1
|
Retore YI, Lucini F, Simionatto S, Rossato L. Synergistic Antifungal Activity of Pentamidine and Auranofin Against Multidrug-Resistant Candida auris. Mycopathologia 2025; 190:41. [PMID: 40360957 DOI: 10.1007/s11046-025-00948-4] [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: 11/29/2024] [Accepted: 04/17/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Candida auris is a significant clinical concern due to its ability to cause outbreaks in healthcare settings and its common resistance to current treatments. This highlights the need for alternative therapies. Drug repurposing offers a promising approach, and the combination of pentamidine (antiprotozoal) and auranofin (anti-rheumatic) has shown potential antifungal activity against Candida species, including C. auris. This study aimed to evaluate the antifungal activity of pentamidine and auranofin, both individually and in combination, against C. auris. METHODS Minimum Inhibitory Concentrations (MICs) were determined following CLSI guidelines, and drug interactions were assessed using the checkerboard microdilution method. Additional evaluations included growth inhibition, antibiofilm activity, cell damage, sorbitol protection, and efflux pump inhibition. Nucleotide leakage and cell membrane permeability were analyzed using biochemical assays. In vivo efficacy was tested using a Tenebrio molitor larvae model infected with C. auris. RESULTS The MICs of pentamidine against C. auris ranged from 16 to 128 μg/mL, showing fungicidal activity. The combination with auranofin had a synergistic effect (FICI: 0.37) and exhibited a fungistatic effect in growth inhibition assays. Auranofin was most effective at inhibiting biofilm formation. Pentamidine impaired mitochondrial function, leading to cellular respiration issues and membrane damage. Efflux pump assays indicated activation by both drugs, potentially influencing resistance. In vivo tests showed both drugs significantly improved survival rates in infected larvae compared to fluconazole. CONCLUSION In conclusion, pentamidine and auranofin, either individually or in combination, are promising treatments for C. auris and warrant further research into optimal dosing and combination strategies.
Collapse
Affiliation(s)
- Yasmim Isabel Retore
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Rodovia Dourados - Itahum km 12, Cidade Universitária, Dourados, Mato Grosso do Sul, CEP: 79804970, Brazil
| | - Fabíola Lucini
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Rodovia Dourados - Itahum km 12, Cidade Universitária, Dourados, Mato Grosso do Sul, CEP: 79804970, Brazil
| | - Simone Simionatto
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Rodovia Dourados - Itahum km 12, Cidade Universitária, Dourados, Mato Grosso do Sul, CEP: 79804970, Brazil
| | - Luana Rossato
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Rodovia Dourados - Itahum km 12, Cidade Universitária, Dourados, Mato Grosso do Sul, CEP: 79804970, Brazil.
| |
Collapse
|
2
|
Vitali V, Zineddu S, Messori L. Metal compounds as antimicrobial agents: 'smart' approaches for discovering new effective treatments. RSC Adv 2025; 15:748-753. [PMID: 39802470 PMCID: PMC11712697 DOI: 10.1039/d4ra07449a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Due to their considerable chemical diversity, metal compounds are attracting increasing and renewed attention from the scientific and medical communities as potential antimicrobial agents to combat the growing problem of antibiotic resistance. The development of metal compounds as antimicrobial agents typically follows classical drug discovery procedures and suffers from the same problems; indeed, these procedures can be very expensive and time-consuming, and carry an intrinsically high risk of failure. Here, we show how some established drug discovery approaches can be conveniently and successfully applied to antimicrobial metal compounds to provide some shortcuts for faster clinical translation of new treatments. Specifically, we refer to (i) drug repurposing, (ii) drug combination and (iii) drug targeting by bioconjugation; some relevant examples will be illustrated.
Collapse
Affiliation(s)
- Valentina Vitali
- Department of Chemistry "Ugo Schiff", University of Florence Sesto Fiorentino 50019 Italy
| | - Stefano Zineddu
- Department of Chemistry "Ugo Schiff", University of Florence Sesto Fiorentino 50019 Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence Sesto Fiorentino 50019 Italy
| |
Collapse
|
3
|
Pengfei S, Yifan Y, Shasha L, Shaowei G, Guanqing H, Dan X, Yong W. Repurposing pinaverium bromide against Staphylococcus and its biofilms with new mechanisms. AMB Express 2024; 14:141. [PMID: 39718732 DOI: 10.1186/s13568-024-01809-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
Antibiotic resistance by methicillin-resistant Staphylococcus aureus (MRSA) is an urgent threat to human health. The biofilm and persister cells formation ability of MRSA and Staphylococcus epidermidis often companied with extremely high antimicrobial resistance. Pinaverium bromide (PVB) is an antispasmodic compound mainly used for irritable bowel syndrome. Here we demonstrate that PVB could rapidly kill MRSA and S. epidermidis planktonic cells and persister cells avoiding resistance occurrence. Moreover, by crystal violet staining, viable cells counting and SYTO9/PI staining, PVB exhibited strong biofilm inhibition and eradication activities on the 96-well plates, glass surface or titanium discs. And the synergistic antimicrobial effects were observed between PVB and conventional antibiotics (ampicillin, oxacillin, and cefazolin). Mechanism study demonstrated the antimicrobial and antibiofilm effects by PVB were mainly mediated by proton motive force disrupting as well as reactive oxygen species inducing. Although, relatively poor pharmacokinetics were observed by systemic use, PVB could significantly reduce the viable bacterial cell loads and inflammatory infiltration in abscess in vivo caused by the biofilm forming strain ATCC 43,300. In all, our results indicated that PVB could be an alternative antimicrobial reagent for the treatment of MRSA, S. epidermidis and its biofilm related skin and soft tissue infections.
Collapse
Affiliation(s)
- She Pengfei
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Yang Yifan
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Liu Shasha
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Guo Shaowei
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China
| | - Huan Guanqing
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China
| | - Xiao Dan
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China
| | - Wu Yong
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China.
| |
Collapse
|
4
|
Coscione F, Zineddu S, Vitali V, Fondi M, Messori L, Perrin E. The Many Lives of Auranofin: How an Old Anti-Rheumatic Agent May Become a Promising Antimicrobial Drug. Antibiotics (Basel) 2024; 13:652. [PMID: 39061334 PMCID: PMC11274207 DOI: 10.3390/antibiotics13070652] [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: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Auranofin (AF) is a gold-based compound with a well-known pharmacological and toxicological profile, currently used in the treatment of some severe forms of rheumatoid arthritis. Over the last twenty years, AF has also been repurposed as antiviral, antitumor, and antibacterial drug. In this review we focused on the antibacterial properties of AF, specifically researching the minimal inhibitory concentrations (MIC) of AF in both mono- and diderm bacteria reported so far in literature. AF proves to be highly effective against monoderm bacteria, while diderm are far less susceptible, probably due to the outer membrane barrier. We also reported the current mechanistic hypotheses concerning the antimicrobial properties of AF, although a conclusive description of its antibacterial mode of action is not yet available. Even if its mechanism of action has not been fully elucidated yet and further studies are required to optimize its delivery strategy, AF deserves additional investigation because of its unique mode of action and high efficacy against a wide range of pathogens, which could lead to potential applications in fighting antimicrobial resistance and improving therapeutic outcomes in infectious diseases.
Collapse
Affiliation(s)
- Francesca Coscione
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| | - Stefano Zineddu
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Valentina Vitali
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Marco Fondi
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Elena Perrin
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| |
Collapse
|
5
|
Mathuria A, Ali N, Kataria N, Mani I. Drug repurposing for fungal infections. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:59-78. [PMID: 38942545 DOI: 10.1016/bs.pmbts.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
The rise of multidrug-resistant bacteria is a well-recognized threat to world health, necessitating the implementation of effective treatments. This issue has been identified as a top priority on the global agenda by the World Health Organization. Certain strains, such as Candida glabrata, Candida krusei, Candida lusitaniae, Candida auris, select cryptococcal species, and opportunistic Aspergillus or Fusarium species, have significant intrinsic resistance to numerous antifungal medicines. This inherent resistance and subsequent suboptimal clinical outcomes underscore the critical imperative for enhanced therapeutic alternatives and management protocols. The challenge of effectively treating fungal infections, compounded by the protracted timelines involved in developing novel drugs, underscores the pressing need to explore alternative therapeutic avenues. Among these, drug repurposing emerges as a particularly promising and expeditious solution, providing cost-effective solutions and safety benefits. In the fight against life-threatening resistant fungal infections, the idea of repurposing existing medications has encouraged research into both established and new compounds as a last-resort therapy. This chapter seeks to provide a comprehensive overview of contemporary antifungal drugs, as well as their key resistance mechanisms. Additionally, it seeks to provide insight into the antimicrobial properties of non-traditional drugs, thereby offering a holistic perspective on the evolving landscape of antifungal therapeutics.
Collapse
Affiliation(s)
- Anshu Mathuria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Namra Ali
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India
| | - Naina Kataria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
6
|
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
|
7
|
da Silva CR, Rebouças JDDO, Cabral VPDF, Rodrigues DS, Barbosa AD, Moreira LEA, Barroso FDD, Coutinho TDNP, de Lima EA, de Andrade CR, de Andrade Neto JB, Lima ISP, Nobre Júnior HV, Gurgel do Amaral Valente Sá L. Promising activity of etomidate against mixed biofilms of fluconazole-resistant Candida albicans and methicillin-resistant Staphylococcus aureus. J Med Microbiol 2024; 73. [PMID: 38385528 DOI: 10.1099/jmm.0.001810] [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: 02/23/2024] Open
Abstract
Introduction. Candida albicans and Staphylococcus aureus are recognized for their development of resistance and biofilm formation. New therapeutic alternatives are necessary in this context.Hypothesis. Etomidate shows potential application in catheters against mixed biofilms of fluconazole-resistant C. albicans and methicillin-resistant S. aureus (MRSA).Aim. The present study aimed to evaluate the activity of etomidate against mixed biofilms of fluconazole-resistant C. albicans and MRSA.Methodology. The action of etomidate against mature biofilms was verified through the evaluation of biomass and cell viability, and its ability to prevent biofilm formation in peripheral venous catheters was determined based on counts of colony forming units (c.f.u.) and confirmed by morphological analysis through scanning electron microscopy (SEM).Results. Etomidate generated a reduction (P<0.05) in biomass and cell viability starting from a concentration of 250 µg ml-1. In addition, it showed significant ability to prevent the formation of mixed biofilms in a peripheral venous catheter, as shown by a reduction in c.f.u. SEM revealed that treatment with etomidate caused substantial damage to the fungal cells.Conclusion. The results showed the potential of etomidate against polymicrobial biofilms of fluconazole-resistant C. albicans and MRSA.
Collapse
Affiliation(s)
- Cecília Rocha da Silva
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Vitória Pessoa de Farias Cabral
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Daniel Sampaio Rodrigues
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Amanda Dias Barbosa
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lara Elloyse Almeida Moreira
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Fátima Daiana Dias Barroso
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | | | - Elaine Aires de Lima
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - João Batista de Andrade Neto
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | - Iri Sandro Pampolha Lima
- Department of Pharmacology, School of Medicine, Federal University of Ceará, Barbalha, CE, Brazil
| | - Hélio Vitoriano Nobre Júnior
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lívia Gurgel do Amaral Valente Sá
- School of Pharmacy, Laboratory of Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| |
Collapse
|
8
|
Di Matteo V, Di Filippo MF, Ballarin B, Gentilomi GA, Bonvicini F, Panzavolta S, Cassani MC. Cellulose/Zeolitic Imidazolate Framework (ZIF-8) Composites with Antibacterial Properties for the Management of Wound Infections. J Funct Biomater 2023; 14:472. [PMID: 37754886 PMCID: PMC10532010 DOI: 10.3390/jfb14090472] [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: 07/18/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Metal-organic frameworks (MOFs) are a class of crystalline porous materials with outstanding physical and chemical properties that make them suitable candidates in many fields, such as catalysis, sensing, energy production, and drug delivery. By combining MOFs with polymeric substrates, advanced functional materials are devised with excellent potential for biomedical applications. In this research, Zeolitic Imidazolate Framework 8 (ZIF-8), a zinc-based MOF, was selected together with cellulose, an almost inexhaustible polymeric raw material produced by nature, to prepare cellulose/ZIF-8 composite flat sheets via an in-situ growing single-step method in aqueous media. The composite materials were characterized by several techniques (IR, XRD, SEM, TGA, ICP, and BET) and their antibacterial activity as well as their biocompatibility in a mammalian model system were investigated. The cellulose/ZIF-8 samples remarkably inhibited the growth of Gram-positive and Gram-negative reference strains, and, notably, they proved to be effective against clinical isolates of Staphylococcus epidermidis and Pseudomonas aeruginosa presenting different antibiotic resistance profiles. As these pathogens are of primary importance in skin diseases and in the delayed healing of wounds, and the cellulose/ZIF-8 composites met the requirements of biological safety, the herein materials reveal a great potential for use as gauze pads in the management of wound infections.
Collapse
Affiliation(s)
- Valentina Di Matteo
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Viale del Risorgimento 4, 40136 Bologna, Italy; (V.D.M.); (B.B.)
| | - Maria Francesca Di Filippo
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (M.F.D.F.); (S.P.)
| | - Barbara Ballarin
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Viale del Risorgimento 4, 40136 Bologna, Italy; (V.D.M.); (B.B.)
- Center for Industrial Research—Fonti Rinnovabili, Ambiente, Mare e Energia CIRI FRAME, University of Bologna, Viale del Risorgimento 2, 40136 Bologna, Italy
- Center for Industrial Research—Advanced Applications in Mechanical Engineering and Materials Technology CIRI MAM, University of Bologna, Viale del Risorgimento 2, 40136 Bologna, Italy
| | - Giovanna Angela Gentilomi
- Department of Pharmacy and Biotechnology, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Francesca Bonvicini
- Department of Pharmacy and Biotechnology, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Silvia Panzavolta
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (M.F.D.F.); (S.P.)
- Center for Industrial Research—Advanced Applications in Mechanical Engineering and Materials Technology CIRI MAM, University of Bologna, Viale del Risorgimento 2, 40136 Bologna, Italy
| | - Maria Cristina Cassani
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Viale del Risorgimento 4, 40136 Bologna, Italy; (V.D.M.); (B.B.)
- Health Sciences and Technologies—Interdepartmental Center for Industrial Research (HST–ICIR), Alma Mater Studiorum—University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy
| |
Collapse
|
9
|
Tharmalingam N, Xu S, Felix LO, Roy B, Xian M, Mylonakis E, Fuchs BB. Gold complex compounds that inhibit drug-resistant Staphylococcus aureus by targeting thioredoxin reductase. FRONTIERS IN ANTIBIOTICS 2023; 2:1179354. [PMID: 39816643 PMCID: PMC11732138 DOI: 10.3389/frabi.2023.1179354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/22/2023] [Indexed: 01/18/2025]
Abstract
Introduction There is a significant need for new antimicrobial compounds that are effective against drug-resistant microbes. Thioredoxin reductase (TrxR) is critical in redox homeostasis and was identified as a potential drug target and confirmed through inhibition by compounds auranofin and Bay11-7085. Methods Additional TrxR inhibitors were designed and found to exhibit antimicrobial activity against Gram-positive (Enterococcus faecium and Staphylococcus aureus) and glutathione-deficient bacteria (Helicobacter pylori). Investigational compounds were tested for antimicrobial activity, anti-biofilm efficacy, target impact, and cytotoxicity. Results The first-generation molecules AU1 and AU5 inhibited TrxR activity and inhibited methicillin-resistant S. aureus strain MW2 with minimal inhibitory concentrations (MIC) of 0.125 and 0.5 μg/mL, respectively. In an S. aureus enzymatic assay, AU1 inhibited TrxR enzymatic activity in a dose-dependent manner causing a decrease in intracellular free thiols. In addition, biofilm studies demonstrated that AU1 and AU5 reduced biofilm formation at 1X MIC and disrupted mature biofilms at 4X MIC. Cytotoxicity profiles were created using human cell lines and primary cells with LD50 exceeding MICs by at least 12X. Discussion Thus, AU1 and AU5 were TrxR inhibitors that yielded low-concentration antimicrobial activity impacting S. aureus in planktonic and biofilm forms with limited toxic liability.
Collapse
Affiliation(s)
- Nagendran Tharmalingam
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, United States
| | - Shi Xu
- Department of Chemistry, Brown University, Providence, RI, United States
| | - Lewis Oscar Felix
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, United States
| | - Biswajit Roy
- Department of Chemistry, Brown University, Providence, RI, United States
| | - Ming Xian
- Department of Chemistry, Brown University, Providence, RI, United States
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, United States
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, United States
| |
Collapse
|
10
|
Maione A, Buonanno A, Galdiero M, de Alteriis E, Petrillo F, Reibaldi M, Guida M, Galdiero E. A Re-Purposing Strategy: Sub-Lethal Concentrations of an Eicosanoid Derived from the Omega-3-Polyunsaturated Fatty Acid Resolvin D1 Affect Dual Species Biofilms. Int J Mol Sci 2023; 24:12876. [PMID: 37629056 PMCID: PMC10454369 DOI: 10.3390/ijms241612876] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The fungal species Candida parapsilosis and the bacterial species Staphylococcus aureus may be responsible for hospital-acquired infections in patients undergoing invasive medical interventions or surgical procedures and often coinfect critically ill patients in complicating polymicrobial biofilms. The efficacy of the re-purposing therapy has recently been reported as an alternative to be used. PUFAs (polyunsaturated fatty acids) may be used alone or in combination with currently available traditional antimicrobials to prevent and manage various infections overcoming antimicrobial resistance. The objectives of the study were to evaluate the effects of Resolvin D1 (RvD1) as an antimicrobial on S. aureus and C. parapsilosis, as well as the activity against the mixed biofilm of the same two species. Microdilution assays and time-kill growth curves revealed bacterial and fungal inhibition at minimum concentration values between 5 and 10 μg mL-1. In single-species structures, an inhibition of 55% and 42% was reported for S. aureus and C. parapsilosis, respectively. Moreover, RvD1 demonstrated an eradication capacity of 60% and 80% for single- and mixed-species biofilms, respectively. In association with the inhibition activity, a downregulation of genes involved in biofilm formation as well as ROS accumulation was observed. Eradication capability was confirmed also on mature mixed biofilm grown on silicone platelets as shown by scanning electron microscopy (SEM). In conclusion, RvD1 was efficient against mono and polymicrobial biofilms in vitro, being a promising alternative for the treatment of mixed bacterial/fungal infections.
Collapse
Affiliation(s)
- Angela Maione
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.B.); (E.d.A.); (M.G.)
| | - Annalisa Buonanno
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.B.); (E.d.A.); (M.G.)
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy;
| | - Elisabetta de Alteriis
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.B.); (E.d.A.); (M.G.)
| | - Francesco Petrillo
- Department of Medical Sciences, Eye Clinic, Turin University, 10126 Turin, Italy; (F.P.); (M.R.)
| | - Michele Reibaldi
- Department of Medical Sciences, Eye Clinic, Turin University, 10126 Turin, Italy; (F.P.); (M.R.)
| | - Marco Guida
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.B.); (E.d.A.); (M.G.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Center for Studies on Bioinspired Agro-Environmental Technology (BAT Center), 80055 Portici, Italy
| | - Emilia Galdiero
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.B.); (E.d.A.); (M.G.)
- Center for Studies on Bioinspired Agro-Environmental Technology (BAT Center), 80055 Portici, Italy
| |
Collapse
|
11
|
Felix L, Whitely C, Tharmalingam N, Mishra B, Vera-Gonzalez N, Mylonakis E, Shukla A, Fuchs BB. Auranofin coated catheters inhibit bacterial and fungal biofilms in a murine subcutaneous model. Front Cell Infect Microbiol 2023; 13:1135942. [PMID: 37313344 PMCID: PMC10258325 DOI: 10.3389/fcimb.2023.1135942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/24/2023] [Indexed: 06/15/2023] Open
Abstract
Microbe entry through catheter ports can lead to biofilm accumulation and complications from catheter-related bloodstream infection and ultimately require antimicrobial treatment and catheter replacement. Although strides have been made with microbial prevention by applying standardized antiseptic techniques during catheter implantation, both bacterial and fungal microbes can present health risks to already sick individuals. To reduce microbial adhesion, murine and human catheters were coated with polyurethane and auranofin using a dip coating method and compared to non-coated materials. Upon passage of fluid through the coated material in vitro, flow dynamics were not impacted. The unique antimicrobial properties of the coating material auranofin has shown inhibitory activity against bacteria such as Staphylococcus aureus and fungi such as Candida albicans. Auranofin coating on catheters at 10mg/mL reduced C. albicans accumulation in vitro from 2.0 x 108 to 7.8 x 105 CFU for mouse catheters and from 1.6 x 107 to 2.8 x 106 for human catheters, showing an impact to mature biofilms. Assessment of a dual microbe biofilm on auranofin-coated catheters resulted in a 2-log reduction in S. aureus and a 3-log reduction in C. albicans compared to uncoated catheters. In vivo assessment in a murine subcutaneous model demonstrated that catheters coated with 10 mg/mL auranofin reduced independent S. aureus and C. albicans accumulation by 4-log and 1-log, respectively, compared to non-coated catheters. In conclusion, the auranofin-coated catheters demonstrate proficiency at inhibiting multiple pathogens by decreasing S. aureus and C. albicans biofilm accumulation.
Collapse
Affiliation(s)
- LewisOscar Felix
- Division of Infectious Diseases, Rhode Island Hospital, The Miriam Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Cutler Whitely
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| | - Nagendran Tharmalingam
- Division of Infectious Diseases, Rhode Island Hospital, The Miriam Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Biswajit Mishra
- Division of Infectious Diseases, Rhode Island Hospital, The Miriam Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Noel Vera-Gonzalez
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, The Miriam Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Anita Shukla
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, The Miriam Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| |
Collapse
|
12
|
Rodrigues ABF, Passos JCDS, Costa MS. Effect of Antimicrobial Photodynamic Therapy, using Toluidine blue on dual-species biofilms of Candida albicans and Candida krusei. Photodiagnosis Photodyn Ther 2023; 42:103600. [PMID: 37150491 DOI: 10.1016/j.pdpdt.2023.103600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/14/2023] [Accepted: 05/05/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Although Candida albicans is the most frequent etiological agent of candidiasis, it has been reported a sizable number of infections related to the non-albicans Candida (NAC) species, Candida krusei. In addition, dual biofilms (biofilms composed by two species) may easily occur in vivo, becoming even more challenging the treatment of an infection. The fungicide effect of Photodynamic Therapy (PDT), using toluidine blue O (TBO) on both C. albicans and C. krusei development has been demonstrated. Thus, the objective of this study was to investigate the effects of PDT on dual-species biofilms of Candida albicans and Candida krusei. METHODS The effect of PDT was observed on the metabolic activity of mature dual-species biofilms of Candida albicans and Candida krusei by a metabolic assay based on the reduction of XTT (2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide sodium salt) assay and the identification of Candida albicans and Candida krusei was performed on CHROMagar Candida medium. RESULTS it was observed a reduction of ∼30% in the metabolic activity of a mature biofilm treated with PDT, using 0.05mg·mL-1 TBO and during biofilm formation a predominance of C. albicans on C. krusei was observed. The inhibition observed was related to reduction in the number of Colony Forming Units (CFU) of Candida albicans from 31.33 ± 3.7 to 17.0 ± 1.5. The number of CFU of C. krusei was not significantly modified. CONCLUSIONS These results demonstrated the efficiency of PDT in inhibiting the dual-species biofilms of Candida albicans and Candida krusei by reducing C. albicans development.
Collapse
Affiliation(s)
- Ana Beatriz Furtado Rodrigues
- Instituto de Pesquisa & Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP. Av. Shishima Hifumi 2911, CEP: 12, 244-000, São José dos Campos, Brazil
| | - Juliene Cristina da Silva Passos
- Instituto de Pesquisa & Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP. Av. Shishima Hifumi 2911, CEP: 12, 244-000, São José dos Campos, Brazil
| | - Maricilia Silva Costa
- Instituto de Pesquisa & Desenvolvimento - IP&D, Universidade do Vale do Paraíba - UNIVAP. Av. Shishima Hifumi 2911, CEP: 12, 244-000, São José dos Campos, Brazil.
| |
Collapse
|
13
|
De Bels D, Maillart E, Van Bambeke F, Redant S, Honoré PM. Existing and emerging therapies for the treatment of invasive candidiasis and candidemia. Expert Opin Emerg Drugs 2022; 27:405-416. [PMID: 36317695 DOI: 10.1080/14728214.2022.2142207] [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: 08/03/2021] [Accepted: 10/27/2022] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Invasive candidiasis or candidemia is a severe infection affecting more than 250,000 people worldwide every year. It is present in up to 16% of ICU patients. The prognosis of these infections is unfavorable, with global death estimated around 50,000 per year, which corresponds to up to 40% depending on patient severity and comorbidities. Therapeutic failure is not rare due to the emergence of multiresistant strains and of new species poorly responsive to current therapies like Candida auris. AREAS COVERED We first review the positioning of antifungal drugs used to treat candidiasis, namely polyenes, azoles, echinocandins and pyrimidine analogues. We then discuss the progresses brought by new formulations, new derivatives within these classes, compounds acting on new targets or repurposed drugs in terms of pharmacokinetic profile, spectrum of activity, potency, safety or risk of drug-drug interactions. EXPERT OPINION While new formulations (amphotericin B cochleate) improve oral bioavailability of the corresponding drugs, new azoles or echinocandins offer higher potency including against strains resistant to former generations of drugs. Repurposed drugs show synergism with current therapies in vitro. Results from ongoing and future clinical trials will be decisive to establish the interest for these drugs in our arsenal.
Collapse
Affiliation(s)
- David De Bels
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
| | - Evelyne Maillart
- Department of Infectious Disease, Brugmann University Hospital, Brussels, Belgium
| | - Françoise Van Bambeke
- Louvain Drug Research Institute, Department of Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Sebastien Redant
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
| | - Patrick M Honoré
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Department of ICU, CHU UCL Godinne-Namur, UCL Louvain Medical School
| |
Collapse
|
14
|
Jampilek J. Novel avenues for identification of new antifungal drugs and current challenges. Expert Opin Drug Discov 2022; 17:949-968. [PMID: 35787715 DOI: 10.1080/17460441.2022.2097659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Some of otherwise useful fungi are pathogenic to humans, and unfortunately, the number of these pathogens is increasing. In addition to common skin infections, these opportunistic pathogens are able to cause severe, often incurable, systemic mycoses. AREAS COVERED : The number of antifungal drugs is limited, especially drugs that can be used for systemic administration, and resistance to these drugs is very common. This review summarizes various approaches to the discovery and development of new antifungal drugs, provides an overview of the most important molecules in terms of basic (laboratory) research and compounds currently in clinical trials, and focuses on drug repurposing strategy, while providing an overview of drugs of other indications that have been tested in vitro for their antifungal activity for possible expansion of antifungal drugs and/or support of existing antimycotics. EXPERT OPINION : Despite the limitations of the research of new antifungal drugs by pharmaceutical manufacturers, in addition to innovated molecules based on clinically used drugs, several completely new small entities with unique mechanisms of actions have been identified. The identification of new molecular targets that offer alternatives for the development of new unique selective antifungal highly effective agents has been an important outcome of repurposing of non-antifungal drugs to antifungal drug. Also, given the advances in monoclonal antibodies and their application to immunosuppressed patients, it may seem possible to predict a more optimistic future for antifungal therapy than has been the case in recent decades.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia.,Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| |
Collapse
|
15
|
Pohl CH. Recent Advances and Opportunities in the Study of Candida albicans Polymicrobial Biofilms. Front Cell Infect Microbiol 2022; 12:836379. [PMID: 35252039 PMCID: PMC8894716 DOI: 10.3389/fcimb.2022.836379] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 01/11/2023] Open
Abstract
It is well known that the opportunistic pathogenic yeast, Candida albicans, can form polymicrobial biofilms with a variety of bacteria, both in vitro and in vivo, and that these polymicrobial biofilms can impact the course and management of disease. Although specific interactions are often described as either synergistic or antagonistic, this may be an oversimplification. Polymicrobial biofilms are complex two-way interacting communities, regulated by inter-domain (inter-kingdom) signaling and various molecular mechanisms. This review article will highlight advances over the last six years (2016-2021) regarding the unique biology of polymicrobial biofilms formed by C. albicans and bacteria, including regulation of their formation. In addition, some of the consequences of these interactions, such as the influence of co-existence on antimicrobial susceptibility and virulence, will be discussed. Since the aim of this knowledge is to inform possible alternative treatment options, recent studies on the discovery of novel anti-biofilm compounds will also be included. Throughout, an attempt will be made to identify ongoing challenges in this area.
Collapse
|
16
|
Liu Y, Lu Y, Xu Z, Ma X, Chen X, Liu W. Repurposing of the gold drug auranofin and a review of its derivatives as antibacterial therapeutics. Drug Discov Today 2022; 27:1961-1973. [DOI: 10.1016/j.drudis.2022.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/22/2022] [Accepted: 02/16/2022] [Indexed: 12/18/2022]
|
17
|
Fatahi Dehpahni M, Chehri K, Azadbakht M. Effect of Silver Nanoparticles and L-Carnitine Supplement on Mixed Vaginitis Caused by Candida albicans/ Staphylococcus aureus in Mouse Models: An Experimental Study. Curr Microbiol 2021; 78:3945-3956. [PMID: 34542662 DOI: 10.1007/s00284-021-02652-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 09/02/2021] [Indexed: 12/01/2022]
Abstract
The evolution of antimicrobial-resistant pathogens is a global health and development threat. Nanomedicine is rapidly becoming the main driving force behind ongoing changes in antimicrobial studies. Among nanoparticles, silver (AgNPs) have attracted attention due to their versatile properties. The study aimed to investigate the effects of AgNPs and L-carnitine (LC) on mixed Candida albicans and Staphylococcus aureus in the mice vaginitis model. Study of antimicrobial activity of AgNPs evaluated by Minimum Inhibitory Concentration (MIC) and Minimum Biocidal Concentration (MBC) assays. AgNPs inhibited biofilm formation of microbial strains, which was tested by using crystal violet staining. In the current study, we evaluated the effects of AgNPs and LC in NMRI mice infected intravaginally with C. albicans/ S. aureus for two weeks. The proportion of mice in each stage of the estrous cycle (proestrus, estrus, metestrus, and diestrus) was examined. Histological properties were assessed by hematoxylin/ eosin (H&E) staining of formalin-fixed, paraffin-embedded vaginal tissue sections. Based on the results, MICs of AgNPs against S. aureus, C. albicans, and their combination were 252.3, 124.8, and 501.8 ppm, and their minimum biofilm inhibitory concentration (MBIC) was 500, 250, and 1000 ppm, respectively. The estrous cycle in the treated group was similar to the control. Vaginal histology and cytology showed that LC can improve tissue damages caused by vaginitis and AgNPs. This study demonstrates the promising use of AgNPs as antimicrobial agents and the combination of AgNPs/ LC could be a great future alternative in the control of vaginitis.
Collapse
Affiliation(s)
| | - Khosrow Chehri
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran.
| | - Mehri Azadbakht
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
18
|
Draft genome sequencing and functional annotation and characterization of biofilm-producing bacterium Bacillus novalis PD1 isolated from rhizospheric soil. Antonie van Leeuwenhoek 2021; 114:1977-1989. [PMID: 34537868 DOI: 10.1007/s10482-021-01655-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/06/2021] [Indexed: 10/20/2022]
Abstract
Biofilm forming bacterium Bacillus novalis PD1 was isolated from the rhizospheric soil of a paddy field. B. novalis PD1 is a Gram-positive, facultatively anaerobic, motile, slightly curved, round-ended, and spore-forming bacteria. The isolate B. novalis PD1 shares 98.45% similarity with B. novalis KB27B. B. vireti LMG21834 and B. drentensis NBRC 102,427 are the closest phylogenetic neighbours for B. novalis PD1. The draft genome RAST annotation showed a linear chromosome with 4,569,088 bp, encoding 6139 coding sequences, 70 transfer RNA (tRNA), and 11 ribosomal RNA (rRNA) genes. The genomic annotation of biofilm forming B. novalis PD1(> 3.6@OD595nm) showed the presence of exopolysaccharide-forming genes (ALG, PSL, and PEL) as well as other biofilm-related genes (comER, Spo0A, codY, sinR, TasA, sipW, degS, and degU). Antibiotic inactivation gene clusters (ANT (6)-I, APH (3')-I, CatA15/A16 family), efflux pumps conferring antibiotic resistance genes (BceA, BceB, MdtABC-OMF, MdtABC-TolC, and MexCD-OprJ), and secondary metabolites linked to phenazine, terpene, and beta lactone gene clusters are part of the genome.
Collapse
|
19
|
Bonvicini F, Belluti F, Bisi A, Gobbi S, Manet I, Gentilomi GA. Improved eradication efficacy of a combination of newly identified antimicrobial agents in C. albicans and S. aureus mixed-species biofilm. Res Microbiol 2021; 172:103873. [PMID: 34389439 DOI: 10.1016/j.resmic.2021.103873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Candida albicans and Staphylococcus aureus are common human pathogens, frequently isolated independently or co-isolated from bloodstream infections, and able to form dense polymicrobial biofilms on various medical devices resulting in strong resistance to conventionally used antimicrobials. New and innovative approaches are therefore needed to ensure the successful management of biofilm related infections. In this study, a chalcone-based derivative and a polycyclic anthracene-maleimide adduct, previously ascertained by us as inhibitors of C. albicans and S. aureus growths, respectively, were reconsidered in a new perspective by evaluating the efficacy of a combined treatment against a polymicrobial biofilm. Both quantitative and qualitative analyses were carried out to delve into their inhibitory potential on the polymicrobial population. Our results indicate that these newly identified antimicrobials are effective in reducing the biomass of the mixed C. albicans-S. aureus biofilm and the viability of fungal-bacterial cells within the polymicrobial community; in addition, confocal laser scanning microscopy demonstrate that compounds 1 and 2 treatment thoroughly modifies the architecture of the dual-species biofilm.
Collapse
Affiliation(s)
- Francesca Bonvicini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy.
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy.
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy.
| | - Ilse Manet
- Institute for Organic and Photoreactivity (ISOF), National Research Council (CNR), Via Gobetti 101, 40129, Bologna, Italy.
| | - Giovanna Angela Gentilomi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| |
Collapse
|
20
|
Drug Repurposing in Medical Mycology: Identification of Compounds as Potential Antifungals to Overcome the Emergence of Multidrug-Resistant Fungi. Pharmaceuticals (Basel) 2021; 14:ph14050488. [PMID: 34065420 PMCID: PMC8161392 DOI: 10.3390/ph14050488] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
Immunodepression, whether due to HIV infection or organ transplantation, has increased human vulnerability to fungal infections. These conditions have created an optimal environment for the emergence of opportunistic infections, which is concomitant to the increase in antifungal resistance. The use of conventional antifungal drugs as azoles and polyenes can lead to clinical failure, particularly in immunocompromised individuals. Difficulties related to treating fungal infections combined with the time required to develop new drugs, require urgent consideration of other therapeutic alternatives. Drug repurposing is one of the most promising and rapid solutions that the scientific and medical community can turn to, with low costs and safety advantages. To treat life-threatening resistant fungal infections, drug repurposing has led to the consideration of well-known and potential molecules as a last-line therapy. The aim of this review is to provide a summary of current antifungal compounds and their main resistance mechanisms, following by an overview of the antifungal activity of non-traditional antimicrobial drugs. We provide their eventual mechanisms of action and the synergistic combinations that improve the activity of current antifungal treatments. Finally, we discuss drug repurposing for the main emerging multidrug resistant (MDR) fungus, including the Candida auris, Aspergillus or Cryptococcus species.
Collapse
|
21
|
Khan F, Bamunuarachchi NI, Pham DTN, Tabassum N, Khan MSA, Kim YM. Mixed biofilms of pathogenic Candida-bacteria: regulation mechanisms and treatment strategies. Crit Rev Microbiol 2021; 47:699-727. [PMID: 34003065 DOI: 10.1080/1040841x.2021.1921696] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mixed-species biofilm is one of the most frequently recorded clinical problems. Mixed biofilms develop as a result of interactions between microorganisms of a single or multiple species (e.g. bacteria and fungi). Candida spp., particularly Candida albicans, are known to associate with various bacterial species to form a multi-species biofilm. Mixed biofilms of Candida spp. have been previously detected in vivo and on the surfaces of many biomedical instruments. Treating infectious diseases caused by mixed biofilms of Candida and bacterial species has been challenging due to their increased resistance to antimicrobial drugs. Here, we review and discuss the clinical significance of mixed Candida-bacteria biofilms as well as the signalling mechanisms involved in Candida-bacteria interactions. We also describe possible approaches for combating infections associated with mixed biofilms, such as the use of natural or synthetic drugs and combination therapy. The review presented here is expected to contribute to the advances in the biomedical field on the understanding of underlying interaction mechanisms of pathogens in mixed biofilm, and alternative approaches to treating the related infections.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea
| | - Nilushi Indika Bamunuarachchi
- Department of Food Science and Technology, Pukyong National University, Busan, South Korea.,Department of Fisheries and Marine Sciences, Ocean University of Sri Lanka, Tangalle, Sri Lanka
| | - Dung Thuy Nguyen Pham
- Center of Excellence for Biochemistry and Natural Products, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam.,NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, South Korea
| | - Mohd Sajjad Ahmad Khan
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea.,Department of Food Science and Technology, Pukyong National University, Busan, South Korea
| |
Collapse
|
22
|
Yaakoub H, Staerck C, Mina S, Godon C, Fleury M, Bouchara JP, Calenda A. Repurposing of auranofin and honokiol as antifungals against Scedosporium species and the related fungus Lomentospora prolificans. Virulence 2021; 12:1076-1090. [PMID: 33825667 PMCID: PMC8032236 DOI: 10.1080/21505594.2021.1909266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The slowing-down de novo drug-discovery emphasized the importance of repurposing old drugs. This is particularly true when combating infections caused by therapy-refractory microorganisms, such as Scedosporium species and Lomentospora prolificans. Recent studies on Scedosporium responses to oxidative stress underscored the importance of targeting the underlying mechanisms. Auranofin, ebselen, PX-12, honokiol, and to a lesser extent, conoidin A are known to disturb redox-homeostasis systems in many organisms. Their antifungal activity was assessed against 27 isolates belonging to the major Scedosporium species: S. apiospermum, S. aurantiacum, S. boydii, S. dehoogii, S. minutisporum, and Lomentospora prolificans. Auranofin and honokiol were the most active against all Scedosporium species (mean MIC50 values of 2.875 and 6.143 μg/ml, respectively) and against L. prolificans isolates (mean MIC50 values of 4.0 and 3.563μg/ml respectively). Combinations of auranofin with voriconazole or honokiol revealed additive effects against 9/27 and 18/27 isolates, respectively. Synergistic interaction between auranofin and honokiol was only found against one isolate of L. prolificans. The effects of auranofin upon exposure to oxidative stress were also investigated. For all species except S. dehoogii, the maximal growth in the presence of auranofin significantly decreased when adding a sublethal dose of menadione. The analysis of the expression of genes encoding oxidoreductase enzymes upon exposure of S. apiospermum to honokiol unveiled the upregulation of many genes, especially those coding peroxiredoxins, thioredoxin reductases, and glutaredoxins. Altogether, these data suggest that auranofin and honokiol act via dampening the redox balance and support their repurposing as antifungals against Scedosporium species and L. prolificans.
Collapse
Affiliation(s)
- Hajar Yaakoub
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Cindy Staerck
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Sara Mina
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Charlotte Godon
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Maxime Fleury
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Jean-Philippe Bouchara
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France.,Département de biologie des agents infectieux , Laboratoire De Parasitologie-Mycologie, Centre Hospitalier Universitaire, Angers, France
| | - Alphonse Calenda
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| |
Collapse
|
23
|
Zhu J, She P, Fu J, Peng C, Wu Y. Identification of Eltrombopag as a Repurposing Drug Against Staphylococcus epidermidis and its Biofilms. Curr Microbiol 2021; 78:1159-1167. [PMID: 33611618 DOI: 10.1007/s00284-021-02386-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/07/2021] [Indexed: 10/22/2022]
Abstract
Staphylococcus epidermidis is a common cause of nosocomial infections, and readily adheres to medical apparatus to form biofilms consisting of highly resistant persister cells. Owing to the refractory infections caused by S. epidermidis biofilms and persisters in immunosuppressed patients, it is crucial to develop new antimicrobials. In the present study, we analyzed the antimicrobial effects of the thrombopoietin receptor agonist eltrombopag (EP) against S. epidermidis planktonic cells, biofilms, and persister cells. EP was significantly toxic to S. epidermidis with the minimal inhibitory concentration of 8 μg/ml, and effectively inhibited the biofilms and persisters in a strain-dependent manner. In addition, EP was only mildly toxic to mammalian cells after 12 to 24 h treatment. It also partially synergized with vancomycin against S. epidermidis, which enhanced its antimicrobial effects and reduced its toxicity to mammalian cells. Taken together, EP is a potential antibiotic for treating refractory infections caused by S. epidermidis.
Collapse
Affiliation(s)
- Juan Zhu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China.
| | - Juhua Fu
- Department of Human Resources, The Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - Canhui Peng
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
24
|
Scorzoni L, Fuchs BB, Junqueira JC, Mylonakis E. Current and promising pharmacotherapeutic options for candidiasis. Expert Opin Pharmacother 2021; 22:867-887. [PMID: 33538201 DOI: 10.1080/14656566.2021.1873951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Candida spp. are commensal yeasts capable of causing infections such as superficial, oral, vaginal, or systemic infections. Despite medical advances, the antifungal pharmacopeia remains limited and the development of alternative strategies is needed.Areas covered: We discuss available treatments for Candida spp. infections, highlighting advantages and limitations related to pharmacokinetics, cytotoxicity, and antimicrobial resistance. Moreover, we present new perspectives to improve the activity of the available antifungals, discussing their immunomodulatory potential and advances on drug delivery carriers. New therapeutic approaches are presented including recent synthesized antifungal compounds (Enchochleated-Amphotericin B, tetrazoles, rezafungin, enfumafungin, manogepix and arylamidine); drug repurposing using a diversity of antibacterial, antiviral and non-antimicrobial drugs; combination therapies with different compounds or photodynamic therapy; and innovations based on nano-particulate delivery systems.Expert opinion: With the lack of novel drugs, the available assets must be leveraged to their best advantage through modifications that enhance delivery, efficacy, and solubility. However, these efforts are met with continuous challenges presented by microbes in their infinite plight to resist and survive therapeutic drugs. The pharmacotherapeutic options in development need to focus on new antimicrobial targets. The success of each antimicrobial agent brings strategic insights to the next phased approach in treatingCandida spp. infections.
Collapse
Affiliation(s)
- Liliana Scorzoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, SP Brazil
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School, Brown University, Providence, RI USA
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, SP Brazil
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School, Brown University, Providence, RI USA
| |
Collapse
|
25
|
Kim JH, Cheng LW, Chan KL, Tam CC, Mahoney N, Friedman M, Shilman MM, Land KM. Antifungal Drug Repurposing. Antibiotics (Basel) 2020; 9:antibiotics9110812. [PMID: 33203147 PMCID: PMC7697925 DOI: 10.3390/antibiotics9110812] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
Control of fungal pathogens is increasingly problematic due to the limited number of effective drugs available for antifungal therapy. Conventional antifungal drugs could also trigger human cytotoxicity associated with the kidneys and liver, including the generation of reactive oxygen species. Moreover, increased incidences of fungal resistance to the classes of azoles, such as fluconazole, itraconazole, voriconazole, or posaconazole, or echinocandins, including caspofungin, anidulafungin, or micafungin, have been documented. Of note, certain azole fungicides such as propiconazole or tebuconazole that are applied to agricultural fields have the same mechanism of antifungal action as clinical azole drugs. Such long-term application of azole fungicides to crop fields provides environmental selection pressure for the emergence of pan-azole-resistant fungal strains such as Aspergillus fumigatus having TR34/L98H mutations, specifically, a 34 bp insertion into the cytochrome P450 51A (CYP51A) gene promoter region and a leucine-to-histidine substitution at codon 98 of CYP51A. Altogether, the emerging resistance of pathogens to currently available antifungal drugs and insufficiency in the discovery of new therapeutics engender the urgent need for the development of new antifungals and/or alternative therapies for effective control of fungal pathogens. We discuss the current needs for the discovery of new clinical antifungal drugs and the recent drug repurposing endeavors as alternative methods for fungal pathogen control.
Collapse
Affiliation(s)
- Jong H. Kim
- Foodborne Toxin Detection and Prevention Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA; (L.W.C.); (K.L.C.); (C.C.T.); (N.M.)
- Correspondence: ; Tel.: +1-510-559-5841
| | - Luisa W. Cheng
- Foodborne Toxin Detection and Prevention Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA; (L.W.C.); (K.L.C.); (C.C.T.); (N.M.)
| | - Kathleen L. Chan
- Foodborne Toxin Detection and Prevention Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA; (L.W.C.); (K.L.C.); (C.C.T.); (N.M.)
| | - Christina C. Tam
- Foodborne Toxin Detection and Prevention Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA; (L.W.C.); (K.L.C.); (C.C.T.); (N.M.)
| | - Noreen Mahoney
- Foodborne Toxin Detection and Prevention Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA; (L.W.C.); (K.L.C.); (C.C.T.); (N.M.)
| | - Mendel Friedman
- Healthy Processed Foods Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA;
| | | | - Kirkwood M. Land
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
26
|
Qian W, Zhang J, Wang W, Liu M, Fu Y, Li X, Wang T, Li Y. Efficacy of Chelerythrine Against Mono- and Dual-Species Biofilms of Candida albicans and Staphylococcus aureus and Its Properties of Inducing Hypha-to-Yeast Transition of C. albicans. J Fungi (Basel) 2020; 6:jof6020045. [PMID: 32252437 PMCID: PMC7345410 DOI: 10.3390/jof6020045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans and Staphylococcus aureus specifically often resulted in biofilm-associated diseases, ranging from superficial mucosal to life-threatening systemic infections. Recent studies reported that chelerythrine displayed antimicrobial activities against a few microorganisms, but its effects on mono- and dual-species biofilms of C. albicans and S. aureus have never been reported. The purpose of this study was to evaluate the efficacy of chelerythrine against mono- and dual-species biofilms, and explore its effect on the hyphal growth and the hypha-to-yeast transition of C. albicans. The results showed that minimum inhibitory concentrations (MICs) and minimum biofilm inhibitory concentration (MBIC90S) of chelerythrine against planktonic cells of mono-species were 4 and 2 μg/mL, while the MIC and MBIC90 were 6 and 3 μg/mL for dual-species. Meanwhile, the decrease in three matrix component levels and tolerance to antibiotics of biofilms formed by mono- and dual-species exposed to chelerythrine were confirmed by a confocal laser scanning microscope, in conjugation with five fluorescent dyes and a gatifloxacin diffusion assay. Moreover, C. albicans and S. aureus mono-species showed a 96.4, and 92.3% reduction, respectively, in 24-h preformed biofilm biomass in the presence of 128 µg/mL of chelerythrine. Similarly, preformed (24 h) dual-species biofilm biomass also displayed a significant reduction (90.7%) when treated with 192 μg/mL chelerythrine. Chelerythrine inhibited hyphae formation of C. albicans at 4 μg/mL, and C. albicans in hypha-form can be converted into yeast-form at 8 μg/mL of chelerythrine. Therefore, chelerythrine shows promise as a potential antimicrobial and antibiofilm agent for clinical effective treatments of mono- and mixed-species and/or biofilm-associated infections.
Collapse
Affiliation(s)
- Weidong Qian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
| | - Jianing Zhang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
| | - Wenjing Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
| | - Miao Liu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
| | - Yuting Fu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
| | - Xiang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
| | - Ting Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (J.Z.); (W.W.); (M.L.); (Y.F.); (X.L.)
- Correspondence: (T.W.); (Y.L.); Tel.: +86-29-86168583 (T.W.)
| | - Yongdong Li
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo 315010, China
- Correspondence: (T.W.); (Y.L.); Tel.: +86-29-86168583 (T.W.)
| |
Collapse
|
27
|
Qian W, Wang W, Zhang J, Liu M, Fu Y, Li X, Wang T, Li Y. Sanguinarine Inhibits Mono- and Dual-Species Biofilm Formation by Candida albicans and Staphylococcus aureus and Induces Mature Hypha Transition of C. albicans. Pharmaceuticals (Basel) 2020; 13:ph13010013. [PMID: 31941090 PMCID: PMC7168937 DOI: 10.3390/ph13010013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 02/07/2023] Open
Abstract
Previous studies have reported that sanguinarine possesses inhibitory activities against several microorganisms, but its effects on mono- and dual-species biofilms of C. albicans and S. aureus have not been fully elucidated. In this study, we aimed to evaluate the efficacy of sanguinarine for mono- and dual-species biofilms and explore its ability to induce the hypha-to-yeast transition of C. albicans. The results showed that the minimum inhibitory concentration (MIC) and minimum biofilm inhibitory concentration (MBIC90) of sanguinarine against C. albicans and S. aureus mono-species biofilms was 4, and 2 μg/mL, respectively, while the MIC and MBIC90 of sanguinarine against dual-species biofilms was 8, and 4 μg/mL, respectively. The decrease in the levels of matrix component and tolerance to antibiotics of sanguinarine-treated mono- and dual-species biofilms was revealed by confocal laser scanning microscopy combined with fluorescent dyes, and the gatifloxacin diffusion assay, respectively. Meanwhile, sanguinarine at 128 and 256 μg/mL could efficiently eradicate the preformed 24-h biofilms by mono- and dual-species, respectively. Moreover, sanguinarine at 8 μg/mL could result in the transition of C. albicans from the mature hypha form to the unicellular yeast form. Hence, this study provides useful information for the development of new agents to combat mono- and dual-species biofilm-associated infections, caused by C. albicans and S. aureus.
Collapse
Affiliation(s)
- Weidong Qian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
| | - Wenjing Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
| | - Jianing Zhang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
| | - Miao Liu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
| | - Yuting Fu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
| | - Xiang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
| | - Ting Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (W.Q.); (W.W.); (J.Z.); (M.L.); (Y.F.); (X.L.)
- Correspondence: (T.W.); (Y.L.); Tel.: +86-29-86168583 (T.W.)
| | - Yongdong Li
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo 315010, China
- Correspondence: (T.W.); (Y.L.); Tel.: +86-29-86168583 (T.W.)
| |
Collapse
|