1
|
Qian W, Lu J, Wang T, Liu Q, Liu N, Chen S, Li Y. Isobavachalcone confers protection against Cryptococcus neoformans-induced ferroptosis in Caenorhabditis elegans via lifespan extension and GSH-GPX-1 axis modulation. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:137969. [PMID: 40154123 DOI: 10.1016/j.jhazmat.2025.137969] [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: 12/19/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
The recent designation of Cryptococcus neoformans as a critical-priority fungal pathogen by the World Health Organization highlights the imperative need for novel antifungal agents with distinct mechanisms of action. This study elucidates the novel ferroptotic pathway underlying C. neoformans-induced cell death in Caenorhabditis elegans and investigates the therapeutic potential of isobavachalcone (IBC) through comprehensive evaluation of core biochemical markers: total glutathione (GSH), malondialdehyde, ferrous iron content, and lipid reactive oxygen species (ROS). Integrated transcriptomic analysis via RNA-seq and subsequent RT-qPCR validation revealed critical gene expression patterns associated with antiferroptotic regulation. Our findings demonstrate that C. neoformans infection initiates ferroptosis in C. elegans through iron-dependent lipid peroxidation cascades. Remarkably, IBC administration conferred significant protection against fungal-induced ferroptosis by restoring redox homeostasis-evidenced by elevated GSH levels, attenuated ROS accumulation, and decreased ferrous iron content. Mechanistic investigations identified IBC-mediated upregulation of SKN-1 and GSH biosynthesis genes, coupled with suppression of GPX-1 activity. These coordinated effects disrupted the iron-ROS amplification loop through modulation of the GSH-GPX-1 axis, ultimately extending host lifespan in C. neoformans-challenged models. Our results position IBC as a ferroptosis inhibitor with dual antioxidant and iron-chelating properties, offering a therapeutic strategy against cryptococcal infections through targeting of evolutionary conserved cell death pathways.
Collapse
Affiliation(s)
- Weidong Qian
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Jiaxing Lu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Ting Wang
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Qiming Liu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Na Liu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Si Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yongdong Li
- Ningbo Municipal Key Laboratory of Virology, Ningbo Municipal Center for Disease Control and Prevention, Ningbo 315010, China.
| |
Collapse
|
2
|
Zhang X, Yang L, Cai T, Liao H, Cai P, Zhao X, Gu N, Yue Z, Gu Y, Zhang F, Wu C, Lu X. The natural symbiotic bacterium Enterococcus faecalis LX10 drives Bombyx mori refractoriness to Nosema bombycis infection via the secretion of enterococcin. BMC Microbiol 2025; 25:303. [PMID: 40382556 PMCID: PMC12084927 DOI: 10.1186/s12866-025-03980-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND The microsporidian Nosema bombycis is an obligate intracellular fungal-related parasites of the Bombyx mori, causing the epidemic disease Pebrine and extensive economic losses in the agricultural and sericulture industry. Enterococcus has emerged as one of the predominant gut microorganisms of the major model organism, Bombyx mori. However, the potential interactions mechanism between B. mori, N. bombycis and Enterococcus have not been well demonstrated. METHODS To address this gap, we used an insect model, silkworm to examine the potential mechanism of the natural symbiotic bacterium Enterococcus faecalis LX10 drives B. mori refractoriness to N. bombycis infection. E. faecalis LX10 was isolated from the gut of healthy silkworms, and its inhibitory activity against N. bombycis was evaluated at both the cellular and individual levels using posttranslational modifications, gene and protein expression analysis, transfected cells, and in vitro immunofluorescence. RESULTS We demonstrated that enterococcin (EntLX), the first antimicrobial protein family in gut commensal bacterium Enterococcus faecalis LX10 of B. mori, contributes to defending against N. bombycis infection resistance depends on the enzyme gelatinase (GelE), disulfide bond and disulfide bond formation proteinA (DsbA). The EntLX protein, abundantly expressed in transgenic BmN cells and gut organs(gut epithelium, peritrophic membrane and contents), can reduce the infection rate of cells and alleviate intestinal damage caused by N. bombycis infection. After simultaneous vaccination with E. faecalis LX10 and N. bombycis, the differentially key metabolites, physiological characteristics(larval mass), or economic traits(cocoon length, cocoon width, whole-cocoon weight, cocoon shell weight, pupation rate and adult emergence rate) showed a certain degrees of recovery and correction compared with those of single N. bombycis inoculation at the individual level. CONCLUSIONS This study advances the understanding of the anti-microsporidia activity of enterococci and paves the way for the expression of these molecules as antifungal agents via the genetic transformation of Enterococcus symbionts from disease-transmitting insects. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xiancui Zhang
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Liang Yang
- Institute of Sericulture, Huzhou Academy of Agricultural Sciences, Huzhou, 313002, China
| | - Tong Cai
- Shandong Center for Disease Control and Prevention, Jinan, 250014, China
| | - Haoran Liao
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Peiwen Cai
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Xinkuang Zhao
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Nengjie Gu
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Zihan Yue
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Yanzhou Gu
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Fan Zhang
- College of Life Science, Shandong Normal University, Jinan, 250358, China
| | - Choufei Wu
- School of Life Science, Huzhou University, Huzhou, 313000, China.
| | - Xingmeng Lu
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
3
|
de Mello Fiallos N, Zanin dos Santos IJ, Brunson DN, Kajfasz JK, Zeng L, de Aguiar Cordeiro R, Lemos JA, Abranches J. Candida albicans impacts carbohydrate metabolism of Enterococcus faecalis in interkingdom biofilms. J Oral Microbiol 2025; 17:2492194. [PMID: 40247862 PMCID: PMC12004723 DOI: 10.1080/20002297.2025.2492194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Objectives This study investigated the transcriptional and physiological responses of Enterococcus faecalis, an opportunistic pathogen linked to endodontic infections, when cultivated in dual-species biofilms with Candida albicans, a yeast pathobiont found in the oral cavity. Methods Forty-eight-hour E. faecalis OG1RF biofilms were developed in BHI medium as mono- or dual-species with C. albicans SC5314. Biofilms were assessed for biomass, colony-forming units (CFUs), and architecture using confocal microscopy. RNA sequencing was performed on an Illumina platform. Mannose-PTS activity and glycerol quantification assays were conducted to investigate changes in carbohydrate metabolism. Results Transcriptomic analysis revealed 149 E. faecalis genes differentially expressed in dual-species biofilms. Genes linked to mannose-PTS and glycerol metabolism were notably upregulated. Mannose-PTS activity was significantly higher in dual-species biofilms. Mannose, as the sole carbohydrate source, increased E. faecalis CFUs and decreased C. albicans CFUs in co-culture, while glucose had no effect. As C. albicans is a glycerol net producer, glycerol levels were always higher when C. albicans was present, likely contributing to the upregulation of glycerol metabolism genes in E. faecalis when in co-cultures. Conclusions The presence of C. albicans alters E. faecalis gene expression and metabolism, suggesting metabolic crosstalk that may influence their pathogenicity and role in oral infections.
Collapse
Affiliation(s)
- Nicole de Mello Fiallos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
- Department of Pathology and Legal Medicine, Federal University of Ceará, College of Medicine, Fortaleza, Brazil
| | - Iriana J. Zanin dos Santos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
- Department of Cariology and Operative Dentistry, Federal University of Ceará, College of Dentistry, Sobral, Brazil
| | - Debra N. Brunson
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jessica K. Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Rossana de Aguiar Cordeiro
- Department of Pathology and Legal Medicine, Federal University of Ceará, College of Medicine, Fortaleza, Brazil
| | - José A. Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jacqueline Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
4
|
Li L, Qiao Y. Opportunities in exploring chemical biology tools for better strategies against Candida albicans. Curr Opin Chem Biol 2025; 86:102595. [PMID: 40184759 DOI: 10.1016/j.cbpa.2025.102595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 04/07/2025]
Abstract
The growing global prevalence of drug-resistant fungal infections and the scarcity of effective clinical antifungal drugs necessitate an urgent need for new treatments and strategies. In the quest for novel antifungal and anti-virulence compounds and alternative drug targets in fungi, we recognize the significant value of chemical biology tools in guiding these endeavors. Focusing on Candida albicans, the major fungal pathogen in humans, this review explores recent antifungal research efforts that utilize chemical biology tools-such as chemical probes and toolkits-that offer valuable biological insights into the cellular processes of C. albicans. In addition, we discuss the wealth of compounds in the host gut microbiota that naturally influence C. albicans invasive growth in the gut habitat, presenting promising yet underexplored opportunities for developing novel antifungal and anti-virulence strategies. Chemical biology tools are uniquely positioned to unlock the potential of gut microbiota-derived molecules and metabolites in combating C. albicans infections.
Collapse
Affiliation(s)
- Lanxin Li
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technical University 637371, Singapore
| | - Yuan Qiao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technical University 637371, Singapore.
| |
Collapse
|
5
|
Wang Y, Xu W, Guo S, Xu S, Wang J, Zhang S, Kuang Y, Jin P. Enterococci for human health: A friend or foe? Microb Pathog 2025; 201:107381. [PMID: 39983880 DOI: 10.1016/j.micpath.2025.107381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Enterococci are widely distributed in nature and exhibit good temperature and pH tolerance, making them suitable for industrial fermentation. It can produce bacteriocins, natural antibacterial substances utilized in food preservation. Some Enterococci are employed as probiotics to regulate human immunity and maintain healthy intestinal environments. However, recent scientific studies have highlighted the pathogenicity and multidrug resistance of Enterococci, classifying it as an important pathogen in clinical infections. Moreover, increasing evidence has linked Enterococcus sp., particularly Enterococcus faecalis and Enterococcus faecium, to clinical diseases, raising concerns about their safety and posing the question, how should we approach the conflicting nature of the pathogenic and beneficial effects of Enterococci? This review provides the recent advancements in Enterococci research and incorporates the perspectives of international authoritative organizations and institutions to comprehensively analyze the beneficial and harmful characteristics of Enterococci in the fields of science, clinical and industrial applications, aiming to address three important questions: whether Enterococci are beneficial or harmful to humans, their potential use in medical treatments, and the criteria to evaluate their safety. The goal is to explore the feasibility of the standardized use of Enterococci and provide guidance on the scientific selection and utilization of probiotics.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Wenfeng Xu
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Sirui Guo
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Shuo Xu
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Jing Wang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Shanshan Zhang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Yongmei Kuang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China
| | - Pengfei Jin
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, PR China.
| |
Collapse
|
6
|
Abe M, Sekizuka T, Miyazaki Y. Gastrointestinal anaerobes and Enterococcus faecalis promote Candida glabrata gastrointestinal colonization and organ dissemination. J Infect Chemother 2025; 31:102658. [PMID: 39956369 DOI: 10.1016/j.jiac.2025.102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Candida glabrata is a common causative pathogen of endogenous candidiasis. It is assumed that the gastrointestinal flora affects C. glabrata gastrointestinal colonization and organ dissemination in the gastrointestinal tract (GIT). However, no reports have yet described the relationships between C. glabrata and bacteria in the GIT. This study aimed to clarify these relationships using a mouse endogenous candidiasis model with cortisone acetate immunosuppression. METHODS Dysbiosis was induced in the GIT by several antibiotic combinations, and then C. glabrata gastrointestinal colonization and organ dissemination were evaluated. Next, metagenomic sequencing analysis of the gastrointestinal flora was performed to identify bacteria associated with C. glabrata organ dissemination. Finally, coinfection experiments were performed using bacteria isolated from the mouse GIT. RESULTS C. glabrata organ dissemination was significantly promoted using specific antibiotics regardless of the amount of colonization in the GIT. Metagenomic sequencing analysis of the gastrointestinal flora showed that Enterococcus species and anaerobes were significantly associated with enhanced organ dissemination, whereas Enterobacterales, such as Escherichia species and Klebsiella species, were associated with the suppression of organ dissemination. In coinfection experiments, Enterococcus faecalis and Faecalibaculum rodentium inoculation, but not either of them, increased C. glabrata organ dissemination without affecting gastrointestinal colonization. CONCLUSIONS Coinfection with gastrointestinal bacteria promoted C. glabrata organ dissemination, which would indicate that gastrointestinal flora could affect C. glabrata dissemination. Therefore, the gastrointestinal flora could be a target for intervention or treatment in clinical settings. Insights from this study would lead to better control of endogenous candidiasis focusing on the gastrointestinal flora.
Collapse
Affiliation(s)
- Masahiro Abe
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshitsugu Miyazaki
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
7
|
Zhang Y, Wang L, Peng L. The Role of Intestinal Fungi in the Pathogenesis and Treatment of Ulcerative Colitis. Microorganisms 2025; 13:794. [PMID: 40284630 PMCID: PMC12029736 DOI: 10.3390/microorganisms13040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease closely associated with dysbiosis of the gut microbiome, encompassing not only bacterial communities but also fungal populations. Despite the growing recognition of the gut microbiome's role in UC pathogenesis, the contribution of intestinal fungi has only recently garnered significant attention. In this review, we comprehensively examine the characteristics of intestinal fungi in both healthy individuals and UC patients, elucidating their role in disease pathogenesis and their interactions with bacterial communities. Additionally, we explore the impact of intestinal fungi on disease severity and therapeutic responses in UC. Furthermore, we evaluate the therapeutic potential of antifungal agents, probiotics, and fecal microbiota transplantation (FMT) in UC management, emphasizing the critical role of fungi in these treatment modalities. Future research should prioritize elucidating the multifunctional roles of fungi in UC pathogenesis and their implications for treatment strategies. Moreover, the identification of fungal biomarkers associated with FMT efficacy could pave the way for precision medicine approaches in FMT, offering novel insights into personalized therapeutic interventions for UC.
Collapse
Affiliation(s)
- Yujing Zhang
- Microbiota Laboratory, Clinical Division of Microbiota, Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (Y.Z.); (L.W.)
- Medical School of Chinese PLA, Beijing 100853, China
| | - Lin Wang
- Microbiota Laboratory, Clinical Division of Microbiota, Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (Y.Z.); (L.W.)
- Medical School of Chinese PLA, Beijing 100853, China
| | - Lihua Peng
- Microbiota Laboratory, Clinical Division of Microbiota, Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (Y.Z.); (L.W.)
| |
Collapse
|
8
|
Lam LN, Savage KE, Shakir CN, Lemos JA. Genome-wide analysis of Enterococcus faecalis genes that facilitate interspecies competition with Lactobacillus crispatus. J Bacteriol 2025; 207:e0043824. [PMID: 39902966 PMCID: PMC11925238 DOI: 10.1128/jb.00438-24] [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: 10/11/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Enterococci are opportunistic pathogens notorious for causing a variety of infections. While both Enterococcus faecalis and Lactobacillus crispatus are commensal residents of the vaginal tract, the molecular mechanisms that enable E. faecalis to take advantage of a vaginal biome with lower counts of lactobacilli to colonize the vaginal tract and induce aerobic vaginitis remain unknown. Here, we show that L. crispatus eradicates E. faecalis in a contact-independent manner. Using transposon sequencing to identify E. faecalis OG1RF transposon (Tn) mutants that are either under-represented or over-represented when co-cultured with L. crispatus, we found that Tn mutants with disruption in the dltABCD operon, that encodes the proteins responsible for the D-alanylation of teichoic acids, and OG1RF_11697 encoding for an uncharacterized hypothetical protein are more susceptible to killing by L. crispatus. Inversely, Tn mutants with disruption in ldh1, which encodes for L-lactate dehydrogenase, are more resistant to L. crispatus killing. Using the Galleria mellonella infection model, we show that co-injection of L. crispatus with E. faecalis OG1RF enhances larvae survival while this L. crispatus-mediated protection was lost in larvae co-infected with either L. crispatus and E. faecalisΔldh1 or Δldh1Δldh2 strains. Last, using RNA sequencing to identify E. faecalis genes that are differently expressed in the presence of L. crispatus, we found major changes in the expression of genes associated with glycerophospholipid metabolism, central metabolism, and general stress responses. The findings in this study provide insights into how E. faecalis mitigate assaults by L. crispatus.IMPORTANCEEnterococcus faecalis is an opportunistic pathogen notorious for causing a multitude of infections. As vaginal commensals, E. faecalis must interact with Lactobacillus crispatus, but how E. faecalis overcomes or mitigate assaults by L. crispatus killing remains unknown. We show that L. crispatus eradicates E. faecalis temporally in a contact-independent manner. Using high-throughput molecular approaches, we identified genetic determinants that enable E. faecalis to compete with L. crispatus. This study represents an important first step for the identification of adaptive genetic traits required for enterococci to tolerate assaults by lactobacilli.
Collapse
Affiliation(s)
- Ling Ning Lam
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Kathryn E. Savage
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Camille N. Shakir
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - José A. Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
9
|
Zhai J, Nie L, Tian J, He Y, Gu Y. Stress resistance of the biofilm and planktonic forms of Lactobacillus delbrueckii in adverse environments. Lett Appl Microbiol 2025; 78:ovaf031. [PMID: 40053510 DOI: 10.1093/lambio/ovaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/09/2025]
Abstract
Biofilm and planktonic forms are different kinds of self-protection mechanisms in microorganisms for resistance to adverse environments. The research explored the physicochemical properties, antimicrobial, and antioxidant activities of Lactobacillus delbrueckii (L. delbrueckii) 5E, focusing on the biofilm's stress tolerance. L. delbrueckii 5E's high lactic acid production (709.1 g·L-1) enhances its antimicrobial activity, which is pH-dependent and decreases when the supernatant is adjusted to pH 6.5. The cell-free fermentation supernatant, bacterial suspension, and cell-free extract of L. delbrueckii 5E showed significant scavenging activity against 2,2-diphenyl-1-picrylhydrazyl (DPPH), hydroxyl, and superoxide anion radicals. L. delbrueckii 5E formed biofilms with a bacterial count of 9.00 × 106 CFU·mL-1 on polyacrylonitrile electrospun membranes. Stress-tolerance tests indicated that the biofilm form of L. delbrueckii 5E exhibited superior survival under high temperatures, osmotic pressures, bile salts, potassium sorbate, and H2O2 exposure. The biofilm consistently released free bacteria, maintaining a stable total colony count of 106 CFU·mL-1. The remarkable antibacterial and antioxidant properties of L. delbrueckii 5E, along with its resilience to harsh environments, establish its potential for applications in the food industry.
Collapse
Affiliation(s)
- Jiangyang Zhai
- College of Food Science and Engineering, Inner Mongolia Agricultural University, 306 Zhaowuda Road, Hohhot, Inner Mongolia, 010018, China
| | - Lili Nie
- College of Food Science and Engineering, Inner Mongolia Agricultural University, 306 Zhaowuda Road, Hohhot, Inner Mongolia, 010018, China
| | - Jianjun Tian
- College of Food Science and Engineering, Inner Mongolia Agricultural University, 306 Zhaowuda Road, Hohhot, Inner Mongolia, 010018, China
| | - Yinfeng He
- College of Food Science and Engineering, Inner Mongolia Agricultural University, 306 Zhaowuda Road, Hohhot, Inner Mongolia, 010018, China
| | - Yue Gu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, 306 Zhaowuda Road, Hohhot, Inner Mongolia, 010018, China
| |
Collapse
|
10
|
Veerapandian R, Paudyal A, Schneider SM, Lee STM, Vediyappan G. A mouse model of immunosuppression facilitates oral Candida albicans biofilms, bacterial dysbiosis and dissemination of infection. Front Cell Infect Microbiol 2025; 14:1467896. [PMID: 39902181 PMCID: PMC11788080 DOI: 10.3389/fcimb.2024.1467896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/26/2024] [Indexed: 02/05/2025] Open
Abstract
Opportunistic pathogens are a major threat to people, especially those with impaired immune systems. Two of the most important microbes in this category are the fungus Candida albicans and Gram-positive bacteria of the genus Enterococcus, which share overlapping niches in the oral cavity, gastrointestinal and urogenital tracts. The clinical importance of oral C. albicans biofilm and its interaction with the host under immunosuppressive conditions remains largely understudied. Here, we used a mouse model of oropharyngeal candidiasis (OPC) with cortisone acetate injection on alternate days and a continuous supply of C. albicans in drinking water for three days, resulting in immunosuppression. Results showed abundant growth of resident oral bacteria and a strong C. albicans biofilm on the tongue consisting of hyphae which damaged papillae, the epidermal layer, and invaded tongue tissue with the accumulation of inflammatory cells as demonstrated by Grocott's methenamine silver and hematoxylin and eosin staining, respectively. The dispersed microbes from the oral biofilm colonized the gastrointestinal (GI) tract and damaged its integrity, disseminating microbes to other organs. Although no visible damage was observed in the kidney and liver, except increased lipid vacuoles in the liver cells, C. albicans was found in the liver homogenate. Intriguingly, we found co-occurrence of Enterococcus faecalis in the tongue, liver, and stool of immunosuppressed control and C. albicans infected organs. Targeted 16S rRNA and ITS2 amplicon sequencing of microbes from the fecal samples of mice confirmed the above results in the stool samples and revealed an inverse correlation of beneficial microbes in the dysbiosis condition. Our study shows that mucosal-oral infection of C. albicans under immunosuppressed conditions causes tissue damage and invasion in local and distant organs; the invasion may be aided by the overgrowth of the resident endogenous Enterobacteriaceae and other members, including the opportunistic pathogen Enterococcus faecalis.
Collapse
Affiliation(s)
- Raja Veerapandian
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Anuja Paudyal
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sarah M. Schneider
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Sonny T. M. Lee
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Govindsamy Vediyappan
- Division of Biology, Kansas State University, Manhattan, KS, United States
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
11
|
Srihawan O, Panichuttra A, Lertchirakarn V, Matangkasombut O. Efficacy of chitosan root canal medicament against cross-kingdom dual-species biofilm of Candida albicans and Enterococcus faecalis in an in vitro root-canal model. Odontology 2024:10.1007/s10266-024-01024-x. [PMID: 39540969 DOI: 10.1007/s10266-024-01024-x] [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: 09/17/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Infection of the root canals with cross-kingdom biofilm of Candida albicans (C. albicans) and Enterococcus faecalis (E. faecalis) causes more inflammation and is resistant to conventional medication. This study aimed to evaluate the efficacy of chitosan paste (chitosan + propylene glycol (PG)) against this dual-species biofilm in comparison to calcium hydroxide (Ca(OH)2) in the root canals of human teeth. Fifty extracted single-rooted human teeth were prepared and inoculated with C. albicans and E. faecalis, and incubated for 3 days for biofilm formation. Four experimental groups (N = 10 each) were applied with different root canal medications for 7 days, including (1) No medication (negative control); (2) Ca(OH)2 + distilled water; (3) PG (vehicle control) and (4) chitosan + PG. The efficacy of root canal medicaments was assessed by determining the percentage of remaining colony-forming units (CFUs) of viable microorganisms in intracanal dentin. The dentin was collected, serially diluted, and spread on solid media for colony count. The biofilm structure in root canals at 3 days and 10 days (ie. before and after treatments) were observed with scanning electron microscopy (SEM) at 5000× and 10,000× magnifications. Data were analyzed using Welch's ANOVA and Games-Howell post-hoc test (α = 5%). The results show that Chitosan + PG group had a significantly lower percentage of remaining microorganisms when compared to PG, Ca(OH)2, and negative control groups (P-value = 0.015, 0.005, and < 0.001, respectively). Consistently, SEM showed the lowest amount of remaining biofilm in Chitosan + PG group. Thus, Chitosan + PG paste was the most efficacious root canal medicament against dual-species biofilm of C. albicans and E. faecalis in this model.
Collapse
Affiliation(s)
- Ornuma Srihawan
- Department of Operative Dentistry, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Patumwan, Bangkok, 10330, Thailand
| | - Anchana Panichuttra
- Department of Operative Dentistry, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Patumwan, Bangkok, 10330, Thailand.
| | - Veera Lertchirakarn
- Department of Microbiology and Center of Excellence On Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Patumwan, Bangkok, 10330, Thailand
| | - Oranart Matangkasombut
- Department of Microbiology and Center of Excellence On Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Patumwan, Bangkok, 10330, Thailand.
- Research Laboratory of Biotechnology, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Laksi, Bangkok, 10120, Thailand.
| |
Collapse
|
12
|
Brown GD, Ballou ER, Bates S, Bignell EM, Borman AM, Brand AC, Brown AJP, Coelho C, Cook PC, Farrer RA, Govender NP, Gow NAR, Hope W, Hoving JC, Dangarembizi R, Harrison TS, Johnson EM, Mukaremera L, Ramsdale M, Thornton CR, Usher J, Warris A, Wilson D. The pathobiology of human fungal infections. Nat Rev Microbiol 2024; 22:687-704. [PMID: 38918447 DOI: 10.1038/s41579-024-01062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
Human fungal infections are a historically neglected area of disease research, yet they cause more than 1.5 million deaths every year. Our understanding of the pathophysiology of these infections has increased considerably over the past decade, through major insights into both the host and pathogen factors that contribute to the phenotype and severity of these diseases. Recent studies are revealing multiple mechanisms by which fungi modify and manipulate the host, escape immune surveillance and generate complex comorbidities. Although the emergence of fungal strains that are less susceptible to antifungal drugs or that rapidly evolve drug resistance is posing new threats, greater understanding of immune mechanisms and host susceptibility factors is beginning to offer novel immunotherapeutic options for the future. In this Review, we provide a broad and comprehensive overview of the pathobiology of human fungal infections, focusing specifically on pathogens that can cause invasive life-threatening infections, highlighting recent discoveries from the pathogen, host and clinical perspectives. We conclude by discussing key future challenges including antifungal drug resistance, the emergence of new pathogens and new developments in modern medicine that are promoting susceptibility to infection.
Collapse
Affiliation(s)
- Gordon D Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Elizabeth R Ballou
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Steven Bates
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elaine M Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Andrew M Borman
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alexandra C Brand
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Carolina Coelho
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Peter C Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rhys A Farrer
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Nelesh P Govender
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - William Hope
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - J Claire Hoving
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rachael Dangarembizi
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Thomas S Harrison
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elizabeth M Johnson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Liliane Mukaremera
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark Ramsdale
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
13
|
Gong M, Yu H, Qu H, Li Z, Liu D, Zhao X. Global research trends and hotspots on human intestinal fungi and health: a bibliometric visualization study. Front Cell Infect Microbiol 2024; 14:1460570. [PMID: 39483119 PMCID: PMC11525014 DOI: 10.3389/fcimb.2024.1460570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
Background This article employs bibliometric methods and visual maps to delineate the research background, collaborative relationships, hotspots, and trends in the study of gut fungi in human diseases and health. Methods Publications related to human gut fungi were retrieved from the Web of Science Core Collection. VOSviewer, CiteSpace, R software and Microsoft Excel were employed to generate visual representations illustrating the contributions made by countries/regions, authors, organizations, and journals. Employing VOSviewer and CiteSpace, we conducted a comprehensive analysis of the retrieved publications, revealing underlying tendencies, research hotspots, and intricate knowledge networks. Results This study analyzed a total of 3,954 publications. The United States ranks first in the number of published papers and has the highest number of citations and h-index. Mostafa S Elshahed is the most prolific author. The University of California System is the institution that published the most papers. Frontiers In Microbiology is the journal with the largest number of publications. Three frequently co-cited references have experienced a citation burst lasting until 2024. Conclusion Advancements in sequencing technologies have intensified research into human gut fungi and their health implications, shifting the research focus from gut fungal infections towards microbiome science. Inflammatory bowel diseases and Candida albicans have emerged as pivotal areas of interest in this endeavor. Through this study, we have gained a deeper insight into global trends and frontier hotspots within this field, thereby enhancing our understanding of the intricate relationship between gut fungi and human health.
Collapse
Affiliation(s)
- Ming Gong
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Huiying Yu
- Laboratory of Basic Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Hong Qu
- Bidding and Procurement Office, The Second Hospital of Dalian Medical University, Dalian, China
| | - Zhexian Li
- Dalian Medical University, Dalian, China
| | - Di Liu
- First Clinical Faculty, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xin Zhao
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Crouch AL, Monsey L, Rambeau M, Ramos C, Yracheta JM, Anderson MZ. Metagenomic discovery of microbial eukaryotes in stool microbiomes. mBio 2024; 15:e0206324. [PMID: 39207108 PMCID: PMC11481512 DOI: 10.1128/mbio.02063-24] [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: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Host-associated microbiota form complex microbial communities that are increasingly associated with host behavior and disease. While these microbes include bacterial, archaeal, viral, and eukaryotic constituents, most studies have focused on bacteria due to their dominance in the human host and available tools for investigation. Accumulating evidence suggests microbial eukaryotes in the microbiome play pivotal roles in host health, but our understandings of these interactions are limited to a few readily identifiable taxa because of technical limitations in unbiased eukaryote exploration. Here, we combined cell sorting, optimized eukaryotic cell lysis, and shotgun sequencing to accelerate metagenomic discovery and analysis of host-associated microbial eukaryotes. Using synthetic communities with a 1% microbial eukaryote representation, the eukaryote-optimized cell lysis and DNA recovery method alone yielded a 38-fold increase in eukaryotic DNA. Automated sorting of eukaryotic cells from stool samples of healthy adults increased the number of microbial eukaryote reads in metagenomic pools by up to 28-fold compared to commercial kits. Read frequencies for identified fungi increased by 10,000× on average compared to the Human Microbiome Project and allowed for the identification of novel taxa, de novo assembly of contigs from previously unknown microbial eukaryotes, and gene prediction from recovered genomic segments. These advances pave the way for the unbiased inclusion of microbial eukaryotes in deciphering determinants of health and disease in the host-associated microbiome.IMPORTANCEMicrobial eukaryotes are common constituents of the human gut where they can contribute to local ecology and host health, but they are often overlooked in microbiome studies. The lack of attention is due to current technical limitations that are heavily biased or poorly recovered DNA from microbial eukaryotes. We developed a method to increase the representation of these eukaryotes in metagenomic sequencing of microbiome samples that allows to improve their detection compared to prior methods and allows for the identification of new species. Application of the technique to gut microbiome samples improved detection of fungi, protists, and helminths. New eukaryotic taxa and their encoded genes could be identified by sequencing a small number of samples. This approach can improve the inclusion of eukaryotes into microbiome research.
Collapse
Affiliation(s)
- Audra L. Crouch
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Laine Monsey
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Molly Rambeau
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Cameron Ramos
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | | | - Matthew Z. Anderson
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
- Center for Genomic Science Innovation, University of Wisconsin - Madison, Madison, Wisconsin, USA
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| |
Collapse
|
15
|
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
|
16
|
Fong JL, Ong Eng Yong V, Yeo C, Adamson C, Li L, Zhang D, Qiao Y. Biochemical Characterization of Recombinant Enterococcus faecalis EntV Peptide to Elucidate Its Antihyphal and Antifungal Mechanisms against Candida albicans. ACS Infect Dis 2024; 10:3408-3418. [PMID: 39137394 DOI: 10.1021/acsinfecdis.4c00515] [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: 08/15/2024]
Abstract
Candida albicans is a common opportunistic fungus in humans, whose morphological switch between yeast and hyphae forms represents a key virulence trait. Developing strategies to inhibit C. albicans hyphal growth may provide insights into designs of novel antivirulent therapeutics. Importantly, the gut commensal bacterium, Enterococcus faecalis, secretes a bacteriocin EntV which has potent antivirulent and antifungal effects against C. albicans in infection models; however, hampered by the challenges to access large quantities of bioactive EntV, the detailed understanding of its mechanisms on C. albicans has remained elusive. In this work, we biochemically reconstituted the proteolytic cleavage reaction to obtain recombinant EntV88-His6 on a large preparative scale, providing facile access to the C-terminal EntV construct. Under in vitro C. albicans hyphal assay with specific inducers, we demonstrated that EntV88-His6 exhibits potent bioactivity against GlcNAc-triggered hyphal growth. Moreover, with fluorescent FITC-EntV88-His6, we revealed that EntV88-His6 enters C. albicans via endocytosis and perturbs the proper localization of the polarisome scaffolding Spa2 protein. Our findings provide important clues on EntV's mechanism of action. Surprisingly, we showed that EntV88-His6 does not affect C. albicans yeast cell growth but potently exerts cytotoxicity against C. albicans under hyphal-inducing conditions in vitro. The combination of EntV88-His6 and GlcNAc displays rapid killing of C. albicans, rendering it a promising antivirulent and antifungal agent.
Collapse
Affiliation(s)
- Jia Li Fong
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Victor Ong Eng Yong
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore
| | - Claresta Yeo
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Christopher Adamson
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Lanxin Li
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| | - Dan Zhang
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore
| | - Yuan Qiao
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University (NTU), 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
17
|
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
|
18
|
Badr EA, Nagy YI, Sayed RM, Kashef MT. Development of a transcription factor decoy-nanocarrier system as a successful inhibitor of Enterococcus faecalis virulence in vitro and in vivo. Microb Pathog 2024; 193:106762. [PMID: 38936638 DOI: 10.1016/j.micpath.2024.106762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/12/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Enterococcus faecalis is a troublesome nosocomial pathogen that acquired resistance to most available antimicrobial agents. Antivirulence agents represent an unconventional treatment approach. Here, transcription factor decoy (TFD)-loaded cationic liposomes (TLL) were developed as an inhibitor of the Fsr quorum-sensing system and its associated virulence traits, in E. faecalis. The consensus sequence of the FsrA binding site was found conserved among 651 E. faecalis annotated genomes. The TFD was synthesized as an 82 bp DNA duplex, containing the conserved binding sequence, and loaded onto cationic liposomes. The optimum loading capacity, mean particle size, and zeta potential of the TLL were characterized. The developed TLL lacked any effect on E. faecalis growth and significantly inhibited the in vitro production of the proteolytic enzymes controlled by the Fsr system; gelatinase and serine protease, in a concentration-dependent manner. This inhibition was accompanied by a significant reduction in the transcription levels of FsrA-regulated genes (fsrB, gelE, and sprE). The developed TLL were safe as evidenced by the nonhemolytic effect on human RBCs and the negligible cytotoxicity on human skin fibroblast cells. Moreover, in the larvae infection model, TLL displayed a significant abolish in the mortality rates of Galleria mellonella larvae infected with E. faecalis. In conclusion, the developed TLL offer a new safe strategy for combating E. faecalis infection through the inhibition of quorum-sensing-mediated virulence; providing a platform for the development of similar agents to combat many other pathogens.
Collapse
Affiliation(s)
- Eslam A Badr
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Yosra Ibrahim Nagy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Rehab Mahmoud Sayed
- Natural Products Research Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mona T Kashef
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
19
|
Sheriff EK, Salvato F, Andersen SE, Chatterjee A, Kleiner M, Duerkop BA. Enterococcal quorum-controlled protease alters phage infection. FEMS MICROBES 2024; 5:xtae022. [PMID: 39156124 PMCID: PMC11328733 DOI: 10.1093/femsmc/xtae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/21/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Increased prevalence of multidrug-resistant bacterial infections has sparked interest in alternative antimicrobials, including bacteriophages (phages). Limited understanding of the phage infection process hampers our ability to utilize phages to their full therapeutic potential. To understand phage infection dynamics, we performed proteomics on Enterococcus faecalis infected with the phage VPE25. We discovered that numerous uncharacterized phage proteins are produced during phage infection of E. faecalis. Additionally, we identified hundreds of changes in bacterial protein abundances during infection. One such protein, enterococcal gelatinase (GelE), an fsr quorum-sensing-regulated protease involved in biofilm formation and virulence, was reduced during VPE25 infection. Plaque assays showed that mutation of either the quorum-sensing regulator fsrA or gelE resulted in plaques with a "halo" morphology and significantly larger diameters, suggesting decreased protection from phage infection. GelE-associated protection during phage infection is dependent on the putative murein hydrolase regulator LrgA and antiholin-like protein LrgB, whose expression have been shown to be regulated by GelE. Our work may be leveraged in the development of phage therapies that can modulate the production of GelE thereby altering biofilm formation and decreasing E. faecalis virulence.
Collapse
Affiliation(s)
- Emma K Sheriff
- Department of Immunology and Microbiology, School of Medicine, University of Colorado – Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, United States
| | - Fernanda Salvato
- Department of Plant and Microbial Biology, North Carolina State University, 112 Derieux Pl., Raleigh, NC 27695, United States
| | - Shelby E Andersen
- Department of Immunology and Microbiology, School of Medicine, University of Colorado – Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, United States
| | - Anushila Chatterjee
- Department of Immunology and Microbiology, School of Medicine, University of Colorado – Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, United States
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, North Carolina State University, 112 Derieux Pl., Raleigh, NC 27695, United States
| | - Breck A Duerkop
- Department of Immunology and Microbiology, School of Medicine, University of Colorado – Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, United States
| |
Collapse
|
20
|
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
|
21
|
Wang X, Jin X, Zhao F, Xu Z, Tan W, Zhang J, Xu Y, Luan X, Fang M, Xie Z, Chang W, Lou H. Structure-Based Optimization of Novel Sterol 24-C-Methyltransferase Inhibitors for the Treatment of Candida albicans Infections. J Med Chem 2024; 67:9318-9341. [PMID: 38764175 DOI: 10.1021/acs.jmedchem.4c00470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Interfering with sterol biosynthesis is an important strategy for developing safe and effective antifungal drugs. We previously identified compound H55 as an allosteric inhibitor of the fungal-specific C-24 sterol methyltransferase Erg6 for treating Candida albicans infections. Herein, 62 derivatives of H55 were designed and synthesized based on target-ligand interactions to identify more active candidates. Among them, d28 displayed the most potent antivirulence ability (MHIC50 = 0.25 μg/mL) by targeting Erg6, exhibiting an 8-fold increase in potency compared with H55. Moreover, d28 significantly outperformed H55 in inhibiting cell adhesion and biofilm formation, and exhibited minimal cytotoxicity and negligible potential to induce drug resistance. Of note, the coadministration of d28 and other sterol biosynthesis inhibitors, such as tridemorph or terbinafine, demonstrated a strong synergistic antifungal action in vitro and in vivo in a murine skin infection model. These results support the potential application of d28 in the treatment of C. albicans infections.
Collapse
Affiliation(s)
- Xue Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xueyang Jin
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zejun Xu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenzhuo Tan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiaozhen Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuliang Xu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China
| | - Xiaoyi Luan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Min Fang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhiyu Xie
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang 461002, China
| | - Wenqiang Chang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
22
|
Mallawaarachchi S, Tonkin-Hill G, Pöntinen A, Calland J, Gladstone R, Arredondo-Alonso S, MacAlasdair N, Thorpe H, Top J, Sheppard S, Balding D, Croucher N, Corander J. Detecting co-selection through excess linkage disequilibrium in bacterial genomes. NAR Genom Bioinform 2024; 6:lqae061. [PMID: 38846349 PMCID: PMC11155488 DOI: 10.1093/nargab/lqae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/15/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024] Open
Abstract
Population genomics has revolutionized our ability to study bacterial evolution by enabling data-driven discovery of the genetic architecture of trait variation. Genome-wide association studies (GWAS) have more recently become accompanied by genome-wide epistasis and co-selection (GWES) analysis, which offers a phenotype-free approach to generating hypotheses about selective processes that simultaneously impact multiple loci across the genome. However, existing GWES methods only consider associations between distant pairs of loci within the genome due to the strong impact of linkage-disequilibrium (LD) over short distances. Based on the general functional organisation of genomes it is nevertheless expected that majority of co-selection and epistasis will act within relatively short genomic proximity, on co-variation occurring within genes and their promoter regions, and within operons. Here, we introduce LDWeaver, which enables an exhaustive GWES across both short- and long-range LD, to disentangle likely neutral co-variation from selection. We demonstrate the ability of LDWeaver to efficiently generate hypotheses about co-selection using large genomic surveys of multiple major human bacterial pathogen species and validate several findings using functional annotation and phenotypic measurements. Our approach will facilitate the study of bacterial evolution in the light of rapidly expanding population genomic data.
Collapse
Affiliation(s)
| | | | - Anna K Pöntinen
- Department of Biostatistics, University of Oslo, Oslo, Norway
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Jessica K Calland
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | | | | | | | - Harry A Thorpe
- Department of Biostatistics, University of Oslo, Oslo, Norway
| | - Janetta Top
- Department of Medical Microbiology, UMC Utrecht, Utrecht, The Netherlands
| | - Samuel K Sheppard
- Ineos Oxford Institute of Antimicrobial Research, Department of Biology, University of Oxford, Oxford, United Kingdom
| | - David Balding
- Melbourne Integrative Genomics, School of BioSciences and School of Mathematics & Statistics, University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas J Croucher
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, United Kingdom
| | - Jukka Corander
- Department of Biostatistics, University of Oslo, Oslo, Norway
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, UK
- Helsinki Institute of Information Technology, Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Sheriff EK, Salvato F, Andersen SE, Chatterjee A, Kleiner M, Duerkop BA. Enterococcal quorum-controlled protease alters phage infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593607. [PMID: 38766208 PMCID: PMC11100838 DOI: 10.1101/2024.05.10.593607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Increased prevalence of multidrug resistant bacterial infections has sparked interest in alternative antimicrobials, including bacteriophages (phages). Limited understanding of the phage infection process hampers our ability to utilize phages to their full therapeutic potential. To understand phage infection dynamics we performed proteomics on Enterococcus faecalis infected with the phage VPE25. We discovered numerous uncharacterized phage proteins are produced during phage infection of Enterococcus faecalis. Additionally, we identified hundreds of changes in bacterial protein abundances during infection. One such protein, enterococcal gelatinase (GelE), an fsr quorum sensing regulated protease involved in biofilm formation and virulence, was reduced during VPE25 infection. Plaque assays showed that mutation of either the fsrA or gelE resulted in plaques with a "halo" morphology and significantly larger diameters, suggesting decreased protection from phage infection. GelE-associated protection during phage infection is dependent on the murein hydrolase regulator LrgA and antiholin-like protein LrgB, whose expression have been shown to be regulated by GelE. Our work may be leveraged in the development of phage therapies that can modulate the production of GelE thereby altering biofilm formation and decreasing E. faecalis virulence.
Collapse
Affiliation(s)
- Emma K. Sheriff
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Fernanda Salvato
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC 27695
| | - Shelby E. Andersen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Anushila Chatterjee
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC 27695
| | - Breck A. Duerkop
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
24
|
Guha S, Cristy SA, Buda De Cesare G, Cruz MR, Lorenz MC, Garsin DA. Optimization of the antifungal properties of the bacterial peptide EntV by variant analysis. mBio 2024; 15:e0057024. [PMID: 38587425 PMCID: PMC11077972 DOI: 10.1128/mbio.00570-24] [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: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 04/09/2024] Open
Abstract
Fungal resistance to commonly used medicines is a growing public health threat, and there is a dire need to develop new classes of antifungals. We previously described a peptide produced by Enterococcus faecalis, EntV, that restricts Candida albicans to a benign form rather than having direct fungicidal activity. Moreover, we showed that one 12-amino acid (aa) alpha helix of this peptide retained full activity, with partial activity down to the 10aa alpha helix. Using these peptides as a starting point, the current investigation sought to identify the critical features necessary for antifungal activity and to screen for new variants with enhanced activity using both biofilm and C. elegans infection assays. First, the short peptides were screened for residues with critical activity by generating alanine substitutions. Based on this information, we used synthetic molecular evolution (SME) to rationally vary the specific residues of the 10aa variant in combination to generate a library that was screened to identify variants with more potent antifungal activity than the parent template. Five gain-of-function peptides were identified. Additionally, chemical modifications to the peptides to increase stability, including substitutions of D-amino acids and hydrocarbon stapling, were investigated. The most promising peptides were additionally tested in mouse models of oropharyngeal and systemic candidiasis where their efficacy in preventing infection was demonstrated. The expectation is that these discoveries will contribute to the development of new therapeutics in the fight against antimicrobial resistant fungi. IMPORTANCE Since the early 1980s, the incidence of disseminated life-threatening fungal infections has been on the rise. Worldwide, Candida and Cryptococcus species are among the most common agents causing these infections. Simultaneously, with this rise of clinical incidence, there has also been an increased prevalence of antifungal resistance, making treatment of these infections very difficult. For example, there are now strains of Candida auris that are resistant to all three classes of currently used antifungal drugs. In this study, we report on a strategy that allows for the development of novel antifungal agents by using synthetic molecular evolution. These discoveries demonstrate that the enhancement of antifungal activity from naturally occurring peptides is possible and can result in clinically relevant agents that have efficacy in multiple in vivo models as well as the potential for broad-spectrum activity.
Collapse
Affiliation(s)
- Shantanu Guha
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shane A. Cristy
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Giuseppe Buda De Cesare
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Melissa R. Cruz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Danielle A. Garsin
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
25
|
Sookkhee S, Khamnoi P, Sastraruji T, Boonkum S, Wikan N, Nimlamool W. Synergistic Inhibition of Synbiotic Cultures among Lactobacilli and Plant Extracts against Vaginal Discharge Causing Candida albicans. Nutrients 2024; 16:1372. [PMID: 38732618 PMCID: PMC11085874 DOI: 10.3390/nu16091372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Vulvovaginal candidiasis (VVC) is the most common cause of vaginal discharge among women. The present study aimed to investigate the synergistic anticandidal effect of lactobacillus cultures supplemented with plant extracts. Among 600 isolates of lactic acid bacteria, 41 isolates exhibited inhibitory activity against Candida albicans ATCC10231. Six out of 41 cell-free supernatants demonstrated the most potent antibacterial and anticandidal activities. They also inhibited the clinical isolates of C. albicans, causing VVC and non-C. albicans. The synergistic effect between Lactobacillus crispatus 84/7 and Limosilactobacillus reuteri 89/4 was demonstrated by the lowest fractional inhibitory concentration index (FICI = 0.5). The synbiotic culture of bacterial combination, cultured with Jerusalem artichoke (H. tuberosus) extract, also exhibited the strongest inhibition against the tested C. albicans. Biofilm formation decreased after 12 h of incubation in the selected cell-free supernatants of this synbiotic culture. The anticandidal activity of crude extracts was lost after treatment with proteinase K and trypsin but not with heating conditions, suggesting that it may be a heat-stable substance. In conclusion, the combination of L. crispatus 84/7 and L. reuteri 89/4 with H. tuberosus may be a promising candidate for inhibiting Candida infection and biofilm formation, with the potential use as ingredients in vaginal biotherapeutic products.
Collapse
Affiliation(s)
- Siriwoot Sookkhee
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Phadungkiat Khamnoi
- Diagnostic Laboratory Unit, Maharaj Nakorn Chiang Mai Hospital, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Thanapat Sastraruji
- Dental Research Center, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sathian Boonkum
- Department of Biotechnology, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Lanna Rice Research Center, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
26
|
Pedro NA, Mira NP. A molecular view on the interference established between vaginal Lactobacilli and pathogenic Candida species: Challenges and opportunities for the development of new therapies. Microbiol Res 2024; 281:127628. [PMID: 38246122 DOI: 10.1016/j.micres.2024.127628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Vaginal infectious diseases caused by viruses and bacteria have been linked to the occurrence of dysbiosis, that is, a reduction in the abundance of the normally dominating vaginal Lactobacillus species. Mucosal infections in the vagina and/or vulva caused by Candida species, usually known as vulvovaginal candidiasis (or VVC), are among the leading causes of diseases in the vaginal tract. The existence of a clear link between the occurrence of dysbiosis and the development of VVC is still unclear, although multiple observations point in that direction. Based on the idea that vaginal health is linked to a microbiota dominated by lactobacilli, several probiotics have been used in management of VVC, either alone or in combination with antifungals, having obtained different degrees of success. In most cases, the undertaken trials resorted to lactobacilli species other than those indigenous to the vaginal tract, although in vitro these vaginal species were shown to reduce growth, viability and virulence of Candida. In this paper we overview the role of lactobacilli and Candida in the vaginal micro- and myco-biomes, while discussing the results obtained in what concerns the establishment of interference mechanisms in vivo and the environmental factors that could determine that. We also overview the molecular mechanisms by which lactobacilli species have been shown to inhibit pathophysiology of Candida, including the description of the genes and pathways determining their ability to thrive in the presence of each other. In a time where concerns are increasing with the emergence of antifungal resistance and the slow pace of discovery of new antifungals, a thorough understanding of the molecular mechanisms underneath the anti-Candida effect prompted by vaginal lactobacilli is of utmost importance to assure a knowledge-based design of what can be a new generation of pharmaceuticals, eventually focusing therapeutic targets other than the usual ones.
Collapse
Affiliation(s)
- Nuno A Pedro
- iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico - Department of Bioengineering, Universidade de Lisboa, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Nuno P Mira
- iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico - Department of Bioengineering, Universidade de Lisboa, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| |
Collapse
|
27
|
Cohen S, Ost KS, Doran KS. Impact of interkingdom microbial interactions in the vaginal tract. PLoS Pathog 2024; 20:e1012018. [PMID: 38457371 PMCID: PMC10923463 DOI: 10.1371/journal.ppat.1012018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Affiliation(s)
- Shirli Cohen
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Kyla S. Ost
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Kelly S. Doran
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| |
Collapse
|
28
|
Ul Haq I, Maryam S, Shyntum DY, Khan TA, Li F. Exploring the frontiers of therapeutic breadth of antifungal peptides: A new avenue in antifungal drugs. J Ind Microbiol Biotechnol 2024; 51:kuae018. [PMID: 38710584 PMCID: PMC11119867 DOI: 10.1093/jimb/kuae018] [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: 02/14/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
The growing prevalence of fungal infections alongside rising resistance to antifungal drugs poses a significant challenge to public health safety. At the close of the 2000s, major pharmaceutical firms began to scale back on antimicrobial research due to repeated setbacks and diminished economic gains, leaving only smaller companies and research labs to pursue new antifungal solutions. Among various natural sources explored for novel antifungal compounds, antifungal peptides (AFPs) emerge as particularly promising. Despite their potential, AFPs receive less focus than their antibacterial counterparts. These peptides have been sourced extensively from nature, including plants, animals, insects, and especially bacteria and fungi. Furthermore, with advancements in recombinant biotechnology and computational biology, AFPs can also be synthesized in lab settings, facilitating peptide production. AFPs are noted for their wide-ranging efficacy, in vitro and in vivo safety, and ability to combat biofilms. They are distinguished by their high specificity, minimal toxicity to cells, and reduced likelihood of resistance development. This review aims to comprehensively cover AFPs, including their sources-both natural and synthetic-their antifungal and biofilm-fighting capabilities in laboratory and real-world settings, their action mechanisms, and the current status of AFP research. ONE-SENTENCE SUMMARY This comprehensive review of AFPs will be helpful for further research in antifungal research.
Collapse
Affiliation(s)
- Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
- Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
- Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Sajida Maryam
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
- Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - Divine Y Shyntum
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Taj A Khan
- Division of Infectious Diseases & Global Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Fan Li
- School of Life Sciences, Peking University, Beijing 100871, People's Republic of China
| |
Collapse
|
29
|
Sobel JD, Vempati YS. Bacterial Vaginosis and Vulvovaginal Candidiasis Pathophysiologic Interrelationship. Microorganisms 2024; 12:108. [PMID: 38257934 PMCID: PMC10820109 DOI: 10.3390/microorganisms12010108] [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: 12/06/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Among the infectious causes of vulvovaginal symptoms, bacterial vaginosis (BV) and vulvovaginal candidiasis (VVC) dominate. Apart from infrequent mixed infections, both are considered independent and caused by unrelated pathogenic mechanisms. Clinical experience, however, is strongly suggestive that in some populations these infections are linked with recurrent BV (RBV) serving as the dominant etiopathogenic trigger for development of recurrent VVC (RVVC) with profound clinical and therapeutic consequences. The biologic basis for this critical interrelationship is discussed and suggests that as a consequence of BV dysbiosis, and not necessarily because of antibiotics prescribed, immune defenses are compromised, neutralizing vaginal yeast tolerance. The consequent BV-induced vaginal proinflammatory environment predisposes to mixed infection or consecutive episodes of post-treatment VVC. Recurrent BV and repeated antimicrobial drug exposure also predispose to acquired fluconazole resistance in C. albicans isolates, contributing to refractory vulvovaginal candidiasis.
Collapse
Affiliation(s)
- Jack D. Sobel
- C.S. Mott Center for Growth and Human Development, 275 E. Hancock St, Detroit, MI 48201, USA
| | - Yogitha Sai Vempati
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| |
Collapse
|
30
|
Srinivasan A, Sajeevan A, Rajaramon S, David H, Solomon AP. Solving polymicrobial puzzles: evolutionary dynamics and future directions. Front Cell Infect Microbiol 2023; 13:1295063. [PMID: 38145044 PMCID: PMC10748482 DOI: 10.3389/fcimb.2023.1295063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/03/2023] [Indexed: 12/26/2023] Open
Abstract
Polymicrobial infections include various microorganisms, often necessitating different treatment methods than a monomicrobial infection. Scientists have been puzzled by the complex interactions within these communities for generations. The presence of specific microorganisms warrants a chronic infection and impacts crucial factors such as virulence and antibiotic susceptibility. Game theory is valuable for scenarios involving multiple decision-makers, but its relevance to polymicrobial infections is limited. Eco-evolutionary dynamics introduce causation for multiple proteomic interactions like metabolic syntropy and niche segregation. The review culminates both these giants to form evolutionary dynamics (ED). There is a significant amount of literature on inter-bacterial interactions that remain unsynchronised. Such raw data can only be moulded by analysing the ED involved. The review culminates the inter-bacterial interactions in multiple clinically relevant polymicrobial infections like chronic wounds, CAUTI, otitis media and dental carries. The data is further moulded with ED to analyse the niche colonisation of two notoriously competitive bacteria: S.aureus and P.aeruginosa. The review attempts to develop a future trajectory for polymicrobial research by following recent innovative strategies incorporating ED to curb polymicrobial infections.
Collapse
Affiliation(s)
| | | | | | | | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
31
|
Ajetunmobi OH, Badali H, Romo JA, Ramage G, Lopez-Ribot JL. Antifungal therapy of Candida biofilms: Past, present and future. Biofilm 2023; 5:100126. [PMID: 37193227 PMCID: PMC10182175 DOI: 10.1016/j.bioflm.2023.100126] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/18/2023] Open
Abstract
Virtually all Candida species linked to clinical candidiasis are capable of forming highly resistant biofilms on different types of surfaces, which poses an additional significant threat and further complicates therapy of these infections. There is a scarcity of antifungal agents, and their effectiveness, particularly against biofilms, is limited. Here we provide a historical perspective on antifungal agents and therapy of Candida biofilms. As we reflect upon the past, consider the present, and look towards the future of antifungal therapy of Candida biofilms, we believe that there are reasons to remain optimistic, and that the major challenges of Candida biofilm therapy can be conquered within a reasonable timeframe.
Collapse
Affiliation(s)
- Olabayo H. Ajetunmobi
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Hamid Badali
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jesus A. Romo
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Gordon Ramage
- Glasgow Biofilm Research Network, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jose L. Lopez-Ribot
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
- Corresponding author. Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
32
|
Delavy M, Sertour N, d'Enfert C, Bougnoux ME. Metagenomics and metabolomics approaches in the study of Candida albicans colonization of host niches: a framework for finding microbiome-based antifungal strategies. Trends Microbiol 2023; 31:1276-1286. [PMID: 37652786 DOI: 10.1016/j.tim.2023.08.002] [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] [Received: 04/11/2023] [Revised: 06/16/2023] [Accepted: 08/02/2023] [Indexed: 09/02/2023]
Abstract
In silico and experimental approaches have allowed an ever-growing understanding of the interactions within the microbiota. For instance, recently acquired data have increased knowledge of the mechanisms that support, in the gut and vaginal microbiota, the resistance to colonization by Candida albicans, an opportunistic fungal pathogen whose overgrowth can initiate severe infections in immunocompromised patients. Here, we review how bacteria from the microbiota interact with C. albicans. We show how recent OMICs-based pipelines, using metagenomics and/or metabolomics, have identified bacterial species and metabolites modulating C. albicans growth. We finally discuss how the combined use of cutting-edge OMICs-based and experimental approaches could provide new means to control C. albicans overgrowth within the microbiota and prevent its consequences.
Collapse
Affiliation(s)
- Margot Delavy
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Natacha Sertour
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Christophe d'Enfert
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Marie-Elisabeth Bougnoux
- Institut Pasteur, Université Paris Cité, INRAE USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France; Assistance Publique des Hôpitaux de Paris (APHP), Hôpital Necker-Enfants-Malades, Unité de Parasitologie-Mycologie, Service de Microbiologie Clinique, Paris, France.
| |
Collapse
|
33
|
Mousavi SM, Archangi B, Zamani I. Antibacterial Properties of Bacteriocin Purified from Serratia marcescens and Computerized Assessment of its Interaction with Antigen 43 in Escherichia coli. ARCHIVES OF RAZI INSTITUTE 2023; 78:1738-1745. [PMID: 38828162 PMCID: PMC11139391 DOI: 10.32592/ari.2023.78.6.2694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/06/2023] [Indexed: 06/05/2024]
Abstract
Bacteriocins are a kind of antimicrobial peptides that kill or inhibit the growth of bacterial strains. The purpose of this study was to investigate the antibacterial effect of Serratia marcescens on several pathogenic bacterial strains. Bacteriocin produced by S. marcescens was purified by chromatography with Sephadex G-75 column, and its antibacterial effect on gram-negative bacteria, including Escherichia coli ATCC 700928, Pseudomonas aeruginosa PTCC 1707, S. marcescens PTCC 1621, Vibrio fischeri PTCC 1693, and Vibrio harveyi PTCC 1755, were evaluated by the disk diffusion method. The structure of bacteriocin was determined by nuclear magnetic resonance spectroscopy. The interaction of bacteriocin with the antigen 43 (Ag43) of E. coli was evaluated by the molecular docking method. Bacteriocin extracted from bacterial isolates had antibacterial activity on E. coli strains but not on other studied strains. Bioinformatics analysis also showed bacteriocin docking with Ag43 with an energy of -159.968 kJ/mol. Natural compounds, such as bacteriocin, can be an alternative to common chemical compounds and antibiotics. To reach a definite conclusion in this regard, there is a need for further research and understanding of their mechanism of action.
Collapse
Affiliation(s)
- S M Mousavi
- Department of Marine Biotechnology, Khorramshahr University of Marine Science and Technology, Khorramshahr, Iran
| | - B Archangi
- Department of Marine Biotechnology, Khorramshahr University of Marine Science and Technology, Khorramshahr, Iran
| | - I Zamani
- Department of Marine Biotechnology, Khorramshahr University of Marine Science and Technology, Khorramshahr, Iran
| |
Collapse
|
34
|
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: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
35
|
Song S, Zhao S, Sun X, Meng L, Wang Z, Tan H, Liu J, Zhang M, Deng Y. Anti-virulence strategy of diaryl chalcogenide compounds against Candida albicans infection. Virulence 2023; 14:2265012. [PMID: 37771181 PMCID: PMC10549196 DOI: 10.1080/21505594.2023.2265012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 09/25/2023] [Indexed: 09/30/2023] Open
Abstract
Candida albicans is an important opportunistic pathogenic fungus that frequently causes serious systemic infection in humans. Due to the vital roles of biofilm formation and the yeast-to-hypha transition in the infection process, we have selected a series of diaryl chalcogenides and tested their efficacy against C. albicans SC5314 pathogenicity by the inhibition of biofilm formation and the yeast-to-hypha transition. The compounds 5-sulfenylindole and 5-selenylindole were found to have excellent abilities to inhibit both biofilm formation and hyphal formation in C. albicans SC5314. Intriguingly, the two leading compounds also markedly attenuated C. albicans SC5314 virulence in human cell lines and mouse infection models at micromolar levels. Furthermore, our results showed that the presence of the compounds at 100 µM resulted in a marked decrease in the expression of genes involved in the cAMP-PKA and MAPK pathways in C. albicans SC5314. Intriguingly, the compounds 5-sulfenylindole and 5-selenylindole not only attenuated the cytotoxicity of Candida species strains but also showed excellent synergistic effects with antifungal agents against the clinical drug-resistant C. albicans strain HCH12. The compound 5-sulfenylindole showed an obvious advantage over fluconazole as it could also restore the composition and richness of the intestinal microbiota in mice infected by C. albicans. Together, these results suggest that diaryl chalcogenides can potentially be designed as novel clinical therapeutic agents against C. albicans infection. The diaryl chalcogenides of 5-sulfenylindole and 5-selenylindole discovered in this study can provide new direction for developing antifungal agents against C. albicans infection.
Collapse
Affiliation(s)
- Shihao Song
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shuo Zhao
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Xiuyun Sun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Lili Meng
- Integrative Microbiology Research Center, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Zijie Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Hunan Children’s Hospital, Changsha, China
| | - Huihui Tan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Jingyun Liu
- Integrative Microbiology Research Center, College of Plant Protection, South China Agricultural University, Guangzhou, China
- Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Min Zhang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, China
| | - Yinyue Deng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
36
|
Kim KS, Lee GH, Bak HR, Park YM, Lee SH, Hong SJ, Lee DW. Complete genome assembly of Enterococcus faecalis strain HL1, isolated from an infant fecal sample. Microbiol Resour Announc 2023; 12:e0055823. [PMID: 37905826 PMCID: PMC10652978 DOI: 10.1128/mra.00558-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/21/2023] [Indexed: 11/02/2023] Open
Abstract
We present the complete genome sequence of Enterococcus faecalis strain HL1, isolated from infant feces. E. faecalis gains significant attention for its therapeutic potential. The genome of E. faecalis HL1 consists of a 2.7 Mb circular chromosome with no plasmids, and it contains a total of 2,546 predicted coding genes.
Collapse
Affiliation(s)
- Kyoung Su Kim
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Gun-Hyeong Lee
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Hyun Ryul Bak
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Yoon Mee Park
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung Hwa Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| |
Collapse
|
37
|
Huang X, Hu M, Sun T, Li J, Zhou Y, Yan Y, Xuan B, Wang J, Xiong H, Ji L, Zhu X, Tong T, Ning L, Ma Y, Zhao Y, Ding J, Guo Z, Zhang Y, Fang JY, Hong J, Chen H. Multi-kingdom gut microbiota analyses define bacterial-fungal interplay and microbial markers of pan-cancer immunotherapy across cohorts. Cell Host Microbe 2023; 31:1930-1943.e4. [PMID: 37944495 DOI: 10.1016/j.chom.2023.10.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/11/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
The effect of gut bacteria on the response to immune checkpoint inhibitors (ICIs) has been studied, but the relationship between fungi and ICI responses is not fully understood. Herein, 862 fecal metagenomes from 9 different cohorts were integrated for the identification of differentially abundant fungi and subsequent construction of random forest (RF) models to predict ICI responses. Fungal markers demonstrate excellent performance, with an average area under the curve (AUC) of 0.87. Their performance improves even further, reaching an average AUC of 0.89 when combined with bacterial markers. Higher enrichment of exhausted T cells is detected in responders, as predicted by fungal markers. Multi-kingdom network and functional analysis reveal that the fungus Schizosaccharomyces octosporus may ferment starch into short-chain fatty acids in responders. This study provides a fungal profile of the ICI response and the identification of multi-kingdom microbial markers with good performance that may improve the overall applicability of ICI therapy.
Collapse
Affiliation(s)
- Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Tiantian Sun
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jiantao Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yilu Zhou
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yuqing Yan
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jilin Wang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Hua Xiong
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Linhua Ji
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China.
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China.
| |
Collapse
|
38
|
Greenwich JL, Fleming D, Banin E, Häussler S, Kjellerup BV, Sauer K, Visick KL, Fuqua C. The biofilm community resurfaces: new findings and post-pandemic progress. J Bacteriol 2023; 205:e0016623. [PMID: 37756166 PMCID: PMC10601713 DOI: 10.1128/jb.00166-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
The ninth American Society for Microbiology Conference on Biofilms was convened in-person on 13-17 November 2022 in Charlotte, NC. As the first of these conferences since prior to the start of the COVID-19 pandemic, the energy among the participants of the conference was clear, and the meeting was a tremendous success. The mixture of >330 oral and poster presentations resoundingly embodied the vitality of biofilm research across a wide range of topics and multiple scientific disciplines. Special activities, including a pre-conference symposium for early career researchers, further enhanced the attendee experience. As a general theme, the conference was deliberately structured to provide high levels of participation and engagement among early career scientists.
Collapse
Affiliation(s)
| | - Derek Fleming
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ehud Banin
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Birthe V. Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Karin Sauer
- Department of Biological Sciences, University of Binghamton, Binghamton, New York, USA
| | - Karen L. Visick
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Clay Fuqua
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
39
|
Robertson EB, Willett JLE. Streptococcus mutans inhibits the growth of Enterococcus via the non-ribosomal cyclic peptide mutanobactin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557362. [PMID: 37745448 PMCID: PMC10515869 DOI: 10.1101/2023.09.12.557362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Enterococcus faecalis is a Gram-positive commensal bacterium in the gastrointestinal tract and an opportunistic pathogen. Enterococci are a leading cause of nosocomial infections, treatment of which is complicated by intrinsic and acquired antibiotic resistance mechanisms. Additionally, E. faecalis has been associated with various oral diseases, and it is frequently implicated in the failure of endodontic treatment. For establishment and persistence in a microbial community, E. faecalis must successfully compete against other bacteria. Streptococcal species play an important role in the establishment of the oral microbiome and co-exist with Enterococcus in the small intestine, yet the nature of interactions between E. faecalis and oral streptococci remains unclear. Here, we describe a mechanism by which Streptococcus mutans inhibits the growth of E. faecalis and other Gram-positive pathogens through the production of mutanobactin, a cyclic lipopeptide. Mutanobactin is produced by a polyketide synthase-nonribosomal peptide synthetase hybrid system encoded by the mub locus. Mutanobactin-producing S. mutans inhibits planktonic and biofilm growth of E. faecalis and is also active against other Enterococcus species and Staphylococcus aureus. Mutanobactin damages the cell envelope of E. faecalis, similar to other lipopeptide antibiotics like daptomycin. E. faecalis resistance to mutanobactin is mediated by the virulence factor gelatinase, a secreted metalloprotease. Our results highlight the anti-biofilm potential of the microbial natural product mutanobactin, provide insight into how E. faecalis interacts with other organisms in the human microbiome, and demonstrate the importance of studying E. faecalis dynamics within polymicrobial communities.
Collapse
Affiliation(s)
- Ethan B. Robertson
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455 USA
| | - Julia L. E. Willett
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455 USA
| |
Collapse
|
40
|
Silva ML, Carneiro MN, Cavalcante RMB, Guerrero JAP, Fontenelle ROS, Lorenzón EN, Cilli EM, Carneiro VA. K-aurein: A notable aurein 1.2-derived peptide that modulates Candida albicans filamentation and reduces biofilm biomass. Amino Acids 2023; 55:1003-1012. [PMID: 37442853 DOI: 10.1007/s00726-023-03288-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/25/2023] [Indexed: 07/15/2023]
Abstract
Candida albicans is considered one of the most important opportunistic fungi due to the large arsenal of virulence factors that help throughout the progress of the infection. In this sense, antimicrobial peptides (AMPs) appear as an alternative, with great antifungal action. Among these, aurein 1.2 has been widely explored, becoming the basis for the discovery of new AMPs, such as K-aurein (K-au). Thus, this study evaluated the anti-C. albicans potential of K-au against virulence factors, planktonic growth, and biofilm formation of clinical isolates. Firstly, K-au antifungal activity was determined by the microdilution method and time-kill curve. The inhibition of hydrolytic enzyme secretion (proteinase, phospholipase, and hemolysin) and germ tube formation was tested. Then, the antibiofilm potential of K-au was verified through biomass quantification and scanning electron microscopy (SEM). All tests were compared with the classical antifungal drug, amphotericin B (AmB). The outcomes showed fungicidal action of K-au at 62.50 µg mL-1 for all isolates, with a time of action around 150-180 min, determined by the time-kill curve. K-au-treated cells decreased by around 40% of the germinative tube compared to the control. Additionally, K-au inhibited the biofilm formation by more than 90% compared to AmB and the control group. SEM images show apparent cellular disaggregation without the formation of filamentous structures. Therefore, the findings suggest a promising anti-C. albicans effect of K-au due to its fungicidal activity against planktonic cells, or its ability to inhibit important virulence factors like germ tube and biofilm formation. Thus, this peptide could be explored as a useful compound against C. albicans-related infection.
Collapse
Affiliation(s)
- Maria Laína Silva
- Laboratory of Biofilms and Antimicrobial Agents (LaBAM), Faculty of Medicine, Federal University of Ceara-UFC, Sobral, 62048-280, Brazil
| | - Maria Nágila Carneiro
- Laboratory of Biofilms and Antimicrobial Agents (LaBAM), Faculty of Medicine, Federal University of Ceara-UFC, Sobral, 62048-280, Brazil
| | - Rafaela Mesquita Bastos Cavalcante
- Laboratory of Biofilms and Antimicrobial Agents (LaBAM), Faculty of Medicine, Federal University of Ceara-UFC, Sobral, 62048-280, Brazil
| | - Jesús Alberto Pérez Guerrero
- Laboratory of Biofilms and Antimicrobial Agents (LaBAM), Faculty of Medicine, Federal University of Ceara-UFC, Sobral, 62048-280, Brazil
| | | | | | - Eduardo Maffud Cilli
- Department of Biochemistry and Organic Chemistry, Estadual University of São Paulo-UNESP, Araraquara, 14800-900, Brazil
| | - Victor Alves Carneiro
- Laboratory of Biofilms and Antimicrobial Agents (LaBAM), Faculty of Medicine, Federal University of Ceara-UFC, Sobral, 62048-280, Brazil.
- Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), University Center INTA-UNINTA, Sobral, 62050-100, Brazil.
| |
Collapse
|
41
|
Rebai Y, Wagner L, Gnaien M, Hammer ML, Kapitan M, Niemiec MJ, Mami W, Mosbah A, Messadi E, Mardassi H, Vylkova S, Jacobsen ID, Znaidi S. Escherichia coli Nissle 1917 Antagonizes Candida albicans Growth and Protects Intestinal Cells from C. albicans-Mediated Damage. Microorganisms 2023; 11:1929. [PMID: 37630490 PMCID: PMC10457924 DOI: 10.3390/microorganisms11081929] [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: 04/22/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 08/27/2023] Open
Abstract
Candida albicans is a pathobiont of the gastrointestinal tract. It can contribute to the diversity of the gut microbiome without causing harmful effects. When the immune system is compromised, C. albicans can damage intestinal cells and cause invasive disease. We hypothesize that a therapeutic approach against C. albicans infections can rely on the antimicrobial properties of probiotic bacteria. We investigated the impact of the probiotic strain Escherichia coli Nissle 1917 (EcN) on C. albicans growth and its ability to cause damage to intestinal cells. In co-culture kinetic assays, C. albicans abundance gradually decreased over time compared with C. albicans abundance in the absence of EcN. Quantification of C. albicans survival suggests that EcN exerts a fungicidal activity. Cell-free supernatants (CFS) collected from C. albicans-EcN co-culture mildly altered C. albicans growth, suggesting the involvement of an EcN-released compound. Using a model of co-culture in the presence of human intestinal epithelial cells, we further show that EcN prevents C. albicans from damaging enterocytes both distantly and through direct contact. Consistently, both C. albicans's filamentous growth and microcolony formation were altered by EcN. Taken together, our study proposes that probiotic-strain EcN can be exploited for future therapeutic approaches against C. albicans infections.
Collapse
Affiliation(s)
- Yasmine Rebai
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
| | - Lysett Wagner
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mayssa Gnaien
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
| | - Merle L. Hammer
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mario Kapitan
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
- Center for Sepsis Control and Care, 07747 Jena, Germany
| | - Maria Joanna Niemiec
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Wael Mami
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Amor Mosbah
- Laboratory of Biotechnology and Bio-Geo Resources Valorization (LR11ES31), Higher Institute of Biotechnology of Sidi Thabet (ISBST), University of Manouba, Tunis 2010, Tunisia
| | - Erij Messadi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Helmi Mardassi
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
| | - Slavena Vylkova
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Ilse D. Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
- Center for Sepsis Control and Care, 07747 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Sadri Znaidi
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
- Institut Pasteur, Institut National de la Recherche Agronomique (INRA), Département Mycologie, Unité Biologie et Pathogénicité Fongiques, 75015 Paris, France
| |
Collapse
|
42
|
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
|
43
|
Conforte JJ, Sousa CA, da Silva ACR, Ribeiro AV, Duque C, Assunção WG. Effect of Enterococcus faecalis Biofilm on Corrosion Kinetics in Titanium Grade 4 Alloys with Different Surface Treatments. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4532. [PMID: 37444846 DOI: 10.3390/ma16134532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023]
Abstract
E. faecalis has been associated with bacteremia, sepsis, and bacterial endocarditis and peri-implantitis. This microorganism can remain in the alveolus even after extraction of the root remnant. This study aimed to evaluate the corrosion on different surfaces of commercially pure titanium (Ti) grade 4 (Ticp-G4) as a function of the bacterial biofilm effect of Enterococcus faecalis. A total of 57 discs were randomly divided according to their surface finish (n = 19). For microbiological analysis (n = 9), the discs were placed in 12-well plates containing E. faecalis culture and incubated at 37 °C for 7 days. The results show that for the intergroup analysis, considering the "electrolyte" factor, there was a difference between the groups. There was greater biofilm formation for the D.A.Zir group, with greater electrochemical exchange for Biofilm, and the presence of biofilm favored greater electrochemical exchange with the medium.
Collapse
Affiliation(s)
- Jadison Junio Conforte
- Department of Dental Materials and Prosthodontic, Araçatuba School of Dentistry, São Paulo State University (UNESP), Sao Paulo 16015-050, Brazil
| | - Cecília Alves Sousa
- Department of Dental Materials and Prosthodontic, Araçatuba School of Dentistry, São Paulo State University (UNESP), Sao Paulo 16015-050, Brazil
| | - Ana Claudia Rodrigues da Silva
- Department of Preventive and Restorative Dentistry, Araçatuba School of Dentistry, São Paulo State University (UNESP), Sao Paulo 16015-050, Brazil
| | | | - Cristiane Duque
- Department of Preventive and Restorative Dentistry, Araçatuba School of Dentistry, São Paulo State University (UNESP), Sao Paulo 16015-050, Brazil
| | - Wirley Gonçalves Assunção
- Department of Dental Materials and Prosthodontic, Araçatuba School of Dentistry, São Paulo State University (UNESP), Sao Paulo 16015-050, Brazil
| |
Collapse
|
44
|
Cong L, Chen C, Mao S, Han Z, Zhu Z, Li Y. Intestinal bacteria-a powerful weapon for fungal infections treatment. Front Cell Infect Microbiol 2023; 13:1187831. [PMID: 37333850 PMCID: PMC10272564 DOI: 10.3389/fcimb.2023.1187831] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
The morbidity and mortality of invasive fungal infections are rising gradually. In recent years, fungi have quietly evolved stronger defense capabilities and increased resistance to antibiotics, posing huge challenges to maintaining physical health. Therefore, developing new drugs and strategies to combat these invasive fungi is crucial. There are a large number of microorganisms in the intestinal tract of mammals, collectively referred to as intestinal microbiota. At the same time, these native microorganisms co-evolve with their hosts in symbiotic relationship. Recent researches have shown that some probiotics and intestinal symbiotic bacteria can inhibit the invasion and colonization of fungi. In this paper, we review the mechanism of some intestinal bacteria affecting the growth and invasion of fungi by targeting the virulence factors, quorum sensing system, secreting active metabolites or regulating the host anti-fungal immune response, so as to provide new strategies for resisting invasive fungal infection.
Collapse
Affiliation(s)
- Liu Cong
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chaoqun Chen
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shanshan Mao
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zibing Han
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zuobin Zhu
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
45
|
Chow EWL, Mei Pang L, Wang Y. Impact of the host microbiota on fungal infections: new possibilities for intervention? Adv Drug Deliv Rev 2023; 198:114896. [PMID: 37211280 DOI: 10.1016/j.addr.2023.114896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Many human fungal pathogens are opportunistic. They are primarily benign residents of the human body and only become infectious when the host's immunity and microbiome are compromised. Bacteria dominate the human microbiome, playing an essential role in keeping fungi harmless and acting as the first line of defense against fungal infection. The Human Microbiome Project, launched by NIH in 2007, has stimulated extensive investigation and significantly advanced our understanding of the molecular mechanisms governing the interaction between bacteria and fungi, providing valuable insights for developing future antifungal strategies by exploiting the interaction. This review summarizes recent progress in this field and discusses new possibilities and challenges. We must seize the opportunities presented by researching bacterial-fungal interplay in the human microbiome to address the global spread of drug-resistant fungal pathogens and the drying pipelines of effective antifungal drugs.
Collapse
Affiliation(s)
- Eve W L Chow
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648
| | - Li Mei Pang
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648
| | - Yue Wang
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore.
| |
Collapse
|
46
|
Jin X, Hou X, Wang X, Zhang M, Chen J, Song M, Zhang J, Zheng H, Chang W, Lou H. Characterization of an allosteric inhibitor of fungal-specific C-24 sterol methyltransferase to treat Candida albicans infections. Cell Chem Biol 2023; 30:553-568.e7. [PMID: 37160123 DOI: 10.1016/j.chembiol.2023.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/02/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023]
Abstract
Filamentation is an important virulence factor of the pathogenic fungus Candida albicans. The abolition of Candida albicans hyphal formation by disrupting sterol synthesis is an important concept for the development of antifungal drugs with high safety. Here, we conduct a high-throughput screen using a C. albicans strain expressing green fluorescent protein-labeled Dpp3 to identify anti-hypha agents by interfering with ergosterol synthesis. The antipyrine derivative H55 is characterized to have minimal cytotoxicity and potent inhibition of C. albicans hyphal formation in multiple cultural conditions. H55 monotherapy exhibits therapeutic efficacy in mouse models of azole-resistant candidiasis. H55 treatment increases the accumulation of zymosterol, the substrate of C-24 sterol methyltransferase (Erg6). The results of enzyme assays, photoaffinity labeling, molecular simulation, mutagenesis, and cellular thermal shift assays support H55 as an allosteric inhibitor of Erg6. Collectively, H55, an inhibitor of the fungal-specific enzyme Erg6, holds potential to treat C. albicans infections.
Collapse
Affiliation(s)
- Xueyang Jin
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xuben Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xue Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ming Zhang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jinyao Chen
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Minghui Song
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Jiaozhen Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Hongbo Zheng
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Wenqiang Chang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
47
|
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
|
48
|
Huang J, Lou Y, Liu J, Bulet P, Cai C, Ma K, Jiao R, Hoffmann JA, Liégeois S, Li Z, Ferrandon D. A Toll pathway effector protects Drosophila specifically from distinct toxins secreted by a fungus or a bacterium. Proc Natl Acad Sci U S A 2023; 120:e2205140120. [PMID: 36917667 PMCID: PMC10041126 DOI: 10.1073/pnas.2205140120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 01/09/2023] [Indexed: 03/16/2023] Open
Abstract
The Drosophila systemic immune response against many Gram-positive bacteria and fungi is mediated by the Toll pathway. How Toll-regulated effectors actually fulfill this role remains poorly understood as the known Toll-regulated antimicrobial peptide (AMP) genes are active only against filamentous fungi and not against Gram-positive bacteria or yeasts. Besides AMPs, two families of peptides secreted in response to infectious stimuli that activate the Toll pathway have been identified, namely Bomanins and peptides derived from a polyprotein precursor known as Baramicin A (BaraA). Unexpectedly, the deletion of a cluster of 10 Bomanins phenocopies the Toll mutant phenotype of susceptibility to infections. Here, we demonstrate that BaraA is required specifically in the host defense against Enterococcus faecalis and against the entomopathogenic fungus Metarhizium robertsii, albeit the fungal burden is not altered in BaraA mutants. BaraA protects the fly from the action of distinct toxins secreted by these Gram-positive and fungal pathogens, respectively, Enterocin V and Destruxin A. The injection of Destruxin A leads to the rapid paralysis of flies, whether wild type (WT) or mutant. However, a larger fraction of wild-type than BaraA flies recovers from paralysis within 5 to 10 h. BaraAs' function in protecting the host from the deleterious action of Destruxin is required in glial cells, highlighting a resilience role for the Toll pathway in the nervous system against microbial virulence factors. Thus, in complement to the current paradigm, innate immunity can cope effectively with the effects of toxins secreted by pathogens through the secretion of dedicated peptides, independently of xenobiotics detoxification pathways.
Collapse
Affiliation(s)
- Jianqiong Huang
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
| | - Yanyan Lou
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
| | - Jiyong Liu
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
| | - Philippe Bulet
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U1209, CNRS, UMR 5309, 38000Grenoble, France
- Platform BioPark Archamps, 74160Archamps, France
| | - Chuping Cai
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
- Université de Strasbourg, Faculté des Sciences de la Vie, 67000Strasbourg, France
- Modèles Insectes d'Immunité Innée, Unité Propre de Recherche 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire du CNRS, 67084Strasbourg, France
| | - Kaiyu Ma
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
| | - Renjie Jiao
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
| | - Jules A. Hoffmann
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
- Université de Strasbourg, Faculté des Sciences de la Vie, 67000Strasbourg, France
- Modèles Insectes d'Immunité Innée, Unité Propre de Recherche 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire du CNRS, 67084Strasbourg, France
- Université de Strasbourg Institute for Advanced Study, 67000Strasbourg, France
| | - Samuel Liégeois
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
- Université de Strasbourg, Faculté des Sciences de la Vie, 67000Strasbourg, France
- Modèles Insectes d'Immunité Innée, Unité Propre de Recherche 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire du CNRS, 67084Strasbourg, France
| | - Zi Li
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
| | - Dominique Ferrandon
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou511436, China
- Université de Strasbourg, Faculté des Sciences de la Vie, 67000Strasbourg, France
- Modèles Insectes d'Immunité Innée, Unité Propre de Recherche 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire du CNRS, 67084Strasbourg, France
| |
Collapse
|
49
|
Juxtaposing Caenorhabditis elegans-Pathogenic Mould Model with Other Models; How Reliable Is This Nematode Model? A Mini Review. Curr Microbiol 2023; 80:105. [PMID: 36790616 DOI: 10.1007/s00284-023-03209-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
The application of Caenorhabditis elegans as a pathogenic model has spanned decades. Its use for pathogenic mould modeling has been attracting some attention lately, though not without some reservations. Several studies have shown C. elegans to be a reliable model for evaluating moulds' virulence factors and patterns as well as for screening the pathogenicity of mutant strains alongside their parental/wild type and revertant/complementary strains. There is a very high degree of reported similarities between the virulence patterns demonstrated in C. elegans and those of other invertebrate and vertebrate models. We have here presented several works in which this nematode model was adopted for virulence evaluation, and other comparative research in which virulence in C. elegans model were juxtaposed with other models. We have further presented possible reasons why there might have been variations of virulence in a few cases, thereby validating C. elegans to be an effective and reliable tool in the study of pathogenic moulds.
Collapse
|
50
|
Secretions from Serratia marcescens Inhibit the Growth and Biofilm Formation of Candida spp. and Cryptococcus neoformans. J Microbiol 2023; 61:221-232. [PMID: 36809632 DOI: 10.1007/s12275-022-00007-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 02/23/2023]
Abstract
Candida spp. and Cryptococcus are conditional pathogenic fungi that commonly infect immunocompromised patients. Over the past few decades, the increase in antifungal resistance has prompted the development of new antifungal agents. In this study, we explored the potential antifungal effects of secretions from Serratia marcescens on Candida spp. and Cryptococcus neoformans. We confirmed that the supernatant of S. marcescens inhibited fungal growth, suppressed hyphal and biofilm formation, and downregulated the expression of hyphae-specific genes and virulence-related genes in Candida spp. and C. neoformans. Furthermore, the S. marcescens supernatant retained biological stability after heat, pH, and protease K treatment. The chemical profile of the S. marcescens supernatant was characterized by ultra-high-performance liquid chromatography-linear ion trap/orbitrap high resolution mass spectrometry analysis and a total of 61 compounds with an mzCloud best match of greater than 70 were identified. In vivo, treatment with the S. marcescens supernatant reduced the mortality of fungi-infected Galleria mellonella. Taken together, our results revealed that the stable antifungal substances in the supernatant of S. marcescens have promising potential applications in the development of new antifungal agents.
Collapse
|