acnes strains from the human skin.
Collapse
Affiliation(s)
- Charlotte Marie Ahle
- Beiersdorf AG, Research & Development, Front End Innovation, 20245, Hamburg, Germany. .,Department of Microbiology and Biotechnology, University of Hamburg, 22609, Hamburg, Germany.
| | | | - Anja Poehlein
- Department of Genomic and Applied Microbiology, Institute of Microbiology and Genetics, University of Göttingen, 37073, Göttingen, Germany
| | - Mechthild Bömeke
- Department of Genomic and Applied Microbiology, Institute of Microbiology and Genetics, University of Göttingen, 37073, Göttingen, Germany
| | - Wolfgang R Streit
- Department of Microbiology and Biotechnology, University of Hamburg, 22609, Hamburg, Germany
| | - Horst Wenck
- Beiersdorf AG, Research & Development, Front End Innovation, 20245, Hamburg, Germany
| | - Jörn Hendrik Reuter
- Beiersdorf AG, Research & Development, Front End Innovation, 20245, Hamburg, Germany
| | - Jennifer Hüpeden
- Beiersdorf AG, Research & Development, Front End Innovation, 20245, Hamburg, Germany
| | - Holger Brüggemann
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark.
| |
Collapse
|
34
|
Candida and the Gram-positive trio: testing the vibe in the ICU patient microbiome using structural equation modelling of literature derived data. Emerg Themes Epidemiol 2022; 19:7. [PMID: 35982466 PMCID: PMC9387012 DOI: 10.1186/s12982-022-00116-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Whether Candida interacts with Gram-positive bacteria, such as Staphylococcus aureus, coagulase negative Staphylococci (CNS) and Enterococci, to enhance their invasive potential from the microbiome of ICU patients remains unclear. Several effective anti-septic, antibiotic, anti-fungal, and non-decontamination based interventions studied for prevention of ventilator associated pneumonia (VAP) and other ICU acquired infections among patients receiving prolonged mechanical ventilation (MV) are known to variably impact Candida colonization. The collective observations within control and intervention groups from numerous ICU infection prevention studies enables tests of these postulated microbial interactions in the clinical context. Methods Four candidate generalized structural equation models (GSEM), each with Staphylococcus aureus, CNS and Enterococci colonization, defined as latent variables, were confronted with blood culture and respiratory tract isolate data derived from 460 groups of ICU patients receiving prolonged MV from 283 infection prevention studies. Results Introducing interaction terms between Candida colonization and each of S aureus (coefficient + 0.40; 95% confidence interval + 0.24 to + 0.55), CNS (+ 0.68; + 0.34 to + 1.0) and Enterococcal (+ 0.56; + 0.33 to + 0.79) colonization (all as latent variables) improved the fit for each model. The magnitude and significance level of the interaction terms were similar to the positive associations between exposure to topical antibiotic prophylaxis (TAP) on Enterococcal (+ 0.51; + 0.12 to + 0.89) and Candida colonization (+ 0.98; + 0.35 to + 1.61) versus the negative association of TAP with S aureus (− 0.45; − 0.70 to − 0.20) colonization and the negative association of anti-fungal exposure and Candida colonization (− 1.41; − 1.6 to − 0.72). Conclusions GSEM modelling of published ICU infection prevention data enables the postulated interactions between Candida and Gram-positive bacteria to be tested using clinically derived data. The optimal model implies interactions occurring in the human microbiome facilitating bacterial invasion and infection. This interaction might also account for the paradoxically high bacteremia incidences among studies of TAP in ICU patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12982-022-00116-9. GSEM modelling of published ICU infection prevention data from > 250 studies enables a test of and provides support to the interaction between Candida and Gram-positive bacteria. The various ICU infection prevention interventions may each broadly impact the patient microbiome.
Collapse
|
35
|
Durand BARN, Pouget C, Magnan C, Molle V, Lavigne JP, Dunyach-Remy C. Bacterial Interactions in the Context of Chronic Wound Biofilm: A Review. Microorganisms 2022; 10:microorganisms10081500. [PMID: 35893558 PMCID: PMC9332326 DOI: 10.3390/microorganisms10081500] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic wounds, defined by their resistance to care after four weeks, are a major concern, affecting millions of patients every year. They can be divided into three types of lesions: diabetic foot ulcers (DFU), pressure ulcers (PU), and venous/arterial ulcers. Once established, the classical treatment for chronic wounds includes tissue debridement at regular intervals to decrease biofilm mass constituted by microorganisms physiologically colonizing the wound. This particular niche hosts a dynamic bacterial population constituting the bed of interaction between the various microorganisms. The temporal reshuffle of biofilm relies on an organized architecture. Microbial community turnover is mainly associated with debridement (allowing transitioning from one major representant to another), but also with microbial competition and/or collaboration within wounds. This complex network of species and interactions has the potential, through diversity in antagonist and/or synergistic crosstalk, to accelerate, delay, or worsen wound healing. Understanding these interactions between microorganisms encountered in this clinical situation is essential to improve the management of chronic wounds.
Collapse
Affiliation(s)
- Benjamin A. R. N. Durand
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Cassandra Pouget
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Chloé Magnan
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, UMR 5235, 34000 Montpellier, France;
| | - Jean-Philippe Lavigne
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Catherine Dunyach-Remy
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
- Correspondence: ; Tel.: +33-466-683-202
| |
Collapse
|
36
|
Tran VN, Khan F, Han W, Luluil M, Truong VG, Yun HG, Choi S, Kim YM, Shin JH, Kang HW. Real-time monitoring of mono- and dual-species biofilm formation and eradication using microfluidic platform. Sci Rep 2022; 12:9678. [PMID: 35690659 PMCID: PMC9188611 DOI: 10.1038/s41598-022-13699-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/26/2022] [Indexed: 11/17/2022] Open
Abstract
In a human host, bacterial Staphylococcus aureus and fungal Candida albicans pathogens form a mixed biofilm that causes severe mortality and morbidity. However, research on the formation and eradication of mixed biofilms under dynamic conditions is lacking. Thus, this study employed a microfluidic technique to analyze the real-time formation of mono- and dual-species (S. aureus and C. albicans) biofilms and noninvasive optical treatment of the established mature biofilm using 405-nm laser light. A herringbone mixer thoroughly mixed both bacterial and fungal cells in the growth media before being injected into the observation channels on the microfluidic chip. At a flow rate of 1.0 µL/min of growth media for 24 h, the bacterial biofilm coverage was up to 15% higher than that of the fungal biofilm (50% for bacteria vs. 35% for fungus). On the other hand, the dual-species biofilm yielded the highest coverage of ~ 96.5% because of the collective interaction between S. aureus and C. albicans. The number of cell proliferation events in S. aureus was higher than that of C. albicans for 12 h, which indicates that the S. aureus biofilm was developed faster than C. albicans. The novel in situ test platform showed a significant bactericidal effect (80%) of the 405-nm laser light at 1080 J/cm2 towards the established S. aureus biofilm, whereas the same treatment removed approximately 69% of the mixed cells in the dual-species biofilm. This study revealed that the developed microfluidic platform could be utilized to monitor the formation of dual-species biofilms in real-time and laser-induced antimicrobial effects on dual-species biofilms.
Collapse
Affiliation(s)
- Van Nam Tran
- Industry 4.0 Convergence Bionics Engineering and Marine-Integrated Biomedical Technology Center, Pukyong National University, Busan, 48513, South Korea
| | - Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, South Korea
| | - Won Han
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, South Korea
| | - Maknuna Luluil
- Industry 4.0 Convergence Bionics Engineering and Marine-Integrated Biomedical Technology Center, Pukyong National University, Busan, 48513, South Korea
| | - Van Gia Truong
- Industry 4.0 Convergence Bionics Engineering and Marine-Integrated Biomedical Technology Center, Pukyong National University, Busan, 48513, South Korea
| | - Hyo Geun Yun
- Department of Electronic Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Sungyoung Choi
- Department of Electronic Engineering, Hanyang University, Seoul, 04763, South Korea.,Department of Biomedical Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, South Korea.,Department of Food Science and Technology, Pukyong National University, Busan, 48513, South Korea
| | - Joong Ho Shin
- Industry 4.0 Convergence Bionics Engineering and Marine-Integrated Biomedical Technology Center, Pukyong National University, Busan, 48513, South Korea. .,Department of Biomedical Engineering, Pukyong National University, Busan, 48513, South Korea.
| | - Hyun Wook Kang
- Industry 4.0 Convergence Bionics Engineering and Marine-Integrated Biomedical Technology Center, Pukyong National University, Busan, 48513, South Korea. .,Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, South Korea. .,Department of Biomedical Engineering, Pukyong National University, Busan, 48513, South Korea.
| |
Collapse
|
37
|
Redfern J, Tosheva L, Malic S, Butcher M, Ramage G, Verran J. The denture microbiome in health and disease: an exploration of a unique community. Lett Appl Microbiol 2022; 75:195-209. [PMID: 35634756 PMCID: PMC9546486 DOI: 10.1111/lam.13751] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022]
Abstract
The United Nations suggests the global population of denture wearers (an artificial device that acts as a replacement for teeth) is likely to rise significantly by the year 2050. Dentures become colonized by microbial biofilms, the composition of which is influenced by complex factors such as patient’s age and health, and the nature of the denture material. Since colonization (and subsequent biofilm formation) by some micro‐organisms can significantly impact the health of the denture wearer, the study of denture microbiology has long been of interest to researchers. The specific local and systemic health risks of denture plaque are different from those of dental plaque, particularly with respect to the presence of the opportunist pathogen Candida albicans and various other nonoral opportunists. Here, we reflect on advancements in our understanding of the relationship between micro‐organisms, dentures, and the host, and highlight how our growing knowledge of the microbiome, biofilms, and novel antimicrobial technologies may better inform diagnosis, treatment, and prevention of denture‐associated infections, thereby enhancing the quality and longevity of denture wearers.
Collapse
Affiliation(s)
- J Redfern
- Department of Natural Sciences, Faculty of Science and Engineering Manchester Metropolitan University UK
| | - L Tosheva
- Department of Natural Sciences, Faculty of Science and Engineering Manchester Metropolitan University UK
| | - S Malic
- Department of Life Sciences, Faculty of Science and Engineering Manchester Metropolitan University UK
| | - M Butcher
- Department of Oral Sciences, Glasgow Dental School, School of Medicine, Dentistry and Nursing University of Glasgow UK
| | - G Ramage
- Department of Oral Sciences, Glasgow Dental School, School of Medicine, Dentistry and Nursing University of Glasgow UK
| | - J Verran
- Department of Life Sciences, Faculty of Science and Engineering Manchester Metropolitan University UK
| |
Collapse
|
38
|
Li H, Dong M, Xie W, Qi W, Teng F, Li H, Yan Y, Wang C, Han C, Xue F. Mixed Vaginitis in the Third Trimester of Pregnancy Is Associated With Adverse Pregnancy Outcomes: A Cross-Sectional Study. Front Cell Infect Microbiol 2022; 12:798738. [PMID: 35419297 PMCID: PMC8995747 DOI: 10.3389/fcimb.2022.798738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/03/2022] [Indexed: 12/03/2022] Open
Abstract
Mixed vaginitis is a complex vaginal dysbiosis that differs from single vaginitis. Vaginitis in the third trimester may lead to adverse maternal and neonatal outcomes. The clinical characteristics, microbiological characteristics, and adverse pregnancy outcomes of mixed vaginitis in late pregnancy are worth studying. Therefore, this study investigated the clinical and microbiological characteristics of vaginitis and adverse pregnancy outcomes of patients with mixed vaginitis. We studied 1,674 women in late pregnancy who attended the Tianjin Medical University General Hospital from November, 2019 to October, 2021. We administered standardized questionnaires, performed vaginal examination and sampling plus microscope examinations, and assessed follow-up pregnancy outcomes. We cultured the vaginal discharge of the patients with mixed vaginitis to isolate pathogens and performed antimicrobial susceptibility tests of the isolated pathogens. For the patients with peripartum infection, we collected a sample to isolate pathogens. Among the 1,674 women, 66 (3.9%) had mixed vaginitis. The independent risk factor for mixed vaginitis in late pregnancy was a history of vaginitis during early and middle pregnancy (OR = 5.637, 95% CI: 3.314-9.580). The signs of vaginal erythema (63.6% vs. 42.0%), yellow discharge (81.8% vs. 59.6%), and malodor (31.8% vs. 18.8%) (P <0.05) were significantly higher in patients with mixed vaginitis than in patients with single vaginitis. Bacterial isolates of the vaginal secretions of patients with mixed bacterial vaginitis were mainly the pathogens of aerobic vaginitis and bacterial vaginosis, such as Gardnerella vaginalis, Streptococcus anginosus, and Staphylococcus epidermidis. Pathogen isolation of the vaginal secretions of patients with mixed fungus and bacteria vaginitis mainly included Candida albicans, followed by S. anginosus, Enterococcus faecalis, Staphylococcus hemolyticus, Staphylococcus aureus, Streptococcus agalactiae and Staphylococcus simulans. Women with mixed vaginitis had an increased incidence and risk of peripartum infections (6.1% vs. 1.4%, P <0.05; OR = 3.985, 95% CI:1.214-13.079). Escherichia coli is the main pathogen that causes peripartum infection. Mixed vaginitis in late pregnancy is characterized by a severe and complex phenotype, complex vaginal dysbiosis, and a long course of vaginal dysbiosis. This can lead to an increased incidence and risk of peripartum infection. Therefore, more attention should be paid to patients with mixed vaginitis in the third trimester of pregnancy.
Collapse
Affiliation(s)
- Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengting Dong
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenjuan Xie
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenhui Qi
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiyang Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
39
|
Pinto M, Borges V, Nascimento M, Martins F, Pessanha MA, Faria I, Rodrigues J, Matias R, Gomes JP, Jordao L. Insights on catheter-related bloodstream infections: a prospective observational study on the catheter colonization and multi-drug resistance. J Hosp Infect 2022; 123:43-51. [DOI: 10.1016/j.jhin.2022.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022]
|
40
|
Khan F, Oh D, Chandika P, Jo DM, Bamunarachchi NI, Jung WK, Kim YM. Inhibitory activities of phloroglucinol-chitosan nanoparticles on mono- and dual-species biofilms of Candida albicans and bacteria. Colloids Surf B Biointerfaces 2021; 211:112307. [PMID: 34971906 DOI: 10.1016/j.colsurfb.2021.112307] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 12/02/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022]
Abstract
Phloroglucinol (PG) was encapsulated into chitosan nanoparticles (CSNPs) using a simple ionic gelification technique, and the inhibitory activity of the resulting nanoparticles on microbial mono- and dual-species biofilms was investigated. PG-CSNPs were determined to be spherical with a rough surface, and had an average diameter and zeta potential of 414.0 ± 48.5 nm and 21.1 ± 1.2 mV, respectively. The rate of PG release from the loaded CSNPs was found to increase in acidic environment. The loading capacity and encapsulation efficiency of PG to CSNPs were determined to be 18.74% and 22.4%, respectively. The prepared PG-CSNPs exhibited inhibitory effects on mono-species biofilms such as Candida albicans, Staphylococcus aureus, Klebsiella pneumoniae, Streptococcus mutans, and dual-species such as C. albicans-K. pneumoniae/S. aureus/S. mutans. The PG-CSNPs were found to be more effective in inhibiting and eradicating mono- and dual-species biofilms than pure PG. In addition, PG-CSNPs were found to enhance the efficacy of several antimicrobial drugs against mature mono- and dual-species biofilms. This work demonstrates that PG-CSNPs may provide an alternative method for treating infections caused by biofilm-forming pathogens.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Dokyung Oh
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Pathum Chandika
- Department of Biomedical Engineering, and New-senior Healthcare Innovation Center (BK21 Plus) Pukyong National University, Busan 48513, Republic of Korea
| | - Du-Min Jo
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | | | - Won-Kyo Jung
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Biomedical Engineering, and New-senior Healthcare Innovation Center (BK21 Plus) Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
41
|
Enhanced Virulence of Candida albicans by Staphylococcus aureus: Evidence in Clinical Bloodstream Infections and Infected Zebrafish Embryos. J Fungi (Basel) 2021; 7:jof7121099. [PMID: 34947081 PMCID: PMC8706905 DOI: 10.3390/jof7121099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
Coinfection with Candida and Staphylococcus results in higher mortality in animal studies. However, the pathogenesis and interplay between C. albicans and S. aureus in bloodstream infections (BSIs) is unclear. This study determines the clinical features and outcomes of mixed C. albicans/S. aureus (CA/SA) BSIs and biofilm formation on pathogenesis during coinfection. Demographics and outcomes for mixed BSIs and monomicrobial candidemia were compared. Compared to 115 monomicrobial C. albicans BSIs, 22 patients with mixed CA/SA BSIs exhibited a significantly higher mortality rate and shorter survival time. In vitro and in vivo biofilm analysis showed that C. albicans accounted for the main biofilm architecture, and S. aureus increased its amount. Antibiotic tolerance in S. aureus, which adhered to Candida hyphae observed by scanning electron microscope, was demonstrated by the presence of wild-type C. albicans co-biofilm. Upregulation in exotoxin genes of S. aureus was evidenced by quantitative RT-PCR when a co-biofilm was formed with C. albicans. Mixed CA/SA BSIs result in a higher mortality rate in patients and in vivo surrogate models experiments. This study demonstrates that the virulence enhancement of C. albicans and S. aureus during co-biofilm formation contributes to the high mortality rate.
Collapse
|
42
|
Interplay between Candida albicans and Lactic Acid Bacteria in the Gastrointestinal Tract: Impact on Colonization Resistance, Microbial Carriage, Opportunistic Infection, and Host Immunity. Clin Microbiol Rev 2021; 34:e0032320. [PMID: 34259567 PMCID: PMC8404691 DOI: 10.1128/cmr.00323-20] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Emerging studies have highlighted the disproportionate role of Candida albicans in influencing both early community assembly of the bacterial microbiome and dysbiosis during allergic diseases and intestinal inflammation. Nonpathogenic colonization of the human gastrointestinal (GI) tract by C. albicans is common, and the role of this single fungal species in modulating bacterial community reassembly after broad-spectrum antibiotics can be readily recapitulated in mouse studies. One of the most notable features of C. albicans-associated dysbiotic states is a marked change in the levels of lactic acid bacteria (LAB). C. albicans and LAB share metabolic niches throughout the GI tract, and in vitro studies have identified various interactions between these microbes. The two predominant LAB affected are Lactobacillus species and Enterococcus species. Lactobacilli can antagonize enterococci and C. albicans, while Enterococcus faecalis and C. albicans have been reported to exhibit a mutualistic relationship. E. faecalis and C. albicans are also causative agents of a variety of life-threatening infections, are frequently isolated together from mixed-species infections, and share certain similarities in clinical presentation-most notably their emergence as opportunistic pathogens following disruption of the microbiota. In this review, we discuss and model the mechanisms used by Lactobacillus species, E. faecalis, and C. albicans to modulate each other's growth and virulence in the GI tract. With multidrug-resistant E. faecalis and C. albicans strains becoming increasingly common in hospital settings, examining the interplay between these three microbes may provide novel insights for enhancing the efficacy of existing antimicrobial therapies.
Collapse
|
43
|
Short B, Delaney C, McKloud E, Brown JL, Kean R, Litherland GJ, Williams C, Martin SL, MacKay WG, Ramage G. Investigating the Transcriptome of Candida albicans in a Dual-Species Staphylococcus aureus Biofilm Model. Front Cell Infect Microbiol 2021; 11:791523. [PMID: 34888261 PMCID: PMC8650683 DOI: 10.3389/fcimb.2021.791523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/27/2021] [Indexed: 11/27/2022] Open
Abstract
Candida albicans is an opportunistic pathogen found throughout multiple body sites and is frequently co-isolated from infections of the respiratory tract and oral cavity with Staphylococcus aureus. Herein we present the first report of the effects that S. aureus elicits on the C. albicans transcriptome. Dual-species biofilms containing S. aureus and C. albicans mutants defective in ALS3 or ECE1 were optimised and characterised, followed by transcriptional profiling of C. albicans by RNA-sequencing (RNA-seq). Altered phenotypes in C. albicans mutants revealed specific interaction profiles between fungus and bacteria. The major adhesion and virulence proteins Als3 and Ece1, respectively, were found to have substantial effects on the Candida transcriptome in early and mature biofilms. Despite this, deletion of ECE1 did not adversely affect biofilm formation or the ability of S. aureus to interact with C. albicans hyphae. Upregulated genes in dual-species biofilms corresponded to multiple gene ontology terms, including those attributed to virulence, biofilm formation and protein binding such as ACE2 and multiple heat-shock protein genes. This shows that S. aureus pushes C. albicans towards a more virulent genotype, helping us to understand the driving forces behind the increased severity of C. albicans-S. aureus infections.
Collapse
Affiliation(s)
- Bryn Short
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom.,School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| | - Christopher Delaney
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| | - Emily McKloud
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| | - Jason L Brown
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| | - Ryan Kean
- Glasgow Biofilms Research Network, Glasgow, United Kingdom.,Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Gary J Litherland
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - Craig Williams
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| | - S Lorraine Martin
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Ireland
| | - William G MacKay
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| | - Gordon Ramage
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom.,Glasgow Biofilms Research Network, Glasgow, United Kingdom
| |
Collapse
|
44
|
Eichelberger KR, Cassat JE. Metabolic Adaptations During Staphylococcus aureus and Candida albicans Co-Infection. Front Immunol 2021; 12:797550. [PMID: 34956233 PMCID: PMC8692374 DOI: 10.3389/fimmu.2021.797550] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/19/2021] [Indexed: 12/21/2022] Open
Abstract
Successful pathogens require metabolic flexibility to adapt to diverse host niches. The presence of co-infecting or commensal microorganisms at a given infection site can further influence the metabolic processes required for a pathogen to cause disease. The Gram-positive bacterium Staphylococcus aureus and the polymorphic fungus Candida albicans are microorganisms that asymptomatically colonize healthy individuals but can also cause superficial infections or severe invasive disease. Due to many shared host niches, S. aureus and C. albicans are frequently co-isolated from mixed fungal-bacterial infections. S. aureus and C. albicans co-infection alters microbial metabolism relative to infection with either organism alone. Metabolic changes during co-infection regulate virulence, such as enhancing toxin production in S. aureus or contributing to morphogenesis and cell wall remodeling in C. albicans. C. albicans and S. aureus also form polymicrobial biofilms, which have greater biomass and reduced susceptibility to antimicrobials relative to mono-microbial biofilms. The S. aureus and C. albicans metabolic programs induced during co-infection impact interactions with host immune cells, resulting in greater microbial survival and immune evasion. Conversely, innate immune cell sensing of S. aureus and C. albicans triggers metabolic changes in the host cells that result in an altered immune response to secondary infections. In this review article, we discuss the metabolic programs that govern host-pathogen interactions during S. aureus and C. albicans co-infection. Understanding C. albicans-S. aureus interactions may highlight more general principles of how polymicrobial interactions, particularly fungal-bacterial interactions, shape the outcome of infectious disease. We focus on how co-infection alters microbial metabolism to enhance virulence and how infection-induced changes to host cell metabolism can impact a secondary infection.
Collapse
Affiliation(s)
- Kara R. Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- *Correspondence: Kara R. Eichelberger, ; James E. Cassat,
| | - James E. Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, United States
- *Correspondence: Kara R. Eichelberger, ; James E. Cassat,
| |
Collapse
|
45
|
Scheunemann G, Fortes BN, Lincopan N, Ishida K. Caspofungin Inhibits Mixed Biofilms of Candida albicans and Methicillin-Resistant Staphylococcus aureus and Displays Effectiveness in Coinfected Galleria mellonella Larvae. Microbiol Spectr 2021; 9:e0074421. [PMID: 34643410 PMCID: PMC8515925 DOI: 10.1128/spectrum.00744-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/01/2021] [Indexed: 01/05/2023] Open
Abstract
Candida albicans and Staphylococcus aureus are pathogens commonly isolated from bloodstream infections worldwide. While coinfection by both pathogens is associated with mixed biofilms and more severe clinical manifestations, due to the combined expression of virulence and resistance factors, effective treatments remain a challenge. In this study, we evaluated the activity of echinocandins, especially caspofungin, against mixed biofilms of C. albicans and methicillin-resistant (MRSA) or methicillin-susceptible S. aureus (MSSA) and their effectiveness in vivo using the Galleria mellonella coinfection model. Although caspofungin (CAS) and micafungin (MFG) inhibited the mixed biofilm formation, with CAS exhibiting inhibitory activity at lower concentrations, only CAS was active against preformed mixed biofilms. CAS significantly decreased the total biomass of mixed biofilms at concentrations of ≥2 μg/ml, whereas the microbial viability was reduced at high concentrations (32 to 128 μg/ml), leading to fungus and bacterium cell wall disruption and fungal cell enlargement. Notably, CAS (20 or 50 mg/kg of body weight) treatment led to an increased survival and improved outcomes of G. mellonella larvae coinfected with C. albicans and MRSA, since a significant reduction of fungal and bacterial burden in larval tissues was achieved with induction of granuloma formation. Our results reveal that CAS can be a therapeutic option for the treatment of mixed infections caused by C. albicans and S. aureus, supporting additional investigation. IMPORTANCE Infections by microorganisms resistant to antimicrobials is a major challenge that leads to high morbidity and mortality rates and increased time and cost with hospitalization. It was estimated that 27 to 56% of bloodstream infections by C. albicans are polymicrobial, with S. aureus being one of the microorganisms commonly coisolated worldwide. About 80% of infections are associated with biofilms by single or mixed species that can be formed on invasive medical devices, e.g., catheter, and are considered a dissemination source. The increased resistance to antimicrobials in bacterial and fungal cells when they are in biofilms is the most medically relevant behavior that frequently results in therapeutic failure. Although there are several studies evaluating treatments for polymicrobial infections associated or not with biofilms, there is still no consensus on an effective antimicrobial therapy to combat the coinfection by bacteria and fungi.
Collapse
Affiliation(s)
- Gaby Scheunemann
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruna N. Fortes
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nilton Lincopan
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Kelly Ishida
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Dias KDC, Barbugli PA, Vergani CE. Insights into the activation of oral keratinocyte cell death by Candida albicans and Staphylococcus aureus biofilms. BIOFOULING 2021; 37:975-983. [PMID: 34708675 DOI: 10.1080/08927014.2021.1994959] [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: 04/16/2021] [Revised: 09/30/2021] [Accepted: 10/14/2021] [Indexed: 06/13/2023]
Abstract
Polymicrobial biofilms comprising Candida albicans and Staphylococcus aureus can increase the frequency and severity of oral diseases. This study assessed oral keratinocyte cell death, apoptosis and/or necrosis, promoted by soluble factors from single and dual biofilms of S. aureus and C. albicans. The soluble factors were obtained from the 16-h biofilm growth media. Cell viability was assessed by MTT and cell membrane damage by LDH. SEM was used for morphology changes. Assessment of apoptosis and necrosis was performed using annexin V and propidium iodide and caspases -2, -3, -6, -8 and -9. Statistical analysis was conducted with ANOVA and Tukey tests (α = 5%). Dual biofilms promoted the greatest harmful effect on oral cells, with a viability rate of 31.76%, damage to cell membranes and LDH released. Dual biofilms also induced higher percentages of necrotic cells (24.95%). Apoptosis was associated with caspases -2, -3, -6 and -8 activation.
Collapse
Affiliation(s)
- Kassia de Carvalho Dias
- Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Paula Aboud Barbugli
- Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Carlos Eduardo Vergani
- Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
47
|
Fatahi Dehpahni M, Chehri K, Azadbakht M. Effect of Silver Nanoparticles and L-Carnitine Supplement on Mixed Vaginitis Caused by Candida albicans/ Staphylococcus aureus in Mouse Models: An Experimental Study. Curr Microbiol 2021; 78:3945-3956. [PMID: 34542662 DOI: 10.1007/s00284-021-02652-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 09/02/2021] [Indexed: 12/01/2022]
Abstract
The evolution of antimicrobial-resistant pathogens is a global health and development threat. Nanomedicine is rapidly becoming the main driving force behind ongoing changes in antimicrobial studies. Among nanoparticles, silver (AgNPs) have attracted attention due to their versatile properties. The study aimed to investigate the effects of AgNPs and L-carnitine (LC) on mixed Candida albicans and Staphylococcus aureus in the mice vaginitis model. Study of antimicrobial activity of AgNPs evaluated by Minimum Inhibitory Concentration (MIC) and Minimum Biocidal Concentration (MBC) assays. AgNPs inhibited biofilm formation of microbial strains, which was tested by using crystal violet staining. In the current study, we evaluated the effects of AgNPs and LC in NMRI mice infected intravaginally with C. albicans/ S. aureus for two weeks. The proportion of mice in each stage of the estrous cycle (proestrus, estrus, metestrus, and diestrus) was examined. Histological properties were assessed by hematoxylin/ eosin (H&E) staining of formalin-fixed, paraffin-embedded vaginal tissue sections. Based on the results, MICs of AgNPs against S. aureus, C. albicans, and their combination were 252.3, 124.8, and 501.8 ppm, and their minimum biofilm inhibitory concentration (MBIC) was 500, 250, and 1000 ppm, respectively. The estrous cycle in the treated group was similar to the control. Vaginal histology and cytology showed that LC can improve tissue damages caused by vaginitis and AgNPs. This study demonstrates the promising use of AgNPs as antimicrobial agents and the combination of AgNPs/ LC could be a great future alternative in the control of vaginitis.
Collapse
Affiliation(s)
| | - Khosrow Chehri
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran.
| | - Mehri Azadbakht
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
48
|
Effects of lipid emulsions on the formation of Escherichia coli-Candida albicans mixed-species biofilms on PVC. Sci Rep 2021; 11:16929. [PMID: 34413406 PMCID: PMC8376934 DOI: 10.1038/s41598-021-96385-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Patients receiving lipid emulsions are at increased risk of contracting catheter-related bloodstream infections (CRBSIs) in the clinic. More than 15% of CRBSIs are polymicrobial. The objective of this study was to explore the effects of lipid emulsions on the formation of Escherichia coli (E. coli)–Candida albicans (C. albicans) mixed-species biofilms (BFs) on polyvinyl chloride (PVC) surfaces and the underlying mechanism. Mixed-species BFs were produced by coculturing E. coli and C. albicans with PVC in various concentrations of lipid emulsions. Crystal violet staining and XTT assays were performed to test the mixed-species BF biomass and the viability of microbes in the BFs. The microstructures of the BFs were observed by an approach that combined confocal laser scanning microscopy, fluorescence in situ hybridization, and scanning electron microscopy. The study found that lipid emulsions could promote the formation of E. coli–C. albicans mixed-species BFs, especially with 10% lipid emulsions. The mechanism by which lipid emulsions promote mixed-species BF formation may involve significant upregulation of the expression of the flhDC, iha, HTA1, and HWP1 genes, which are associated with bacterial motility, adhesion, and BF formation. The results derived from this study necessitate strict aseptic precautions when handling lipid emulsions and avoiding the use of high concentrations of lipid emulsions for as long as possible.
Collapse
|
49
|
Hu Y, Niu Y, Ye X, Zhu C, Tong T, Zhou Y, Zhou X, Cheng L, Ren B. Staphylococcus aureus Synergized with Candida albicans to Increase the Pathogenesis and Drug Resistance in Cutaneous Abscess and Peritonitis Murine Models. Pathogens 2021; 10:pathogens10081036. [PMID: 34451500 PMCID: PMC8398722 DOI: 10.3390/pathogens10081036] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/05/2021] [Accepted: 08/12/2021] [Indexed: 01/21/2023] Open
Abstract
The mixed species of Staphylococcus aureus and Candida albicans can cause infections on skin, mucosa or bloodstream; however, mechanisms of their cross-kingdom interactions related to pathogenesis and drug resistance are still not clear. Here an increase of S. aureus proliferation and biofilm formation was observed in S. aureus and C. albicans dual-species culture, and the synergistic pathogenic effect was then confirmed in both local (cutaneous abscess) and systemic infection (peritonitis) murine models. According to the transcriptome analysis of the dual-species culture, virulence factors of S. aureus were significantly upregulated. Surprisingly, the beta-lactams and vancomycin-resistant genes in S. aureus as well as azole-resistant genes in C. albicans were also significantly increased. The synergistic effects on drug resistance to both antibacterial and antifungal agents were further proved both in vitro and in cutaneous abscess and peritonitis murine models treated by methicillin, vancomycin and fluconazole. The synergistic interactions between S. aureus and C. albicans on pathogenesis and drug resistance highlight the importance of targeting the microbial interactions in polyspecies-associated infections.
Collapse
Affiliation(s)
- Yao Hu
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
| | - Yulong Niu
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610041, China;
| | - Xingchen Ye
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
| | - Chengguang Zhu
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
| | - Ting Tong
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
| | - Yujie Zhou
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
- Correspondence: (X.Z.); (L.C.); (B.R.)
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
- Correspondence: (X.Z.); (L.C.); (B.R.)
| | - Biao Ren
- State Key Laboratory of Oral Diseases, West China School of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610064, China; (Y.H.); (X.Y.); (C.Z.); (T.T.); (Y.Z.)
- Correspondence: (X.Z.); (L.C.); (B.R.)
| |
Collapse
|
50
|
Cheong JZA, Johnson CJ, Wan H, Liu A, Kernien JF, Gibson ALF, Nett JE, Kalan LR. Priority effects dictate community structure and alter virulence of fungal-bacterial biofilms. THE ISME JOURNAL 2021; 15:2012-2027. [PMID: 33558690 PMCID: PMC8245565 DOI: 10.1038/s41396-021-00901-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
Polymicrobial biofilms are a hallmark of chronic wound infection. The forces governing assembly and maturation of these microbial ecosystems are largely unexplored but the consequences on host response and clinical outcome can be significant. In the context of wound healing, formation of a biofilm and a stable microbial community structure is associated with impaired tissue repair resulting in a non-healing chronic wound. These types of wounds can persist for years simmering below the threshold of classically defined clinical infection (which includes heat, pain, redness, and swelling) and cycling through phases of recurrent infection. In the most severe outcome, amputation of lower extremities may occur if spreading infection ensues. Here we take an ecological perspective to study priority effects and competitive exclusion on overall biofilm community structure in a three-membered community comprised of strains of Staphylococcus aureus, Citrobacter freundii, and Candida albicans derived from a chronic wound. We show that both priority effects and inter-bacterial competition for binding to C. albicans biofilms significantly shape community structure on both abiotic and biotic substrates, such as ex vivo human skin wounds. We further show attachment of C. freundii to C. albicans is mediated by mannose-binding lectins. Co-cultures of C. freundii and C. albicans trigger the yeast-to-hyphae transition, resulting in a significant increase in neutrophil death and inflammation compared to either species alone. Collectively, the results presented here facilitate our understanding of fungal-bacterial interactions and their effects on host-microbe interactions, pathogenesis, and ultimately, wound healing.
Collapse
Affiliation(s)
- J Z Alex Cheong
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Chad J Johnson
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Hanxiao Wan
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Aiping Liu
- Department of Surgery, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - John F Kernien
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Angela L F Gibson
- Department of Surgery, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Jeniel E Nett
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Lindsay R Kalan
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA.
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|