1
|
Chugh S, Létisse F, Neyrolles O. The exometabolome as a hidden driver of bacterial virulence and pathogenesis. Trends Microbiol 2025; 33:546-557. [PMID: 39701858 DOI: 10.1016/j.tim.2024.11.009] [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: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
The traditional view of metabolism as merely supplying energy and biosynthetic precursors is undergoing a paradigm shift. Metabolic dynamics not only regulates gene expression but also orchestrates cellular processes with remarkable precision. Most bacterial pathogens exhibit exceptional metabolic plasticity, enabling them to adapt to diverse environments, including hostile conditions within a host. While the role of intracellular bacterial metabolism in pathogen-host interactions has been extensively studied, the contributions of the extracellularly released or secreted bacterial metabolites (referred to here as the bacterial 'exometabolome') to metabolic adaptations and disease pathogenesis remain largely unexplored. In this review, we highlight the significant and intriguing roles of bacterial exometabolomes in drug tolerance, immune suppression, and disease pathogenesis, opening a new frontier in our understanding of bacterial-host interactions.
Collapse
Affiliation(s)
- Saurabh Chugh
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Fabien Létisse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
2
|
Kulshrestha A, Gupta P. Multi-computational screening identifies homovanillic acid as a potential SAP5 inhibitor against Candida albicans biofilms. Comput Biol Chem 2025; 118:108453. [PMID: 40222055 DOI: 10.1016/j.compbiolchem.2025.108453] [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: 12/31/2024] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
This work aims to find inhibitors of SAP5, a virulence factor in Candida albicans polymicrobial biofilms. The methodology included docking simulations, MMGBSA calculations, and molecular dynamics simulations. Of the 107 phenolic acids retrieved from PubChem, 20 passed ADMET screening. The research finds homovanillic acid to be a possible SAP5 inhibitor, with a binding energy of -19.92 kcal/mol as shown by molecular docking and MMGBSA analysis. The compound showed favorable ADMET properties, indicating low toxicity and high drug-likeness. Molecular dynamics simulations over 100 nanoseconds confirmed stable protein-ligand interactions. These findings suggest homovanillic acid's potential in treating AMR-associated biofilms and establish a foundation for experimental validation. The study demonstrates how computational methods can accelerate the discovery of novel antifungal medicines targeting polymicrobial infections.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh 492010, India.
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh 492010, India.
| |
Collapse
|
3
|
Cha J, Kim TG, Ryu JH. Conversation between skin microbiota and the host: from early life to adulthood. Exp Mol Med 2025; 57:703-713. [PMID: 40164684 PMCID: PMC12045987 DOI: 10.1038/s12276-025-01427-y] [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: 10/28/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 04/02/2025] Open
Abstract
Host life is inextricably linked to commensal microbiota, which play a crucial role in maintaining homeostasis and immune activation. A diverse array of commensal microbiota on the skin interacts with the host, influencing the skin physiology in various ways. Early-life exposure to commensal microbiota has long-lasting effects, and disruption of the epidermal barrier or transient exposure to these microorganisms can lead to skin dysbiosis and inflammation. Several commensal skin microbiota have the potential to function as either commensals or pathogens, both influencing and being influenced by the pathogenesis of skin inflammatory diseases. Here we explore the impact of various commensal skin microbiota on the host and elucidate the interactions between skin microbiota and host systems. A deeper understanding of these interactions may open new avenues for developing effective strategies to address skin diseases.
Collapse
Affiliation(s)
- Jimin Cha
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Hwan Ryu
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Karzani S, Sharifzadeh A, Nayeri-Fasaei B, Khosravi AR, Hassan J, Sharifi A, Shahrestani AP. Impact of oleuropein on Candida albicans and Staphylococcus aureus adhesion and its mediated toxicity in Zebrafish ( Danio rerio) embryos. IRANIAN JOURNAL OF MICROBIOLOGY 2025; 17:312-320. [PMID: 40337685 PMCID: PMC12053420 DOI: 10.18502/ijm.v17i2.18390] [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] [Indexed: 05/09/2025]
Abstract
Background and Objectives The rising prevalence of antibiotic resistance and biofilm-associated infections poses significant challenges in clinical settings. This study investigates the antimicrobial and anti-adhesive properties of oleuropein, a compound derived from olive leaves, against Candida albicans and Staphylococcus aureus. Materials and Methods This study was conducted on Candida albicans (fluconazole-resistant/susceptible) and Staphylococcus aureus (methicillin-resistant/susceptible). The antifungal, antibacterial, anti-adhesion, and cell surface hydrophobicity (CSH) effects of oleuropein were evaluated. The impact of oleuropein on germ tube formation (GTF) in C. albicans was assessed. Finally, the toxicity of oleuropein was evaluated in zebrafish embryos. Results Oleuropein exhibited MIC values of 10 mg/ml for C. albicans and 5 mg/ml for S. aureus. It significantly (P< 0.05) reduced the adhesion of both microorganisms in a dose-dependent manner, with inhibition percentages of 78.43% and 75.91% for C. albicans and S. aureus, respectively. Additionally, oleuropein reduced the CSH of C. albicans, indicating its potential to interfere with adhesion mechanisms. In addition, oleuropein exhibited inhibition of GTF in C. albicans. Conclusion Oleuropein demonstrates significant antimicrobial and anti-adhesive properties against C. albicans and S. aureus, indicating its potential as a therapeutic agent for preventing biofilm-related infections. However, careful dosage management is crucial due to its observed toxicity at higher concentrations.
Collapse
Affiliation(s)
- Samira Karzani
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Aghil Sharifzadeh
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Bahar Nayeri-Fasaei
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Reza Khosravi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Jalal Hassan
- Department of Comparative Bioscience, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Aram Sharifi
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran
| | - Ali Pourshaban Shahrestani
- Department of Comparative Bioscience, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
5
|
Bordea MA, Nutz BTG, Chiorean AD, Samasca G, Lupan I, Simon LM, Pepelea L. Microbial Interactions in Nature: The Impact of Gram-Negative Bacilli on the Hyphal Growth of Candida albicans. Pathogens 2025; 14:327. [PMID: 40333113 PMCID: PMC12030758 DOI: 10.3390/pathogens14040327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 05/09/2025] Open
Abstract
The escalating global prevalence of fungal and bacterial co-infections underscores the significant and multifaceted impact of ubiquitous microorganisms on both environmental equilibria and human well-being. The human microbiome, a complex ecosystem of bacterial communities, harbors opportunistic pathogens capable of inducing superinfections or concurrent infections with Candida spp. The intricate interplay, exemplified by the interaction between Candida albicans and diverse bacteria, necessitates rigorous investigation to elucidate mechanisms by which this polymicrobial behavior potentiates fungal virulence, particularly in immunocompromised individuals. Our study aims to comprehensively examine the ramifications of these interactions, with a specific focus on their influence on fungal virulence and the consequent exacerbation of disease severity. Achieving a comprehensive understanding of these complex relationships is paramount for informing effective clinical management strategies for infectious diseases, and the accurate identification of fungal-bacterial co-infections holds substantial implications for optimizing clinical treatment paradigms, especially in vulnerable immunocompromised hosts.
Collapse
Affiliation(s)
- Madalina Adriana Bordea
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400151 Cluj-Napoca, Romania; (M.A.B.); (B.T.G.N.); (L.P.)
| | - Benjamin Thomas Georg Nutz
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400151 Cluj-Napoca, Romania; (M.A.B.); (B.T.G.N.); (L.P.)
| | - Alin-Dan Chiorean
- Departmernt of Molecular Biology, Iuliu Hatieganu University of Medicine and Pharmacy, 400151 Cluj-Napoca, Romania;
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400151 Cluj-Napoca, Romania
| | - Iulia Lupan
- Departmernt of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania;
| | - Laura Mihaela Simon
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400151 Cluj-Napoca, Romania; (M.A.B.); (B.T.G.N.); (L.P.)
| | - Lia Pepelea
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400151 Cluj-Napoca, Romania; (M.A.B.); (B.T.G.N.); (L.P.)
| |
Collapse
|
6
|
Oduselu GO, Ajani OO, Ogunnupebi TA, Elebiju OF, Bodun DS, Opebiyi OT, Adebiyi E. Synthesis, in silico and in vitro antimicrobial efficacy of some amidoxime-based benzimidazole and benzimidamide derivatives. RSC Med Chem 2025:d5md00114e. [PMID: 40162201 PMCID: PMC11950986 DOI: 10.1039/d5md00114e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Amidoximes are employed as building blocks to synthesise heterocyclic motifs with biological significance. They are very reactive and are used as prodrugs of amidine. This present study unveils the synthesis of amidoxime-based benzimidazole and benzimidamide motifs and evaluates their in silico and in vitro antimicrobial potential as future drug candidates. The compounds (2a, 2b, 4a-c) were synthesized using multi-step synthetic pathways. The synthesised compounds were characterised using physico-chemical examination, 1H- and 13C-NMR, DEPT-135, and FT-IR spectroscopic analyses. The in silico antimicrobial potentials of the synthesized compounds were carried out against glucosamine-6-phosphate synthase of E. coli (PDB ID: 2VF5), and N-myristoyltransferase (NMT) of C. albicans (PDB ID: 1IYL), while the in vitro antimicrobial screening was investigated against selected bacteria and fungi. The in silico studies were carried out using predicted ADMET screening, molecular docking, MM-GBSA, induced-fit docking (IFD), and molecular dynamics (MD) simulation studies. Furthermore, the in vitro experimental validations were performed using the agar diffusion method and the standard antibacterial and antifungal drugs used were gentamicin and ketoconazole respectively. The predicted toxicity test of the compounds showed no significant risk, except for 4c, which showed high tumorigenic risk. Compounds 2b and 2a gave better binding energies; -8.0 kcal mol-1 for 2VF5 and -11.7 kcal mol-1 for 1IYL, respectively. The antimicrobial zone of inhibition and minimum inhibitory concentration values were 40 mm and 3.90 mg mL-1 against S. mutans, then 42 mm and 1.90 mg mL-1 against C. albicans. Potential antimicrobial drug candidates have been identified in this report and should be explored for future preclinical research.
Collapse
Affiliation(s)
- Gbolahan O Oduselu
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana P. O Box LG 54, Legon Accra Ghana
| | - Olayinka O Ajani
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- Department of Chemistry, Covenant University Km 10 Idiroko Road P.M.B. 1023 Ota Ogun State Nigeria
| | - Temitope A Ogunnupebi
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- Department of Chemistry, Covenant University Km 10 Idiroko Road P.M.B. 1023 Ota Ogun State Nigeria
| | - Oluwadunni F Elebiju
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- Department of Chemistry, Covenant University Km 10 Idiroko Road P.M.B. 1023 Ota Ogun State Nigeria
| | - Damilola S Bodun
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
| | | | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- African Center of Excellence in Bioinformatics and Data Intensive Science, Makerere University Kampala Uganda
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) Heidelberg Germany
| |
Collapse
|
7
|
Odu-Onikosi SG, Momoh TA, Abarra ST, Wood NE, Amulejoye FD, Emery M, Harper GM, Eynon B, Kuri V, Kühlwein H, Merrifield DL. Impact of Autolysed Brewer's Yeast and Soluble Dried Yeast Extract on Growth Performance and Mucosal Health of Atlantic Salmon ( Salmo salar) Parr. Animals (Basel) 2025; 15:323. [PMID: 39943093 PMCID: PMC11816051 DOI: 10.3390/ani15030323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Yeast-based feed additives have emerged as promising functional feed additives (FFAs) to promote sustainable aquaculture development through enhanced gut health and immune modulation in fish. The present study evaluated the impact of autolysed brewer's yeast (ABY) and soluble dried yeast extract (SDYE) in improving the intestinal and skin histology, immune response, and intestinal microbiome of Atlantic salmon parr (Salmo salar) over a 9-week feeding trial. Three experimental diets were produced: a control diet, a diet supplemented with ABY at 2.5 g/kg, and a diet supplemented with SDYE at 2.5 g/kg. These diets were administered to triplicate tanks of Atlantic salmon. The yeast-supplemented diets, especially ABY, improved intestinal mucosal fold length, lamina propria width, microvilli density, and intestinal goblet cell counts, as well as skin goblet cell counts. The yeast additives had no detrimental effects on the fish haematology, with no significant differences in haemoglobin concentration, red blood cell counts, and white blood cell counts among the treatment groups. Gene expression analysis revealed upregulation of il-1β and muc-2 in fish fed the ABY diet, indicating enhanced immune function and potentially mucosal protection. Intestinal microbiota analysis revealed Firmicutes as the most dominant phylum in all groups, followed by Actinobacteriota. Distinct bacterial community shifts were observed between the treatment groups, with a significant increase in the relative abundance of taxa such as Staphylococcus in yeast-supplemented diets and a significant decrease in Streptococcus and Weissella. Collectively, these findings suggest that the yeast additives, especially ABY, enhance gut health and immune function without compromising growth performance.
Collapse
Affiliation(s)
- Sheu G. Odu-Onikosi
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
- Department of Fisheries and Aquaculture, Lagos State University of Science and Technology, Ikorodu 104101, Nigeria
| | - Taofik A. Momoh
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Sherilyn T. Abarra
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Noah E. Wood
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Folasade D. Amulejoye
- Department of Fisheries and Aquaculture Technology, Olusegun Agagu University of Science and Technology, P.M.B. 353, Okitipupa 350105, Nigeria;
| | - Matthew Emery
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Glenn M. Harper
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Benjamin Eynon
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Victor Kuri
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | | | - Daniel L. Merrifield
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| |
Collapse
|
8
|
Kwikima MM. The biological and chemical water quality of retail bottled water brands: a case of Dodoma, Tanzania. NUTRITION & FOOD SCIENCE 2025; 55:165-179. [DOI: 10.1108/nfs-05-2024-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Purpose
Ensuring high water quality is crucial for safeguarding public health, as contaminated water can pose significant risks to consumers’ well-being. This study aims to evaluate the microbiological and chemical quality of bottled water brands commonly consumed in Dodoma, Tanzania.
Design/methodology/approach
A total of 36 samples from 12 brands were collected between January and March 2023 and analyzed for microbiological and general water quality parameters.
Findings
Microbial analysis found that 42% of brands tested positive for coliform bacteria, while opportunistic pathogens Aeromonas hydrophila and Pseudomonas aeruginosa were detected in 25% and 17% of samples, respectively. For chemical composition, 42% of brands exceeded the World Health Organization guideline value of 1.5 mg/L for fluoride. However, no other parameters exceeded national drinking water standards. Statistical analysis revealed significantly higher measured fluoride levels compared to values declared on product labels (paired t-test, p = 0.003). A moderate positive correlation between fluoride and conductivity (r = 0.52, p = 0.045) indicated possible geological influences on water chemistry.
Research limitations/implications
To enhance the study’s comprehensiveness, exploring temporal and spatial variations among water brands, including samples from typically clean environments such as supermarkets, could have been beneficial in identifying underlying factors. Additionally, investigating the entire manufacturing process, from production to end-user, could have provided insights into unforeseen deviations in quality. Furthermore, the use of pour plating techniques at 37°C for microbial analysis, while suitable for resource-limited settings, may not have fully captured coliform diversity compared to membrane filtration and differential temperature incubation as per standard methods. This could partly explain the detection of heterotrophs without higher coliform counts in some samples.
Practical implications
This study provides baseline data on the bacteriological and inorganic chemical quality of bottled water in Dodoma. Detectable microbial contaminants and significant exceedances of fluoride guidelines in some products raise public health concerns.
Originality/value
While existing studies focus on bottled water quality at production facilities, this research highlights the overlooked risks at retail points, where consumers are directly affected.
Collapse
|
9
|
Vávrová P, Janďourek O, Diepoltová A, Nachtigal P, Konečná K. The appropriate nutrient conditions for methicillin-resistant Staphylococcus aureus and Candida albicans dual-species biofilm formation in vitro. Sci Rep 2025; 15:183. [PMID: 39747199 PMCID: PMC11696109 DOI: 10.1038/s41598-024-83745-1] [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: 07/19/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Polymicrobial biofilms, the reason for most chronic wound infections, play a significant role in increasing antibiotic resistance. The in vivo effectiveness of the new anti-biofilm therapy is conditioned by the profound evaluation using appropriate in vitro biofilm models. Since nutrient availability is crucial for in vitro biofilm formation, this study is focused on the impact of four selected cultivation media on the properties of methicillin-resistant Staphylococcus aureus and Candida albicans dual-species biofilms. To reflect the wound environment, Tryptic soy broth, RPMI 1640 with and without glucose, and Lubbock medium were supplemented with different amounts of host effector molecules present in human plasma or sheep red blood cells. The study demonstrates that the Lubbock medium provided the most appropriate amount of nutrients regarding the biomass structure and the highest degree of tolerance to selected antimicrobials with the evident contribution of the biofilm matrix. Our results allow the rational employment of nutrition conditions within methicillin-resistant Staphylococcus aureus and Candida albicans dual-species biofilm formation in vitro for preclinical research. Additionally, one of the potential targets of a complex antibiofilm strategy, carbohydrates, was revealed since they are prevailing molecules in the matrices regardless of the cultivation media.
Collapse
Affiliation(s)
- Pavlína Vávrová
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Králové, 2089, Zborovská, Hradec Králové, 500 03, Czech Republic
| | - Ondřej Janďourek
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Králové, 2089, Zborovská, Hradec Králové, 500 03, Czech Republic
| | - Adéla Diepoltová
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Králové, 2089, Zborovská, Hradec Králové, 500 03, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Králové, 2089, Zborovská, Hradec Králové, 500 03, Czech Republic
| | - Klára Konečná
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Králové, 2089, Zborovská, Hradec Králové, 500 03, Czech Republic.
| |
Collapse
|
10
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
11
|
Jacobs S, Boccarella G, van den Berg P, Van Dijck P, Carolus H. Unlocking the potential of experimental evolution to study drug resistance in pathogenic fungi. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:48. [PMID: 39843963 PMCID: PMC11721431 DOI: 10.1038/s44259-024-00064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/15/2024] [Indexed: 01/24/2025]
Abstract
Exploring the dynamics and molecular mechanisms of antimicrobial drug resistance provides critical insights for developing effective strategies to combat it. This review highlights the potential of experimental evolution methods to study resistance in pathogenic fungi, drawing on insights from bacteriology and innovative approaches in mycology. We emphasize the versatility of experimental evolution in replicating clinical and environmental scenarios and propose that incorporating evolutionary modelling can enhance our understanding of antifungal resistance evolution. We advocate for a broader application of experimental evolution in medical mycology to improve our still limited understanding of drug resistance in fungi.
Collapse
Affiliation(s)
- Stef Jacobs
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Giorgio Boccarella
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Pieter van den Berg
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
- Evolutionary Modelling Group, Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
- KU Leuven One Health Institute, KU Leuven, Leuven, Belgium
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Parveen H, Mukhtar S, Albalawi MO, Khasim S, Ahmad A, Wani MY. Concomitant Inhibition and Collaring of Dual-Species Biofilms Formed by Candida auris and Staphylococcus aureus by Triazole Based Small Molecule Inhibitors. Pharmaceutics 2024; 16:1570. [PMID: 39771549 PMCID: PMC11677466 DOI: 10.3390/pharmaceutics16121570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Biofilm-associated infections, particularly those involving Candida auris and Staphylococcus aureus, pose significant challenges in clinical settings due to their resilience and resistance to conventional treatments. This study aimed to synthesize novel triazole derivatives containing a piperazine ring via click chemistry and evaluate their efficacy in disrupting biofilms formed by these pathogens. METHODS Triazole derivatives were synthesized using click chemistry techniques. The antimicrobial activity of the compounds was tested against planktonic cells of C. auris and S. aureus in single and dual-species culture conditions. Biofilm disruption efficacy was assessed, alongside the evaluation of physicochemical properties, oral bioavailability potential, and toxicity profiles. RESULTS The compound T3 demonstrated potent antimicrobial activity against planktonic cells of C. auris and S. aureus in both single and dual-species cultures. T3 exhibited significant efficacy in reducing microbial viability within biofilms formed by these pathogens. Physicochemical analyses revealed favorable solubility and permeability profiles, supporting its potential for oral bioavailability. Toxicity assessments showed a non-toxic profile, highlighting a promising safety margin for further development. CONCLUSIONS This study underscores the anti-biofilm properties of novel triazole-piperazine derivatives, particularly T3, against single and dual-species biofilms of C. auris and S. aureus. These findings position T3 as a promising candidate for developing therapies targeting polymicrobial infections and provide a foundation for future research into alternative strategies for combating biofilm-associated infections.
Collapse
Affiliation(s)
- Humaira Parveen
- Organic and Medicinal Chemistry Research Laboratory, Department of Chemistry, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.M.); (M.O.A.)
| | - Sayeed Mukhtar
- Organic and Medicinal Chemistry Research Laboratory, Department of Chemistry, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.M.); (M.O.A.)
| | - Mona O. Albalawi
- Organic and Medicinal Chemistry Research Laboratory, Department of Chemistry, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.M.); (M.O.A.)
| | - Syed Khasim
- Advanced Materials Research Laboratory, Department of Physics, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Aijaz Ahmad
- Department of Clinical Microbiology and Infectious Diseases, Faculty of Health Sciences, School of Pathology, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| |
Collapse
|
13
|
Thakur B, Kaur S, Tripathi M, Upadhyay SK. Exploring the potential of lactic acid bacteria and its molecular mechanism of action in the development of biosurfactants: Current finding and future outlook. Biotechnol Genet Eng Rev 2024; 40:4737-4768. [PMID: 37226486 DOI: 10.1080/02648725.2023.2216421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
Biosurfactants generated from lactic acid bacteria (LAB) offer an advantage over standard microbial surfactants due to their antifungal, antibacterial and antiviral capabilities. Many LAB strains have been related to the manufacture of biosurfactant, an essential chemical with uses in the treatment of a number of illnesses. Furthermore, their effectiveness as anti-adhesive agents against a diverse variety of pathogens proves their utility as anti-adhesive coating agents for medical insertional materials, reducing hospital infections without the need of synthetic drugs and chemicals. LAB produces both low and high molecular weight biosurfactants. Biosurfactants from L. pentosus, L. gasseri and L. jensenii have been reported to produce glycolipopeptides that comprise carbohydrates, proteins and lipids in the ratio of 1:3:6 with palmitic, stearic acid, and linoelaidic acid as the major fatty acid component, whereas L. plantarum has been reported to make surlactin due to the presence of non-ribosomal peptide synthetase genes (NRPS) genes. Antimicrobial activity of sophorolipids and rhamnolipids generated from LAB against B. subtilis, P. aeruginosa, S. epidermidis, Propionibacterium acnes and E. coli has been demonstrated. The safety of biosurfactants is being evaluated in compliance with a number of regulatory standards that emphasize the importance of safety in the pharmaceutical industry. This review attempts, for the first time, to provide a comprehensive evaluation of several approaches for the synthesis of biosurfactant-mediated molecular modulation in terms of their biological value. Future biosurfactant directions, as well as regulatory considerations that are crucial for the synthesis of biosurfactants from novel LAB, have also been explored.
Collapse
Affiliation(s)
- Babita Thakur
- Department of Biotechnology, Chandigarh University, Mohali, India
| | | | - Manikant Tripathi
- Biotechnology Program, Dr. Rammanohar Lohia Avadh University, Ayodhya, India
| | - Sudhir K Upadhyay
- Department of Environmental Science, Veer Bahadur Singh Purvanchal University, Jaunpur, India
| |
Collapse
|
14
|
Arévalo-Jaimes BV, Torrents E. Candida albicans enhances Staphylococcus aureus virulence by progressive generation of new phenotypes. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100316. [PMID: 39649408 PMCID: PMC11621768 DOI: 10.1016/j.crmicr.2024.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
Candida albicans and Staphylococcus aureus have been co-isolated from several biofilm-associated diseases, including those related to medical devices. This association confers advantages to both microorganisms, resulting in detrimental effects on the host. To elucidate this phenomenon, the present study investigated colony changes derived from non-physical interactions between C. albicans and S. aureus. We performed proximity assays by confronting colonies of the yeast and the bacteria on agar plates at six different distances for 9-10 days. We found that colony variants of S. aureus originated progressively after prolonged exposure to C. albicans proximity, specifically in response to pH neutralization of the media by the fungi. The new phenotypes of S. aureus were more virulent in a Galleria mellonella larvae model compared to colonies grown without C. albicans influence. This event was associated with an upregulation of RNA III and agrA expression, suggesting a role for α-toxin. Our findings indicate that C. albicans enhances S. aureus virulence by inducing the formation of more aggressive colonies. This highlights the importance of understanding the intricate connection between environmental responses, virulence and, fitness in S. aureus pathogenesis.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac Street 10, 08037, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal Street 647, 08028, Barcelona, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac Street 10, 08037, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal Street 647, 08028, Barcelona, Spain
| |
Collapse
|
15
|
Passos JCDS, Furtado Rodrigues AB, Alberto-Silva C, Costa MS. The arrangement of dual-species biofilms of Candida albicans and Issatchenkia orientalis can be modified by the medium: effect of Voriconazole. BIOFOULING 2024; 40:527-537. [PMID: 39115404 DOI: 10.1080/08927014.2024.2389848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/26/2024]
Abstract
Both Candida albicans and Issatchenkia orientalis have been isolated from different types of infections over the years. They have the ability to form communities of microorganisms known as biofilms. It has been demonstrated that the medium employed in studies may affect the biofilm development. The aim of this study was to investigate the arrangement of dual-species biofilms of C. albicans and I. orientalis cultivated on either RPMI-1640 or Sabouraud Dextrose Broth (SDB), as well as the inhibitory effect of Voriconazole (VRC). For the experiments performed, ATCC strains were used, and yeast-mixed suspensions were inoculated in 96-well plates with either RPMI-1640 or SDB, in the presence or absence of VRC. The results were observed by counting the number of CFU obtained from scraping off the biofilms produced and plating the content on CHROMagar Candida medium. It was observed that for all conditions tested the medium chosen affected the arrangement of dual-species biofilms: when RPMI-1640 was used, there was a prevalence of C. albicans, while the opposite was noted when SDB was used. It could be suggested that the medium and environment could regulate interactions between both yeast species, including the response to different antifungal drugs.
Collapse
Affiliation(s)
| | - Ana Beatriz Furtado Rodrigues
- Instituto de Pesquisa & Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, São José dos Campos, Brazil
| | - Carlos Alberto-Silva
- Experimental Morphophysiology Laboratory, Natural and Humanities Science Center (CCNH), Federal University of ABC - UFABC, São Bernardo do Campo, Brazil
| | - Maricilia Silva Costa
- Instituto de Pesquisa & Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP, São José dos Campos, Brazil
| |
Collapse
|
16
|
Nartop D, Hasanoğlu Özkan E, Öğütcü H, Kurnaz Yetim N, Özdemir İ. Novel α-N-heterocyclic thiosemicarbazone complexes: synthesis, characterization, and antimicrobial of properties investigation. RSC Adv 2024; 14:29308-29318. [PMID: 39285885 PMCID: PMC11403394 DOI: 10.1039/d4ra04002c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
In this paper, eight novel α-N-heterocyclic thiosemicarbazone complexes were synthesized in search of new biologically active compounds, and characterized via organic elemental analysis, nuclear magnetic resonance spectroscopy, infrared spectra, thermogravimetric analysis, ultraviolet-visible spectroscopy, molar conductance and magnetic susceptibility measurements. The in vitro antimicrobial activity of these complexes was examined against ten disease-causing pathogens: Gram-positive bacteria (Micrococcus luteus ATCC9341, Staphylococcus epidermidis ATCC12228, Bacillus cereus RSKK863) and Gram-negative bacteria (Pseudomonas aeroginosa ATCC27853, Klebsiella pneumonia ATCC27853, Enterobacter aerogenes ATCC51342, Salmonella typhi H NCTC9018394, Shigella dysenteria NCTC2966, Proteus vulgaris RSKK96026) and yeast (Candida albicans Y-1200-NIH). The results revealed that the α-N-heterocyclic thiosemicarbazone compounds showed potent activity. It was observed that all thiosemicarbazone complexes were more susceptible to Gram-negative strains based on the presence of an electron-withdrawing substituent (-Br/-Cl/-F). It was determined that thiosemicarbazone Cu2+complexes showed stronger antifungal effects.
Collapse
Affiliation(s)
- Dilek Nartop
- Department of Chemistry, Faculty of Arts and Sciences, Düzce University Düzce Turkiye
| | | | - Hatice Öğütcü
- Department of Field Crops, Faculty of Agriculture, Kırşehir Ahi Evran University Kırşehir Turkiye
| | - Nurdan Kurnaz Yetim
- Department of Chemistry, Faculty of Arts and Sciences, Kırklareli University Kırklareli Turkiye
| | - İnci Özdemir
- Kocaeli University, Izmit Vocational School, Property Protection and Security Department 41285 Kocaeli Turkiye
| |
Collapse
|
17
|
Thakur B, Kaur S, Dwibedi V, Albadrani GM, Al-Ghadi MQ, Abdel-Daim MM. Unveiling the antimicrobial and antibiofilm potential of biosurfactant produced by newly isolated Lactiplantibacillus plantarum strain 1625. Front Microbiol 2024; 15:1459388. [PMID: 39318434 PMCID: PMC11420119 DOI: 10.3389/fmicb.2024.1459388] [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: 07/04/2024] [Accepted: 08/09/2024] [Indexed: 09/26/2024] Open
Abstract
The present study aimed to characterize the biosurfactants synthesized by lactic acid bacteria (LAB) obtained from fermented foods, optimize the conditions for increasing the yield of biosurfactants and explore their antimicrobial and antibiofilm potential. Out of the 26 LAB isolates, isolate BS2 showed the highest biosurfactant production as indicated in the oil displacement test, drop collapse and emulsification activity. BS2 was identified as Lactiplantibacillus plantarum 1625 using 16S-rRNA gene sequencing and phylogenetic analysis. The biosurfactant produced by BS2 was identified as an anionic glycol-lipo-proteins by employing Fourier Transform Infrared Spectroscopy (FTIR) and Gas Chromatography-Mass Spectrometry (GC-MS) analysis. The biosurfactants produced by L. plantarum 1625 demonstrated strong antibacterial and antibiofilm characteristics against pathogenic strains such as Staphylococcus aureus MTCC 1049, Escherichia coli MTCC 1587, and Pseudomonas putida MTCC 1655. The minimal inhibition concentration value of antibacterial activity was found to be 0.1 mg/mL with the inhibition percentage ranging from 90 to 95%. Further, the effect of temperature, pH, and substrate composition on biosurfactant production was also studied to enhance it production using the Box-Behnken Design approach of Response surface methodology (RSM). Application of biosurfactant led to a considerable decrease in biofilm-forming harmful bacteria, as proven by scanning electron microscopy analysis. The results highlight the potential uses of biosurfactants in distinct industries, and biotechnological contexts, especially in the creation of new antimicrobial and antibiofilm agents.
Collapse
Affiliation(s)
- Babita Thakur
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Sukhminderjit Kaur
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Vagish Dwibedi
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Muath Q. Al-Ghadi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
18
|
Al-Hussein IAH, Keshavarzi F, Abd Aun Jwad BAA, Maroofi Naghadehi M, Hasan KMA. Association between IL6 rs1800795, IL10 rs1800871 and 1,800,872 polymorphisms with periodontitis. HUMAN GENE 2024; 41:201302. [DOI: 10.1016/j.humgen.2024.201302] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
|
19
|
Pasman R, Zhang J, Zaat SAJ, Brul S, Krom BP. A customizable and defined medium supporting culturing of Candida albicans, Staphylococcus aureus, and human oral epithelial cells. Appl Environ Microbiol 2024; 90:e0036024. [PMID: 39072650 PMCID: PMC11337806 DOI: 10.1128/aem.00360-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Candida albicans, an opportunistic oral pathogen, synergizes with Staphylococcus aureus, allowing bacteria to co-invade and systemically disseminate within the host. Studying human-microbe interactions creates the need for a universal culture medium that supports fungal, bacterial, and human cell culturing, while allowing sensitive analytical approaches such as OMICs and chromatography techniques. In this study, we established a fully defined, customizable adaptation of Dulbecco's modified Eagle medium (DMEM), allowing multi-kingdom culturing of S. aureus, C. albicans, and human oral cell lines, whereas minimal version of DMEM (mDMEM) did not support growth of S. aureus, and neither did supplementation with dextrose, MEM non-essential amino acids, pyruvate, and Glutamax. This new medium composition, designated as "mDMEM-DMP," promoted growth of all tested S. aureus strains. Addition of 25 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) further improved growth, while higher concentrations did not improve growth any further. Higher concentrations of HEPES did result in prolonged stabilization of medium pH. mDMEM-DMP promoted (hyphal) C. albicans monoculturing and co-culturing on both solid and semi-solid surfaces. In contrast to S. aureus, addition of HEPES reduced C. albicans maximum culture optical density (OD). Finally, only buffered mDMEM-DMP (100 mM HEPES) was successful in maintaining the metabolic activity of human oral Ca9-22 and HO1N1 cell lines for 24 hours. Altogether, our findings show that mDMEM-DMP is a versatile and potent culture medium for both microbial and human cell culturing, providing a customizable platform to study human as well as microbial molecular physiology and putative interactions. IMPORTANCE Interaction between microbes and the host are in the center of interest both in disease and in health. In order to study the interactions between microbes of different kingdoms and the host, alternative media are required. Synthetic media are useful as they allow addition of specific components. In addition, well-defined media are required if high-resolution analyses such as metabolomics and proteomics are desired. We describe the development of a synthetic medium to study the interactions between C. albicans, S. aureus, and human oral epithelial cells. Our findings show that mDMEM-DMP is a versatile and potent culture medium for both microbial and human cell culturing, providing a customizable platform to study human as well as microbial molecular physiology and putative interactions.
Collapse
Affiliation(s)
- Raymond Pasman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Jianbo Zhang
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Sebastian A. J. Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Stanley Brul
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Free University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
20
|
Mourad DF, Radwan S, Hamdy R, Elkhashab DM, Kamel MM, Abdel-Moneim AS, Kadry DY. Identification of Lower Respiratory Tract Pathogens in Cancer Patients: Insights into Fatal Outcomes. Microorganisms 2024; 12:1686. [PMID: 39203528 PMCID: PMC11356771 DOI: 10.3390/microorganisms12081686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
This study aimed to investigate LRTIs in cancer patients, focusing on pathogen distribution, and outcomes based on tumor types and antimicrobial treatments. The study included 110 cancer patients exhibiting symptoms of lower respiratory tract infections (LRTIs), consisting of 67 males and 43 females across a wide age range from under 1 year to over 60 years old. Exclusion of SARS-CoV-2 infection was conducted before admission. In addition to classical microbiological methods, fast-track detection using Multiplex Real-Time PCR was employed, utilizing the FTD-33 test kit. The findings revealed a diverse landscape of infections, notably Klebsiella pneumoniae, Haemophilus influenzae and Staphylococcus aureus. Parainfluenza 3 and 4 viruses, rhinovirus, influenza A subtype H1N1pdm09, influenza B and C viruses, HCoV-229, HCoV-OC43, and HCoV-HKU1 were infrequently detected. Furthermore, the existence of mixed infection highlighted the complexity of disease conditions in cancer patients. An analysis of antimicrobial treatment highlighted significant variations in fatal outcomes for carbapenem and colistimethate sodium. It was concluded that mixed infections were commonly identified as potential causes of LRTIs among cancer patients, while viral infections were less frequently detected. It underscores the complexity of antimicrobial treatment outcomes.
Collapse
Affiliation(s)
- Dalia F. Mourad
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Samah Radwan
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Rana Hamdy
- Pediatric Oncology Department, National Cancer Institute, Cairo University, Cairo 12613, Egypt
| | - Dina M. Elkhashab
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Mahmoud M. Kamel
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Ahmed S. Abdel-Moneim
- Department of Microbiology, College of Medicine, Taif University, Al-Taif 21944, Saudi Arabia
| | - Dalia Y. Kadry
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| |
Collapse
|
21
|
Barman S, Dey R, Ghosh S, Mukherjee R, Mukherjee S, Haldar J. Amino Acid-Conjugated Polymer-Silver Bromide Nanocomposites for Eradicating Polymicrobial Biofilms and Treating Burn Wound Infections. ACS Infect Dis 2024; 10:2999-3012. [PMID: 39082818 DOI: 10.1021/acsinfecdis.4c00342] [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: 08/10/2024]
Abstract
The rise in antimicrobial resistance, the increasing occurrence of bacterial, and fungal infections, and the challenges posed by polymicrobial biofilms necessitate the exploration of innovative therapeutic strategies. Silver-based antimicrobials have garnered attention for their broad-spectrum activity and multimodal mechanisms of action. However, their effectiveness against single-species or polymicrobial biofilms remains limited. In this study, we present the fabrication of polymer-silver bromide nanocomposites using amino acid conjugated polymers (ACPs) through a green and water-based in situ technique. The nanocomposite architecture facilitated prolonged and controlled release of the active components. Remarkably, the nanocomposites exhibited broad-spectrum activity against multidrug-resistant (MDR) human pathogenic bacteria (MIC = 2-16 μg/mL) and fungi (MIC = 1-8 μg/mL), while displaying no detectable toxicity to human erythrocytes (HC50 > 1024 μg/mL). In contrast to existing antimicrobials and silver-based therapies, the nanocomposite effectively eradicated bacterial, fungal, and polymicrobial biofilms, and prevented the development of microbial resistance due to their membrane-active properties. Furthermore, the lead polymer-silver bromide nanocomposite demonstrated a 99% reduction in the drug-resistant Pseudomonas aeruginosa burden in a murine model of burn wound infection, along with excellent in vivo biocompatibility.
Collapse
Affiliation(s)
- Swagatam Barman
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Rajib Dey
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Sreyan Ghosh
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Riya Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| |
Collapse
|
22
|
Gupta P, Meher MK, Tripathi S, Poluri KM. Nanoformulations for dismantling fungal biofilms: The latest arsenals of antifungal therapy. Mol Aspects Med 2024; 98:101290. [PMID: 38945048 DOI: 10.1016/j.mam.2024.101290] [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: 08/13/2023] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Globally, fungal infections have evolved as a strenuous challenge for clinicians, particularly in patients with compromised immunity in intensive care units. Fungal co-infection in Covid-19 patients has made the situation more formidable for healthcare practitioners. Surface adhered fungal population known as biofilm often develop at the diseased site to elicit antifungal tolerance and recalcitrant traits. Thus, an innovative strategy is required to impede/eradicate developed biofilm and avoid the formation of new colonies. The development of nanocomposite-based antibiofilm solutions is the most appropriate way to withstand and dismantle biofilm structures. Nanocomposites can be utilized as a drug delivery medium and for fabrication of anti-biofilm surfaces capable to resist fungal colonization. In this context, the present review comprehensively described different forms of nanocomposites and mode of their action against fungal biofilms. Amongst various nanocomposites, efficacy of metal/organic nanoparticles and nanofibers are particularly emphasized to highlight their role in the pursuit of antibiofilm strategies. Further, the inevitable concern of nanotoxicology has also been introduced and discussed with the exigent need of addressing it while developing nano-based therapies. Further, a list of FDA-approved nano-based antifungal formulations for therapeutic usage available to date has been described. Collectively, the review highlights the potential, scope, and future of nanocomposite-based antibiofilm therapeutics to address the fungal biofilm management issue.
Collapse
Affiliation(s)
- Payal Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India; Department of Biotechnology, Graphic Era (Demmed to be Unievrsity), Dehradun, 248001, Uttarakhand, India
| | - Mukesh Kumar Meher
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Shweta Tripathi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| |
Collapse
|
23
|
Jing Q, Liu R, Jiang Q, Liu Y, He J, Zhou X, Yu OY, Chu CH, Cheng L, Ren B, Li M. Staphylococcus aureus wraps around Candida albicans and synergistically escapes from Neutrophil extracellular traps. Front Immunol 2024; 15:1422440. [PMID: 39050841 PMCID: PMC11266059 DOI: 10.3389/fimmu.2024.1422440] [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: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Background NETs, a unique neutrophil immune mechanism, are vital in defending against microbial invasions. Understanding the mechanisms of co-infection by Candida albicans and Staphylococcus aureus, which often leads to higher mortality and poorer prognosis, is crucial for studying infection progression. Methods In our study, we established a mouse model of subcutaneous infection to characterize the inflammation induced by co-infection. By purifying and extracting NETs to interact with microorganisms, we delve into the differences in their interactions with various microbial species. Additionally, we investigated the differences in NETs production by neutrophils in response to single or mixed microorganisms through the interaction between neutrophils and these microorganisms. Furthermore, we analyzed the gene expression differences during co-infection using transcriptomics. Results In vivo, C. albicans infections tend to aggregate, while S. aureus infections are more diffuse. In cases of co-infection, S. aureus adheres to and wraps C. albicans. NETs exhibit strong killing capability against C. albicans but weaker efficacy against S. aureus. When NETs interact with mixed microorganisms, they preferentially target and kill the outer layer of S. aureus. In the early stages, neutrophils primarily rely on phagocytosis to kill S. aureus, but as the bacteria accumulate, they stimulate neutrophils to produce NETs. Interestingly, in the presence of neutrophils, S. aureus promotes the proliferation and hyphal growth of C. albicans. Conclusion Our research has showed substantial differences in the progression of co-infections compared to single-microbial infections, thereby providing scientific evidence for NETs as potential therapeutic targets in the treatment of co-infections.
Collapse
Affiliation(s)
- Qi Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qingsong Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yingshuang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jinzhi He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ollie Yiru Yu
- Faculty of Dentistry, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chun-Hung Chu
- Faculty of Dentistry, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Paul S, Todd OA, Eichelberger KR, Tkaczyk C, Sellman BR, Noverr MC, Cassat JE, Fidel PL, Peters BM. A fungal metabolic regulator underlies infectious synergism during Candida albicans-Staphylococcus aureus intra-abdominal co-infection. Nat Commun 2024; 15:5746. [PMID: 38982056 PMCID: PMC11233573 DOI: 10.1038/s41467-024-50058-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.
Collapse
Affiliation(s)
- Saikat Paul
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Olivia A Todd
- Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine Tkaczyk
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, USA
| | - Bret R Sellman
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, USA
| | - Mairi C Noverr
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul L Fidel
- Department of Oral and Craniofacial Biology, Louisiana State University Health - School of Dentistry, New Orleans, LA, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
25
|
Gourari-Bouzouina K, Boucherit-Otmani Z, Seghir A, Baba Ahmed-Kazi Tani ZZ, Bendoukha I, Benahmed A, Aissaoui M, Boucherit K. Evaluation of mixed biofilm production by Candida spp. and Staphylococcus aureus strains co-isolated from cystic fibrosis patients in northwest Algeria. Diagn Microbiol Infect Dis 2024; 109:116321. [PMID: 38677054 DOI: 10.1016/j.diagmicrobio.2024.116321] [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: 01/30/2024] [Revised: 04/13/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Cystic fibrosis patients' lungs are chronically colonized by multiple microbial species capable of forming biofilms. This study aimed to characterize the polymicrobial biofilm formed by Candida spp. and S. aureus, co-isolated from sputum samples of cystic fibrosis patients regarding microbial density, metabolic activity, and structure. 67 samples from 28 patients were collected with a 96% alteration rate. 34% showed alterations by both Candida spp. and Gram-positive bacteria, predominantly Candida spp. and S. aureus in 77% of cases, accounting for 6 associations. Biofilm biomass was quantified using the crystal violet assay, and metabolic activity was assessed using the MTT reduction assay. Scanning electron microscopy analyzed the C. tropicalis/S. aureus24 biofilm architecture. Candida spp. isolates demonstrated the ability to form mixed biofilms with S. aureus. The C. tropicalis/S. aureus24 association exhibited the highest production of biofilm and metabolic activity, along with the C. albicans17/C. rugosa/S. aureus7 in both single and mixed biofilms.
Collapse
Affiliation(s)
- Karima Gourari-Bouzouina
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria.
| | - Zahia Boucherit-Otmani
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| | - Abdelfettah Seghir
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| | - Zahira Zakia Baba Ahmed-Kazi Tani
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| | - Imene Bendoukha
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| | - Abdeselem Benahmed
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| | - Mohammed Aissaoui
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria; Department of Biology, Faculty of Sciences and Technology, University of Tamanghasset, Tamanghasset 11000, Algeria
| | - Kebir Boucherit
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LAPSAB), Department of Biology, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| |
Collapse
|
26
|
Kulshrestha A, Gupta P. Real-time biofilm detection techniques: advances and applications. Future Microbiol 2024; 19:1003-1016. [PMID: 38904296 PMCID: PMC11318681 DOI: 10.1080/17460913.2024.2350285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/29/2024] [Indexed: 06/22/2024] Open
Abstract
Microbial biofilms, complex assemblies enveloped in extracellular matrices, are significant contributors to various infections. Traditional in vitro biofilm characterization methods, though informative, often disrupt the biofilm structure. The need to address biofilm-related infections urgently emphasizes the importance of continuous monitoring and timely interventions. This review provides a focused examination of advancements in real-time biofilm detection techniques, specifically in electrochemical, optical and mechanical systems. The potential applications of real-time detection in managing and monitoring biofilm growth in industrial settings, preventing medical infections, comprehending biofilm dynamics and evaluating control strategies highlight the necessity for it. Crucially, the review emphasizes the importance of evaluating these methods for their accuracy and reliability in real-time biofilm detection, offering valuable insights for precise interventions across various applications.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh, India
| |
Collapse
|
27
|
Hamion G, Aucher W, Mercier A, Tewes F, Menard M, Bertaux J, Girardot M, Imbert C. Insights into betulinic acid as a promising molecule to fight the interkingdom biofilm Staphylococcus aureus-Candida albicans. Int J Antimicrob Agents 2024; 63:107166. [PMID: 38570017 DOI: 10.1016/j.ijantimicag.2024.107166] [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: 10/24/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
The demand for antibiofilm molecules has increased over several years due to their potential to fight biofilm-associated infections, such as those including the interkingdom Staphylococcus aureus-Candida albicans occurring in clinical settings worldwide. Recently, we identified a pentacyclic triterpenoid compound, betulinic acid, from invasive macrophytes, with interesting antibiofilm properties. The aim of the present study was to provide insights into the mechanism of action of betulinic acid against the clinically relevant bi-species S. aureus-C. albicans biofilms. Microscopy examinations, flow cytometry and crystal violet assays confirmed that betulinic acid was effective at damaging mature S. aureus-C. albicans biofilms or inhibiting their formation, reducing biofilm biomass by 70% on average and without microbicidal activity. The results suggested an action of betulinic acid on cell membranes, inducing changes in properties such as composition, hydrophobicity and fluidity as observed in C. albicans, which may hinder the early adhesion step, biofilm growth and the physical interactions of both microbial species. Further results of real-time polymerase chain reaction argued in favour of a reduction in S. aureus-C. albicans physical interaction due to betulinic acid by the modulation of biofilm-related gene expression, as observed in early stages of biofilm formation. This study revealed the potential of betulinic acid as a candidate agent for the prevention and treatment of S. aureus-C. albicans biofilm-related infections.
Collapse
Affiliation(s)
- Guillaume Hamion
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France.
| | - Willy Aucher
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France
| | - Anne Mercier
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France
| | - Frederic Tewes
- Pharmacology of Antimicrobial Agents and Antibioresistance, University of Poitiers, INSERM U1070, Poitiers, France
| | - Maëlenn Menard
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France
| | - Joanne Bertaux
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France
| | - Marion Girardot
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France
| | - Christine Imbert
- Ecology and Biology of Interactions, University of Poitiers, UMR CNRS 7267, Poitiers, France
| |
Collapse
|
28
|
Gourari-Bouzouina K, Boucherit-Otmani Z, Halla N, Seghir A, Baba Ahmed-Kazi Tani ZZ, Boucherit K. Exploring the dynamics of mixed-species biofilms involving Candida spp. and bacteria in cystic fibrosis. Arch Microbiol 2024; 206:255. [PMID: 38734793 DOI: 10.1007/s00203-024-03967-9] [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: 01/30/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Cystic fibrosis (CF) is an inherited disease that results from mutations in the gene responsible for the cystic fibrosis transmembrane conductance regulator (CFTR). The airways become clogged with thick, viscous mucus that traps microbes in respiratory tracts, facilitating colonization, inflammation and infection. CF is recognized as a biofilm-associated disease, it is commonly polymicrobial and can develop in biofilms. This review discusses Candida spp. and both Gram-positive and Gram-negative bacterial biofilms that affect the airways and cause pulmonary infections in the CF context, with a particular focus on mixed-species biofilms. In addition, the review explores the intricate interactions between fungal and bacterial species within these biofilms and elucidates the underlying molecular mechanisms that govern their dynamics. Moreover, the review addresses the multifaceted issue of antimicrobial resistance in the context of CF-associated biofilms. By synthesizing current knowledge and research findings, this review aims to provide insights into the pathogenesis of CF-related infections and identify potential therapeutic approaches to manage and combat these complex biofilm-mediated infections.
Collapse
Affiliation(s)
- Karima Gourari-Bouzouina
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LapSab), Department of Biology, Faculty of Sciences, University of Tlemcen, BP 119, 13000, Tlemcen, Algeria.
| | - Zahia Boucherit-Otmani
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LapSab), Department of Biology, Faculty of Sciences, University of Tlemcen, BP 119, 13000, Tlemcen, Algeria
| | - Noureddine Halla
- Laboratory of Biotoxicology, Pharmacognosy and Biological Recovery of Plants, Department of Biology, Faculty of Sciences, University of Moulay-Tahar, 20000, Saida, Algeria
| | - Abdelfettah Seghir
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LapSab), Department of Biology, Faculty of Sciences, University of Tlemcen, BP 119, 13000, Tlemcen, Algeria
| | - Zahira Zakia Baba Ahmed-Kazi Tani
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LapSab), Department of Biology, Faculty of Sciences, University of Tlemcen, BP 119, 13000, Tlemcen, Algeria
| | - Kebir Boucherit
- Antibiotics Antifungal Laboratory, Physical Chemistry, Synthesis and Biological Activity (LapSab), Department of Biology, Faculty of Sciences, University of Tlemcen, BP 119, 13000, Tlemcen, Algeria
| |
Collapse
|
29
|
Wnorowska S, Grzegorczyk A, Kurzepa J, Maggi F, Strzemski M. Fractionation of Carlina acaulis L. Root Methanolic Extract as a Promising Path towards New Formulations against Bacillus cereus and Methicillin-Resistant Staphylococcus aureus. Molecules 2024; 29:1939. [PMID: 38731430 PMCID: PMC11085459 DOI: 10.3390/molecules29091939] [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: 03/15/2024] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
The root of Carlina acaulis L. has been widely used in traditional medicine for its antimicrobial properties. In this study, the fractionation of methanol extract from the root was conducted. Four fractions (A, B, C, and D) were obtained and tested against a range of bacteria and fungi. The results showed promising antibacterial activity, especially against Bacillus cereus, where the minimal inhibitory concentration (MIC) was determined to be equal to 0.08 mg/mL and 0.16 mg/mL for heptane (fraction B) and ethyl acetate (fraction C), respectively. In the case of the methicillin-resistant Staphylococcus aureus (MRSA) ATCC 43300 strain, the same fractions yielded higher MIC values (2.5 and 5.0 mg/mL, respectively). This was accompanied by a lack of apparent cytotoxicity to normal human BJ foreskin fibroblasts, enterocytes derived from CaCo2 cells, and zebrafish embryos. Further analyses revealed the presence of bioactive chlorogenic acids in the fractionated extract, especially in the ethyl acetate fraction (C). These findings support the traditional use of the root from C. acaulis and pave the way for the development of new formulations for treating bacterial infections. This was further evaluated in a proof-of-concept experiment where fraction C was used in the ointment formulation, which maintained high antimicrobial activity against MRSA and displayed low toxicity towards cultured fibroblasts.
Collapse
Affiliation(s)
- Sylwia Wnorowska
- Department of Medical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Agnieszka Grzegorczyk
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Jacek Kurzepa
- Department of Medical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Filippo Maggi
- Chemistry Interdisciplinary Project (ChIP), School of Pharmacy, University of Camerino, Via Madonna Delle Carceri, 62032 Camerino, Italy;
| | - Maciej Strzemski
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
30
|
Manohar P, Loh B, Nachimuthu R, Leptihn S. Phage-antibiotic combinations to control Pseudomonas aeruginosa-Candida two-species biofilms. Sci Rep 2024; 14:9354. [PMID: 38653744 DOI: 10.1038/s41598-024-59444-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Phage-antibiotic combinations to treat bacterial infections are gaining increased attention due to the synergistic effects often observed when applying both components together. Most studies however focus on a single pathogen, although in many clinical cases multiple species are present at the site of infection. The aim of this study was to investigate the anti-biofilm activity of phage-antibiotic/antifungal combinations on single- and dual-species biofilms formed by P. aeruginosa and the fungal pathogen Candida albicans. The Pseudomonas phage Motto in combination with ciprofloxacin had significant anti-biofilm activity. We then compared biofilms formed by P. aeruginosa alone with the dual-species biofilms formed by bacteria and C. albicans. Here, we found that the phage together with the antifungal fluconazole was active against 6-h-old dual-species biofilms but showed only negligible activity against 24-h-old biofilms. This study lays the first foundation for potential therapeutic approaches to treat co-infections caused by bacteria and fungi using phage-antibiotic combinations.
Collapse
Affiliation(s)
- Prasanth Manohar
- School of Bioscience and Technology, Vellore Institute of Technology (VIT), Vellore, India
- Center for Phage Technology, Department of Biochemistry and Biophysics, Texas A&M AgriLife Research, Texas A&M University, College Station, TX, 77843, USA
| | - Belinda Loh
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103, Leipzig, Germany
| | - Ramesh Nachimuthu
- School of Bioscience and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Sebastian Leptihn
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103, Leipzig, Germany.
- Department of Biochemistry, Health and Medical University, Erfurt, Anger 66/73, 99084, Erfurt, Germany.
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
31
|
Robertson SN, Romero M, Fenn S, Kohler Riedi PL, Cámara M. Development, characterization, and evaluation of a simple polymicrobial colony biofilm model for testing of antimicrobial wound dressings. J Appl Microbiol 2024; 135:lxae042. [PMID: 38366933 DOI: 10.1093/jambio/lxae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/12/2024] [Accepted: 02/15/2024] [Indexed: 02/19/2024]
Abstract
Chronic wound infections are generally of polymicrobial nature with aerobic and anaerobic bacteria, as well as fungi frequently observed in them. Wound treatment involves a series of steps, including debridement of the wound, flushing, and often the use of multiple wound dressings many of which are antimicrobial. Yet, many wound dressings are tested versus single species of planktonic microbes, which fails to mirror the real-life presence of biofilms. AIMS Simple biofilm models are the first step to testing of any antimicrobial and wound dressing; therefore, the aim of this study was to develop and validate a simple polymicrobial colony biofilm wound model comprised of Pseudomonas aeruginosa, Staphylococcus aureus, and Candida albicans on RPMI-1640 agar. The model was then used to evaluate the topical disinfectant chlorohexidine and four commercially available wound dressings using the polymicrobial model. The model used was as a starting point to mimic debridement in clinical care of wounds and the effectiveness of wound dressings evaluated afterwards. METHODS AND RESULTS Planktonic assessment using AATCC100-2004 demonstrated that all antimicrobial wound dressings reduced the planktonic microbial burden below the limit of detection; however, when challenged with polymicrobial colony biofilms, silver wound dressings showed limited effectiveness (1-2 log CFU reductions). In contrast, a single iodine releasing wound dressing showed potent antibiofilm activity reducing all species CFUs below the limit of detection (>6-10 log) depending on the species. A disrupted biofilm model challenge was performed to represent the debridement of a wound and wound silver-based wound dressings were found to be marginally more effective than in whole colony biofilm challenges while the iodine containing wound dressing reduced microbial recovery below the limit of detection. CONCLUSIONS In this model, silver dressings were ineffective versus the whole colony biofilms but showed some recovery of activity versus the disrupted colony biofilm. The iodine wound dressing reduced the viability of all species below the level of detection. This suggests that mode of action of wound dressing should be considered for the type of biofilm challenge as should the clinical use, e.g. debridement.
Collapse
Affiliation(s)
- Shaun N Robertson
- National Biofilms Innovation Centre, School of Life Sciences, Biodiscovery Institute, University of Nottingham, NG7 2RD Nottingham, United Kingdom
| | - Manuel Romero
- National Biofilms Innovation Centre, School of Life Sciences, Biodiscovery Institute, University of Nottingham, NG7 2RD Nottingham, United Kingdom
- Department of Microbiology and Parasitology, Faculty of Biology-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Samuel Fenn
- National Biofilms Innovation Centre, School of Life Sciences, Biodiscovery Institute, University of Nottingham, NG7 2RD Nottingham, United Kingdom
- Schools of Microbiology and Medicine, University College Cork, and APC Microbiome Ireland, Cork T12 TP07, Ireland
| | | | - Miguel Cámara
- National Biofilms Innovation Centre, School of Life Sciences, Biodiscovery Institute, University of Nottingham, NG7 2RD Nottingham, United Kingdom
| |
Collapse
|
32
|
Hefny ZA, Ji B, Elsemman IE, Nielsen J, Van Dijck P. Transcriptomic meta-analysis to identify potential antifungal targets in Candida albicans. BMC Microbiol 2024; 24:66. [PMID: 38413885 PMCID: PMC10898158 DOI: 10.1186/s12866-024-03213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Candida albicans is a fungal pathogen causing human infections. Here we investigated differential gene expression patterns and functional enrichment in C. albicans strains grown under different conditions. METHODS A systematic GEO database search identified 239 "Candida albicans" datasets, of which 14 were selected after rigorous criteria application. Retrieval of raw sequencing data from the ENA database was accompanied by essential metadata extraction from dataset descriptions and original articles. Pre-processing via the tailored nf-core pipeline for C. albicans involved alignment, gene/transcript quantification, and diverse quality control measures. Quality assessment via PCA and DESeq2 identified significant genes (FDR < = 0.05, log2-fold change > = 1 or <= -1), while topGO conducted GO term enrichment analysis. Exclusions were made based on data quality and strain relevance, resulting in the selection of seven datasets from the SC5314 strain background for in-depth investigation. RESULTS The meta-analysis of seven selected studies unveiled a substantial number of genes exhibiting significant up-regulation (24,689) and down-regulation (18,074). These differentially expressed genes were further categorized into 2,497 significantly up-regulated and 2,573 significantly down-regulated Gene Ontology (GO) IDs. GO term enrichment analysis clustered these terms into distinct groups, providing insights into the functional implications. Three target gene lists were compiled based on previous studies, focusing on central metabolism, ion homeostasis, and pathogenicity. Frequency analysis revealed genes with higher occurrence within the identified GO clusters, suggesting their potential as antifungal targets. Notably, the genes TPS2, TPS1, RIM21, PRA1, SAP4, and SAP6 exhibited higher frequencies within the clusters. Through frequency analysis within the GO clusters, several key genes emerged as potential targets for antifungal therapies. These include RSP5, GLC7, SOD2, SOD5, SOD1, SOD6, SOD4, SOD3, and RIM101 which exhibited higher occurrence within the identified clusters. CONCLUSION This comprehensive study significantly advances our understanding of the dynamic nature of gene expression in C. albicans. The identification of genes with enhanced potential as antifungal drug targets underpins their value for future interventions. The highlighted genes, including TPS2, TPS1, RIM21, PRA1, SAP4, SAP6, RSP5, GLC7, SOD2, SOD5, SOD1, SOD6, SOD4, SOD3, and RIM101, hold promise for the development of targeted antifungal therapies.
Collapse
Affiliation(s)
- Zeinab Abdelmoghis Hefny
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Kasteelpark Arenberg 31, Leuven, B-3001, Belgium
| | - Boyang Ji
- BioInnovation Institute, Ole Maaløes Vej 3, Copenhagen, DK2200, Denmark
| | - Ibrahim E Elsemman
- Department of Information Systems, Faculty of Computers and Information, Assiut University, Assiut, 2071515, Egypt
| | - Jens Nielsen
- BioInnovation Institute, Ole Maaløes Vej 3, Copenhagen, DK2200, Denmark.
- Department of Life Sciences, Chalmers University of Technology, Kemivägen 10, SE41296, Gothenburg, SE41296, Sweden.
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Kasteelpark Arenberg 31, Leuven, B-3001, Belgium.
| |
Collapse
|
33
|
Li P, Li Y, Zhang Y, Zhu S, Pei Y, Zhang Q, Liu J, Bao J, Sun M. A dynamic nomogram to predict invasive fungal super-infection during healthcare-associated bacterial infection in intensive care unit patients: an ambispective cohort study in China. Front Cell Infect Microbiol 2024; 14:1281759. [PMID: 38469345 PMCID: PMC10925706 DOI: 10.3389/fcimb.2024.1281759] [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: 08/23/2023] [Accepted: 02/08/2024] [Indexed: 03/13/2024] Open
Abstract
Objectives Invasive fungal super-infection (IFSI) is an added diagnostic and therapeutic dilemma. We aimed to develop and assess a nomogram of IFSI in patients with healthcare-associated bacterial infection (HABI). Methods An ambispective cohort study was conducted in ICU patients with HABI from a tertiary hospital of China. Predictors of IFSI were selected by both the least absolute shrinkage and selection operator (LASSO) method and the two-way stepwise method. The predictive performance of two models built by logistic regression was internal-validated and compared. Then external validity was assessed and a web-based nomogram was deployed. Results Between Jan 1, 2019 and June 30, 2023, 12,305 patients with HABI were screened in 14 ICUs, of whom 372 (3.0%) developed IFSI. Among the fungal strains causing IFSI, the most common was C.albicans (34.7%) with a decreasing proportion, followed by C.tropicalis (30.9%), A.fumigatus (13.9%) and C.glabrata (10.1%) with increasing proportions year by year. Compared with LASSO-model that included five predictors (combination of priority antimicrobials, immunosuppressant, MDRO, aCCI and S.aureus), the discriminability of stepwise-model was improved by 6.8% after adding two more predictors of COVID-19 and microbiological test before antibiotics use (P<0.01).And the stepwise-model showed similar discriminability in the derivation (the area under curve, AUC=0.87) and external validation cohorts (AUC=0.84, P=0.46). No significant gaps existed between the proportion of actual diagnosed IFSI and the frequency of IFSI predicted by both two models in derivation cohort and by stepwise-model in external validation cohort (P=0.16, 0.30 and 0.35, respectively). Conclusion The incidence of IFSI in ICU patients with HABI appeared to be a temporal rising, and our externally validated nomogram will facilitate the development of targeted and timely prevention and control measures based on specific risks of IFSI.
Collapse
Affiliation(s)
- Peng Li
- Department of Infection Control, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Li
- Department of Infection Control, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Youjian Zhang
- Department of Infection Control, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Shichao Zhu
- Central Intensive Care Unit, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongju Pei
- Respiratory Intensive Care Unit, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Zhang
- Department of Clinical Microbiology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Junping Liu
- Department of Infectious Disease, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Junzhe Bao
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Mingjie Sun
- Department of Infection Control, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
34
|
Paul S, Todd OA, Eichelberger KR, Tkaczyk C, Sellman BR, Noverr MC, Cassat JE, Fidel PL, Peters BM. A fungal metabolic regulator underlies infectious synergism during Candida albicans - Staphylococcus aureus intra-abdominal co-infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580531. [PMID: 38405692 PMCID: PMC10888754 DOI: 10.1101/2024.02.15.580531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) uncovered synergistic lethality that was driven by Candida -induced upregulation of functional S. aureus ⍺-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken and revealed that zcf13 Δ/Δ failed to drive augmented ⍺-toxin or lethal synergism during co-infection. Using a combination of transcriptional and phenotypic profiling approaches, ZCF13 was shown to regulate genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments revealed that ribose inhibited the staphylococcal agr quorum sensing system and concomitantly repressed toxicity. Unlike wild-type C. albicans , zcf13 Δ/Δ was unable to effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13 Δ/Δ mutant fully restored pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.
Collapse
|
35
|
Baharvand R, Fallah F, Jafari P, Azimi L. Co-colonization of methicillin-resistant Staphylococcus aureus and Candida spp. in children with malignancies. AMB Express 2024; 14:22. [PMID: 38351284 PMCID: PMC10864235 DOI: 10.1186/s13568-024-01667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
This study aimed to evaluate the interaction between methicillin-resistant Staphylococcus aureus(MRSA) and Candida spp. in the oral cavity of children with malignancies under chemotherapy. We evaluated the expression level of Als3p and mecA in Candida spp. and MRSA strains in both single colonization and co-colonization condition. Oral and nasal samples were collected by dry sponge swabs in 10 ml of sterile phosphate-buffered saline. The MRSA and Candida spp. was confirmed using the PCR method and mecA and Als3p genes, respectively. The SYBR Green-based quantitative real-time PCR was used to evaluate the relative expression levels of mecA and Als3p genes in MRSA and Candida spp., respectively. The frequency of S. aureus in oral-only and nasal-only swab samples were 14.1% (n = 24/170). 58.3% (n = 14/24) and 29.2% (n = 7/24) of S. aureus isolated from oral and nasal samples were MRSA, respectively. Among Candida species, C. albicans (n = 28/170; 16.5%) had the highest frequency. The oral co-colonization of MRSA and Candida spp. was detected in 4.7% (n = 8/170) patients. The overall average of gene expression levels among all Candida spp. and MRSA isolates indicated that the mecA and Als3p genes expression increased six and two times in co-colonization conditions compared to single colonization conditions, respectively. Our findings revealed the importance of polymicrobial infection in clinical settings and stated that it is possible that Candida spp. facilitates the infection of S. aureus and can lead to systemic infection in co-colonized patients.
Collapse
Affiliation(s)
- Raziyeh Baharvand
- Department of Microbiology, Faculty of Science, Islamic Azad University, Arak branch, Arak, Iran
| | - Fatemeh Fallah
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvaneh Jafari
- Department of Microbiology, Faculty of Science, Islamic Azad University, Arak branch, Arak, Iran.
| | - Leila Azimi
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
To TT, Oparaugo NC, Kheshvadjian AR, Nelson AM, Agak GW. Understanding Type 3 Innate Lymphoid Cells and Crosstalk with the Microbiota: A Skin Connection. Int J Mol Sci 2024; 25:2021. [PMID: 38396697 PMCID: PMC10888374 DOI: 10.3390/ijms25042021] [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: 12/01/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Innate lymphoid cells (ILCs) are a diverse population of lymphocytes classified into natural killer (NK) cells, ILC1s, ILC2s, ILC3s, and ILCregs, broadly following the cytokine secretion and transcription factor profiles of classical T cell subsets. Nonetheless, the ILC lineage does not have rearranged antigen-specific receptors and possesses distinct characteristics. ILCs are found in barrier tissues such as the skin, lungs, and intestines, where they play a role between acquired immune cells and myeloid cells. Within the skin, ILCs are activated by the microbiota and, in turn, may influence the microbiome composition and modulate immune function through cytokine secretion or direct cellular interactions. In particular, ILC3s provide epithelial protection against extracellular bacteria. However, the mechanism by which these cells modulate skin health and homeostasis in response to microbiome changes is unclear. To better understand how ILC3s function against microbiota perturbations in the skin, we propose a role for these cells in response to Cutibacterium acnes, a predominant commensal bacterium linked to the inflammatory skin condition, acne vulgaris. In this article, we review current evidence describing the role of ILC3s in the skin and suggest functional roles by drawing parallels with ILC3s from other organs. We emphasize the limited understanding and knowledge gaps of ILC3s in the skin and discuss the potential impact of ILC3-microbiota crosstalk in select skin diseases. Exploring the dialogue between the microbiota and ILC3s may lead to novel strategies to ameliorate skin immunity.
Collapse
Affiliation(s)
- Thao Tam To
- Division of Dermatology, Department of Medicine, University of California (UCLA), Los Angeles, CA 90095, USA
| | - Nicole Chizara Oparaugo
- Division of Dermatology, Department of Medicine, University of California (UCLA), Los Angeles, CA 90095, USA
| | - Alexander R. Kheshvadjian
- Division of Dermatology, Department of Medicine, University of California (UCLA), Los Angeles, CA 90095, USA
| | - Amanda M. Nelson
- Department of Dermatology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - George W. Agak
- Division of Dermatology, Department of Medicine, University of California (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
37
|
da Silva CR, Sá LGDAV, Andrade Neto JBD, Barroso FDD, Cabral VPDF, Rodrigues DS, da Silva LJ, Lima ISP, Pérez L, Ramos da Silva A, Moreira DR, Ricardo NMPS, Nobre HV. Antimicrobial potential of a biosurfactant gel for the prevention of mixed biofilms formed by fluconazole-resistant C. albicans and methicillin-resistant S. aureus in catheters. BIOFOULING 2024; 40:165-176. [PMID: 38425095 DOI: 10.1080/08927014.2024.2324028] [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: 11/08/2023] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Dual-species biofilms formed by Candida albicans and Staphylococcus aureus have high virulence and drug resistance. In this context, biosurfactants produced by Pseudomonas aeruginosa have been widely studied, of which a new derivative (RLmix_Arg) stands out for possible application in formulations. The objective of this study was to evaluate the antibiofilm activity of RLmix_Arg, both alone and incorporated in a gel prepared with Pluronic F-127, against dual-species biofilms of fluconazole-resistant C. albicans (FRCA) and methicillin-resistant S. aureus (MRSA) in impregnated catheters. Broth microdilution tests, MTT reduction assays of mature biofilms, impregnation of RLmix_Arg and its gel in peripheral venous catheters, durability tests and scanning electron microscopy (SEM) were performed. RLmix_Arg showed antimicrobial activity against Candida spp. and S. aureus, by reducing the cell viability of mixed biofilms of FRCA and MRSA, and preventing their formation in a peripheral venous catheter. The incorporation of this biosurfactant in the Pluronic F-127 gel considerably enhanced its antibiofilm activity. Thus, RLmix_Arg has potential application in gels for impregnation in peripheral venous catheters, helping to prevent development of dual-species biofilms of FRCA and MRSA.
Collapse
Affiliation(s)
- Cecília Rocha da Silva
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lívia Gurgel do Amaral Valente Sá
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | - João Batista de Andrade Neto
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | - Fátima Daiana Dias Barroso
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | - Vitória Pessoa de Farias Cabral
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Daniel Sampaio Rodrigues
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lisandra Juvêncio da Silva
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Iri Sandro Pampolha Lima
- Departament of Pharmacology, School of Medicine, Federal University of Ceará, Barbalha, CE, Brazil
| | - Lourdes Pérez
- Department of Surfactants and Nanobiotechnology, IQAC-CSIC, Barcelona, Spain
| | | | - Denise Ramos Moreira
- Laboratory of Polymers and Materials Innovation, Department of Organic and Inorganic Chemistry, Sciences Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Nágila Maria Pontes Silva Ricardo
- Laboratory of Polymers and Materials Innovation, Department of Organic and Inorganic Chemistry, Sciences Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélio Vitoriano Nobre
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Center of Drug Research and Development, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
38
|
Vasileva EA, Berdyshev DV, Mishchenko NP, Gerasimenko AV, Menchinskaya ES, Pislyagin EA, Chingizova EA, Kaluzhskiy LA, Dautov SS, Fedoreyev SA. Phanogracilins A-C, New Bibenzochromenones of Crinoid Phanogenia gracilis (Hartlaub, 1890). Biomolecules 2024; 14:151. [PMID: 38397388 PMCID: PMC10887160 DOI: 10.3390/biom14020151] [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: 12/28/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Three new bibenzochromenones named phanogracilins A-C (1-3) were isolated from the crinoid Phanogenia gracilis. The structure of 1 was established using X-ray crystallography as 5,5',6,6',8,8'-hexahydroxy-2,2'-dipropyl-4H,4'H-[7,9'-bibenzo[g]chromene]-4,4'-dione. This allowed us to assign reliably 2D NMR signals for compound 1 and subsequently for its isomer 2 that differed in the connecting position of two benzochromenone moieties (7,10' instead of 7,9'), and compound for 3 that differed in the length of the aliphatic chain of one of the fragments. Compound 4 was derived from 1 in alkaline conditions, and its structure was elucidated as 5,5',6',8,8'-pentahydroxy-2,2'-dipropyl-4H,4'H-[7,9'-bibenzo[g]chromene]-4,4',6,9-tetraone. Even though compounds 1-4 did not contain stereo centers, they possessed notable optical activity due to sterical hindrances, which limited the internal rotation of two benzochromenone fragments around C(7)-C(9'/10') bonds. Isolated bibenzochromenones 1-4 were tested for their antiradical, neuroprotective and antimicrobial activities. Compounds 1, 3 and 4 demonstrated significant antiradical properties towards ABTS radicals higher than the positive control trolox. Compounds 1 and 4 exhibited moderate neuroprotective activity, increasing the viability of rotenone-treated Neuro-2a cells at a concentration of 1 µM by 9.8% and 11.8%, respectively. Compounds 1 and 3 at concentrations from 25 to 100 μM dose-dependently inhibited the growth of Gram-positive bacteria S. aureus and yeast-like fungi C. albicans, and they also prevented the formation of their biofilms. Compounds 2 and 4 exhibited low antimicrobial activity.
Collapse
Affiliation(s)
- Elena A. Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| | - Dmitrii V. Berdyshev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| | - Natalia P. Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| | - Andrey V. Gerasimenko
- Institute of Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia;
| | - Ekaterina S. Menchinskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| | - Evgeniy A. Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| | - Ekaterina A. Chingizova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| | | | - Salim Sh. Dautov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690041, Russia;
| | - Sergey A. Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.B.); (N.P.M.); (E.S.M.); (E.A.P.); (E.A.C.); (S.A.F.)
| |
Collapse
|
39
|
Baxter KJ, Sargison FA, Fitzgerald JR, McConnell G, Hoskisson PA. Time-lapse mesoscopy of Candida albicans and Staphylococcus aureus dual-species biofilms reveals a structural role for the hyphae of C. albicans in biofilm formation. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001426. [PMID: 38261525 PMCID: PMC10866020 DOI: 10.1099/mic.0.001426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
Polymicrobial infection with Candida albicans and Staphylococcus aureus may result in a concomitant increase in virulence and resistance to antimicrobial drugs. This enhanced pathogenicity phenotype is mediated by numerous factors, including metabolic processes and direct interaction of S. aureus with C. albicans hyphae. The overall structure of biofilms is known to contribute to their recalcitrance to treatment, although the dynamics of direct interaction between species and how it contributes to pathogenicity is poorly understood. To address this, a novel time-lapse mesoscopic optical imaging method was developed to enable the formation of C. albicans/S. aureus whole dual-species biofilms to be followed. It was found that yeast-form or hyphal-form C. albicans in the biofilm founder population profoundly affects the structure of the biofilm as it matures. Different sub-populations of C. albicans and S. aureus arise within each biofilm as a result of the different C. albicans morphotypes, resulting in distinct sub-regions. These data reveal that C. albicans cell morphology is pivotal in the development of global biofilm architecture and the emergence of colony macrostructures and may temporally influence synergy in infection.
Collapse
Affiliation(s)
- Katherine J. Baxter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Fiona A. Sargison
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
40
|
Cox CA, Manavathu EK, Wakade S, Myntti M, Vazquez JA. Efficacy of biofilm disrupters against Candida auris and other Candida species in monomicrobial and polymicrobial biofilms. Mycoses 2024; 67:e13684. [PMID: 38214428 DOI: 10.1111/myc.13684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 10/19/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
Candida species are now considered global threats by the CDC and WHO. Candida auris specifically is on the critical pathogen threat list along with Candida albicans. In addition, it is not uncommon to find Candida spp. in a mixed culture with bacterial organisms, especially Staphylococcus aureus producing polymicrobial infections. To eradicate these organisms from the environment and from patient surfaces, surface agents such as chlorhexidine (CHD) and Puracyn are used. Biofilm disrupters (BDs) are novel agents with a broad spectrum of antimicrobial activity and have been used in the management of chronic wounds and to sterilise environmental surfaces for the past several years. The goal of this study was to evaluate BDs (BlastX, Torrent, NSSD) and CHD against Candida spp. and S. aureus using zone of inhibition assays, biofilm and time-kill assays. All BDs and CHD inhibited C. auris growth effectively in a concentration-dependent manner. Additionally, CHD and the BDs showed excellent antimicrobial activity within polymicrobial biofilms. A comparative analysis of the BDs and CHD against C. auris and C. albicans using biofilm kill-curves showed at least 99.999% killing. All three BDs and CHD have excellent activity against different Candida species, including C. auris. However, one isolate of C. auris in a polymicrobial biofilm assay showed resistance/tolerance to CHD, but not to the BDs. The fungicidal activity of these novel agents will be valuable in eradicating surface colonisation of Candida spp, especially C. auris from colonised environmental surfaces and from wounds in colonised patients.
Collapse
Affiliation(s)
- Claudia A Cox
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Elias K Manavathu
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Sushama Wakade
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | | | - Jose A Vazquez
- Division of Infectious Diseases, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
41
|
Al Mousa AA, Abouelela ME, Al Ghamidi NS, Abo-Dahab Y, Mohamed H, Abo-Dahab NF, Hassane AMA. Anti-Staphylococcal, Anti-Candida, and Free-Radical Scavenging Potential of Soil Fungal Metabolites: A Study Supported by Phenolic Characterization and Molecular Docking Analysis. Curr Issues Mol Biol 2023; 46:221-243. [PMID: 38248318 PMCID: PMC10814734 DOI: 10.3390/cimb46010016] [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: 12/04/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/23/2024] Open
Abstract
Staphylococcus and Candida are recognized as causative agents in numerous diseases, and the rise of multidrug-resistant strains emphasizes the need to explore natural sources, such as fungi, for effective antimicrobial agents. This study aims to assess the in vitro anti-staphylococcal and anti-candidal potential of ethyl acetate extracts from various soil-derived fungal isolates. The investigation includes isolating and identifying fungal strains as well as determining their antioxidative activities, characterizing their phenolic substances through HPLC analysis, and conducting in silico molecular docking assessments of the phenolics' binding affinities to the target proteins, Staphylococcus aureus tyrosyl-tRNA synthetase and Candida albicans secreted aspartic protease 2. Out of nine fungal species tested, two highly potent isolates were identified through ITS ribosomal gene sequencing: Aspergillus terreus AUMC 15447 and A. nidulans AUMC 15444. Results indicated that A. terreus AUMC 15447 and A. nidulans AUMC 15444 extracts effectively inhibited S. aureus (concentration range: 25-0.39 mg/mL), with the A. nidulans AUMC 15444 extract demonstrating significant suppression of Candida spp. (concentration range: 3.125-0.39 mg/mL). The A. terreus AUMC 15447 extract exhibited an IC50 of 0.47 mg/mL toward DPPH radical-scavenging activity. HPLC analysis of the fungal extracts, employing 18 standards, revealed varying degrees of detected phenolics in terms of their presence and quantities. Docking investigations highlighted rutin as a potent inhibitor, showing high affinity (-16.43 kcal/mol and -12.35 kcal/mol) for S. aureus tyrosyl-tRNA synthetase and C. albicans secreted aspartic protease 2, respectively. The findings suggest that fungal metabolites, particularly phenolics, hold significant promise for the development of safe medications to combat pathogenic infections.
Collapse
Affiliation(s)
- Amal A. Al Mousa
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 145111, Riyadh 4545, Saudi Arabia;
| | - Mohamed E. Abouelela
- Department of Pharmacognosy, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo P.O. Box 11884, Egypt;
| | - Nadaa S. Al Ghamidi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 145111, Riyadh 4545, Saudi Arabia;
| | | | - Hassan Mohamed
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt; (H.M.); (N.F.A.-D.)
| | - Nageh F. Abo-Dahab
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt; (H.M.); (N.F.A.-D.)
| | - Abdallah M. A. Hassane
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt; (H.M.); (N.F.A.-D.)
| |
Collapse
|
42
|
Wang T, Jia Z, An C, Ren P, Yang Y, Wang W, Su L. The Protective Effect of Auricularia cornea var. Li. Polysaccharide on Alcoholic Liver Disease and Its Effect on Intestinal Microbiota. Molecules 2023; 28:8003. [PMID: 38138493 PMCID: PMC10745760 DOI: 10.3390/molecules28248003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/26/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
This study's objective was to examine the protective effect and mechanism of a novel polysaccharide (AYP) from Auricularia cornea var. Li. on alcoholic liver disease in mice. AYP was extracted from the fruiting bodies of Auricularia cornea var. Li. by enzymatic extraction and purified by DEAE-52 and Sephacryl S-400. Structural features were determined using high-performance liquid chromatography, ion exchange chromatography and Fourier-transform infrared analysis. Additionally, alcoholic liver disease (ALD) mice were established to explore the hepatoprotective activity of AYP (50, 100 and 200 mg/kg/d). Here, our results showed that AYP presented high purity with a molecular weight of 4.64 × 105 Da. AYP was composed of galacturonic acid, galactose, glucose, arabinose, mannose, xylose, rhamnose, ribos, glucuronic acid and fucose (molar ratio: 39.5:32.9:23.6:18.3:6.5:5.8:5.8:3.3:2:1.1). Notably, AYP remarkably reduced liver function impairment (alanine aminotransferase (ALT), aspartate aminotransferase (AST), triglyceride (TG), total cholesterol (TC)), nitric oxide (NO) and malondialdehyde (MDA) of the liver and enhanced the activity of antioxidant enzymes (superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and glutathione (gGSH)) in mice with ALD. Meanwhile, the serum level of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) were reduced in ALD mice treated by AYP. Furthermore, the AYPH group was the most effective and was therefore chosen to further investigate its effect on the intestinal microbiota (bacteria and fungi) of ALD mice. Based on 16s rRNA and ITS-1 sequencing data, AYP influenced the homeostasis of intestinal microbiota to mitigate the damage of ALD mice, possibly by raising the abundance of favorable microbiota (Muribaculaceae, Lachnospiraceae and Kazachstania) and diminishing the abundance of detrimental microbiota (Lactobacillus, Mortierella and Candida). This discovery opens new possibilities for investigating physiological activity in A. cornea var. Li. and provides theoretical references for natural liver-protecting medication research.
Collapse
Affiliation(s)
- Tianci Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (T.W.); (Z.J.)
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China;
| | - Zikun Jia
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (T.W.); (Z.J.)
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China;
| | - Canghai An
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China;
| | - Ping Ren
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yiting Yang
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Wanting Wang
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Ling Su
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (T.W.); (Z.J.)
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China;
| |
Collapse
|
43
|
Eichelberger KR, Paul S, Peters BM, Cassat JE. Candida-bacterial cross-kingdom interactions. Trends Microbiol 2023; 31:1287-1299. [PMID: 37640601 PMCID: PMC10843858 DOI: 10.1016/j.tim.2023.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
While the fungus Candida albicans is a common colonizer of healthy humans, it is also responsible for mucosal infections and severe invasive disease. Understanding the mechanisms that allow C. albicans to exist as both a benign commensal and as an invasive pathogen have been the focus of numerous studies, and recent findings indicate an important role for cross-kingdom interactions on C. albicans biology. This review highlights how C. albicans-bacteria interactions influence healthy polymicrobial community structure, host immune responses, microbial pathogenesis, and how dysbiosis may lead to C. albicans infection. Finally, we discuss how cross-kingdom interactions represent an opportunity to identify new antivirulence compounds that target fungal infections.
Collapse
Affiliation(s)
- Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Saikat Paul
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
44
|
Vaculík O, Bernatová S, Rebrošová K, Samek O, Šilhan L, Růžička F, Šerý M, Šiler M, Ježek J, Zemánek P. Rapid identification of pathogens in blood serum via Raman tweezers in combination with advanced processing methods. BIOMEDICAL OPTICS EXPRESS 2023; 14:6410-6421. [PMID: 38420303 PMCID: PMC10898560 DOI: 10.1364/boe.503628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/06/2023] [Accepted: 10/21/2023] [Indexed: 03/02/2024]
Abstract
Pathogenic microbes contribute to several major global diseases that kill millions of people every year. Bloodstream infections caused by these microbes are associated with high morbidity and mortality rates, which are among the most common causes of hospitalizations. The search for the "Holy Grail" in clinical diagnostic microbiology, a reliable, accurate, low cost, real-time, and easy-to-use diagnostic method, is one of the essential issues in clinical practice. These very critical conditions can be met by Raman tweezers in combination with advanced analysis methods. Here, we present a proof-of-concept study based on Raman tweezers combined with spectral mixture analysis that allows for the identification of microbial strains directly from human blood serum without user intervention, thus eliminating the influence of a data analyst.
Collapse
Affiliation(s)
- Ondřej Vaculík
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Silvie Bernatová
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Katarína Rebrošová
- Department of Microbiology, Faculty of Medicine of Masaryk University and St. Anne's, University Hospital, Pekařská 53, Brno, 65691, Czech Republic
| | - Ota Samek
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Lukáš Šilhan
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Filip Růžička
- Department of Microbiology, Faculty of Medicine of Masaryk University and St. Anne's, University Hospital, Pekařská 53, Brno, 65691, Czech Republic
| | - Mojmír Šerý
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Martin Šiler
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Jan Ježek
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| | - Pavel Zemánek
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, Brno, 61264, Czech Republic
| |
Collapse
|
45
|
Khan F, Jeong GJ, Javaid A, Thuy Nguyen Pham D, Tabassum N, Kim YM. Surface adherence and vacuolar internalization of bacterial pathogens to the Candida spp. cells: Mechanism of persistence and propagation. J Adv Res 2023; 53:115-136. [PMID: 36572338 PMCID: PMC10658324 DOI: 10.1016/j.jare.2022.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The co-existence of Candida albicans with the bacteria in the host tissues and organs displays interactions at competitive, antagonistic, and synergistic levels. Several pathogenic bacteria take advantage of such types of interaction for their survival and proliferation. The chemical interaction involves the signaling molecules produced by the bacteria or Candida spp., whereas the physical attachment occurs by involving the surface proteins of the bacteria and Candida. In addition, bacterial pathogens have emerged to internalize inside the C. albicans vacuole, which is one of the inherent properties of the endosymbiotic relationship between the bacteria and the eukaryotic host. AIM OF REVIEW The interaction occurring by the involvement of surface protein from diverse bacterial species with Candida species has been discussed in detail in this paper. An in silico molecular docking study was performed between the surface proteins of different bacterial species and Als3P of C. albicans to explain the molecular mechanism involved in the Als3P-dependent interaction. Furthermore, in order to understand the specificity of C. albicans interaction with Als3P, the evolutionary relatedness of several bacterial surface proteins has been investigated. Furthermore, the environmental factors that influence bacterial pathogen internalization into the Candida vacuole have been addressed. Moreover, the review presented future perspectives for disrupting the cross-kingdom interaction and eradicating the endosymbiotic bacterial pathogens. KEY SCIENTIFIC CONCEPTS OF REVIEW With the involvement of cross-kingdom interactions and endosymbiotic relationships, the bacterial pathogens escape from the environmental stresses and the antimicrobial activity of the host immune system. Thus, the study of interactions between Candida and bacterial pathogens is of high clinical significance.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Aqib Javaid
- Department of Biotechnology and Bioinformatics, University of Hyderabad, India
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
46
|
Katrak C, Garcia BA, Dornelas-Figueira LM, Nguyen M, Williams RB, Lorenz MC, Abranches J. Catalase produced by Candida albicans protects Streptococcus mutans from H 2O 2 stress-one more piece in the cross-kingdom synergism puzzle. mSphere 2023; 8:e0029523. [PMID: 37607054 PMCID: PMC10597455 DOI: 10.1128/msphere.00295-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 06/30/2023] [Indexed: 08/24/2023] Open
Abstract
Co-infection with Streptococcus mutans and Candida albicans is associated with dental caries, and their co-cultivation results in enhanced biofilm matrix production that contributes to increased virulence and caries risk. Moreover, the catalase-negative S. mutans demonstrates increased oxidative stress tolerance when co-cultivated in biofilms with C. albicans, a catalase-producing yeast. Here, we sought to obtain mechanistic insights into the increased H2O2 tolerance of S. mutans when co-cultivated with clinical isolates of Candida glabrata, Candida tropicalis, and C. albicans. Additionally, the C. albicans SC5314 laboratory strain, its catalase mutant (SC5314Δcat1), and S. mutans UA159 and its glucosyltransferase B/C mutant (UA159ΔgtfB/C) were grown as single- and dual-species biofilms. Time-kill assays revealed that upon acute H2O2 challenge, the survival rates of S. mutans in dual-species biofilms with the clinical isolates and C. albicans SC5314 were greater than when paired with SC5314Δcat1 or as a single-species biofilm. Importantly, this protection was independent of glucan production through S. mutans GtfB/C. Transwell assays and treatment with H2O2-pre-stimulated C. albicans SC5314 supernatant revealed that this protection is contact-dependent. Biofilm stability assays with sublethal H2O2 or peroxigenic Streptococcus A12 challenge resulted in biomass reduction of single-species S. mutans UA159 and dual-species with SC5314Δcat1 biofilms compared to UA159 biofilms co-cultured with C. albicans SC5314. S. mutans oxidative stress genes were upregulated in single-species biofilms when exposed to H2O2, but not when S. mutans was co-cultivated with C. albicans SC5314. Here, we uncovered a novel, contact-dependent, synergistic interaction in which the catalase of C. albicans protects S. mutans against H2O2. IMPORTANCE It is well established that co-infection with the gram-positive caries-associated bacterium Streptococcus mutans and the yeast pathobiont Candida albicans results in aggressive forms of caries in humans and animal models. Together, these microorganisms form robust biofilms through enhanced production of extracellular polysaccharide matrix. Further, co-habitation in a biofilm community appears to enhance these microbes' tolerance to environmental stressors. Here, we show that catalase produced by C. albicans protects S. mutans from H2O2 stress in a biofilm matrix-independent manner. Our findings uncovered a novel synergistic trait between these two microorganisms that could be further exploited for dental caries prevention and control.
Collapse
Affiliation(s)
- Callahan Katrak
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Bruna A. Garcia
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
- Department of Restorative Dental Sciences, University of Florida College of Dentistry, Gainesville, Florida, USA
| | | | - Mary Nguyen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Robert B. Williams
- Department of Microbiology and Molecular Genetics, McGovern Medical School, Houston, Texas, USA
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, McGovern Medical School, Houston, Texas, USA
| | - Jacqueline Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| |
Collapse
|
47
|
Tsikopoulos A, Tsikopoulos K, Meroni G, Drago L, Triaridis S, Papaioannidou P. Strategies for Inhibition of Biofilm Formation on Silicone Rubber Voice Prostheses: A Systematic Review. J Voice 2023:S0892-1997(23)00222-9. [PMID: 37625903 DOI: 10.1016/j.jvoice.2023.07.015] [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: 04/04/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Lifetime elongation of the silicone voice rubber prostheses by inhibition of biofilm formation is a primary objective in voice restoration of laryngectomized patients. This systematic review sought to explore the existing strategies in this direction. MATERIALS We conducted a systematic search of both in vitro and in vivo literature published in PubMed, Scopus, and Cochrane Central Register of Controlled Trials, until December 31, 2022, for published and unpublished trials assessing the strategies for inhibiting biofilm formation on silicone rubber voice prostheses, and appraised quality assessment with the modified Consolidated Standards of Reporting Trials tool. We analyzed the infection prevention capacity of the included antibacterial and antifungal agents. RESULTS The qualitative synthesis showed that both surface modification methods and prophylactic treatment of silicone rubber voice prostheses present adequate antibiofilm activity. Of note, the majority of the suggested prosthetic surfaces were not chronically exposed to both human fluids and biofilm-forming microorganisms. CONCLUSION Various experimental methods provide promising antibiofilm activity and, thus, possible lifespan elongation of silicone rubber voice prostheses.
Collapse
Affiliation(s)
- Alexios Tsikopoulos
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Konstantinos Tsikopoulos
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gabriele Meroni
- One Health Unit, Department of Biomedical, Surgical and Dental Sciences, School of Medicine, University of Milan, Milan, Italy
| | - Lorenzo Drago
- Laboratory of Clinical Microbiology & Microbiome, Department of Biomedical Sciences for Health, School of Medicine, University of Milan, Milan, Italy
| | - Stefanos Triaridis
- 1st Department of Otorhinolaryngology - Head and Neck Surgery, AHEPA University General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paraskevi Papaioannidou
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
48
|
Tsikopoulos A, Tsikopoulos K, Meroni G, Gravalidis C, Soukouroglou P, Chatzimoschou A, Drago L, Triaridis S, Papaioannidou P. Νanomaterial-Loaded Polymer Coating Prevents the In Vitro Growth of Candida albicans Biofilms on Silicone Biomaterials. Antibiotics (Basel) 2023; 12:1103. [PMID: 37508199 PMCID: PMC10376674 DOI: 10.3390/antibiotics12071103] [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: 05/08/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Early failure of silicone voice prostheses resulting from fungal colonization and biofilm formation poses a major concern in modern ear nose throat surgery. Therefore, developing new infection prevention techniques to prolong those implants' survivorship is crucial. We designed an in vitro laboratory study to include nanomaterial-enhanced polymer coating with a plasma spraying technique against Candida albicans growth to address this issue. The anti-biofilm effects of high- and low-dose Al2O3 nanowire and TiO2 nanoparticle coatings were studied either alone or in conjunction with each other using checkerboard testing. It was demonstrated that both nanomaterials were capable of preventing fungal biofilm formation regardless of the anti-fungal agent concentration (median absorbance for high-dose Al2O3-enhanced polymer coating was 0.176 [IQR = 0.207] versus control absorbance of 0.805 [IQR = 0.381], p = 0.003 [98% biofilm reduction]; median absorbance for high-dose TiO2-enhanced polymer coating was 0.186 [IQR = 0.024] versus control absorbance of 0.766 [IQR = 0.458], p < 0.001 [93% biofilm reduction]). Furthermore, synergy was revealed when the Bliss model was applied. According to the findings of this work, it seems that simultaneous consideration of Al2O3 and TiO2 could further increase the existing antibiofilm potential of these nanomaterials and decrease the likelihood of localized toxicity.
Collapse
Affiliation(s)
- Alexios Tsikopoulos
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (P.P.)
| | - Konstantinos Tsikopoulos
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (P.P.)
| | - Gabriele Meroni
- One Health Unit, Department of Biomedical, Surgical and Dental Sciences, School of Medicine, University of Milan, 20133 Milan, Italy;
| | | | | | | | - Lorenzo Drago
- Laboratory of Clinical Microbiology & Microbiome, Department of Biomedical Sciences for Health, School of Medicine, University of Milan, 20133 Milan, Italy;
| | - Stefanos Triaridis
- 1st Department of Otorhinolaryngology-Head and Neck Surgery, AHEPA General Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Paraskevi Papaioannidou
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (P.P.)
| |
Collapse
|
49
|
Friedrich B, Tietze R, Dümig M, Sover A, Boca MA, Schreiber E, Band J, Janko C, Krappmann S, Alexiou C, Lyer S. Magnetic Removal of Candida albicans Using Salivary Peptide-Functionalized SPIONs. Int J Nanomedicine 2023; 18:3231-3246. [PMID: 37337577 PMCID: PMC10276999 DOI: 10.2147/ijn.s409559] [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: 02/21/2023] [Accepted: 05/07/2023] [Indexed: 06/21/2023] Open
Abstract
Purpose Magnetic separation of microbes can be an effective tool for pathogen identification and diagnostic applications to reduce the time needed for sample preparation. After peptide functionalization of superparamagnetic iron oxide nanoparticles (SPIONs) with an appropriate interface, they can be used for the separation of sepsis-associated yeasts like Candida albicans. Due to their magnetic properties, the magnetic extraction of the particles in the presence of an external magnetic field ensures the accumulation of the targeted yeast. Materials and Methods In this study, we used SPIONs coated with 3-aminopropyltriethoxysilane (APTES) and functionalized with a peptide originating from GP340 (SPION-APTES-Pep). For the first time, we investigate whether this system is suitable for the separation and enrichment of Candida albicans, we investigated its physicochemical properties and by thermogravimetric analysis we determined the amount of peptide on the SPIONs. Further, the toxicological profile was evaluated by recording cell cycle and DNA degradation. The separation efficiency was investigated using Candida albicans in different experimental settings, and regrowth experiments were carried out to show the use of SPION-APTES-Pep as a sample preparation method for the identification of fungal infections. Conclusion SPION-APTES-Pep can magnetically remove more than 80% of the microorganism and with a high selective host-pathogen distinction Candida albicans from water-based media and about 55% in blood after 8 minutes processing without compromising effects on the cell cycle of human blood cells. Moreover, the separated fungal cells could be regrown without any restrictions.
Collapse
Affiliation(s)
- Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rainer Tietze
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michaela Dümig
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Alexandru Sover
- Faculty of Engineering, Ansbach University of Applied Sciences, Ansbach, Germany
| | - Marius-Andrei Boca
- Faculty of Engineering, Ansbach University of Applied Sciences, Ansbach, Germany
| | - Eveline Schreiber
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Band
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sven Krappmann
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Controlled Nanomaterials, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
50
|
Kulshrestha A, Gupta P. Combating polymicrobial biofilm: recent approaches. Folia Microbiol (Praha) 2023:10.1007/s12223-023-01070-y. [PMID: 37310652 DOI: 10.1007/s12223-023-01070-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/04/2023] [Indexed: 06/14/2023]
Abstract
The polymicrobial biofilm (PMBF) is formed when microbes from multiple species co-aggregate into an envelope made of extra polymeric substances (EPS) that keep the microbes safe from external stresses. The formation of PMBF has been linked to a variety of human infections, including cystic fibrosis, dental caries, urinary tract infections, etc. Multiple microbial species co-aggregation during an infection results in a recalcitrant biofilm formation, which is a seriously threatening phenomenon. It is challenging to treat polymicrobial biofilms since they contain multiple microbes which show drug resistance to various antibiotics/antifungals. The present study discusses various approaches by which an antibiofilm compound works. Depending on their mode of action, antibiofilm compounds can block the adhesion of cells to one another, modify membranes/walls, or disrupt quorum-sensing systems.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology, Raipur, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology, Raipur, India.
| |
Collapse
|