51
|
Possamai Rossatto FC, Tharmalingam N, Escobar IE, d’Azevedo PA, Zimmer KR, Mylonakis E. Antifungal Activity of the Phenolic Compounds Ellagic Acid (EA) and Caffeic Acid Phenethyl Ester (CAPE) against Drug-Resistant Candida auris. J Fungi (Basel) 2021; 7:jof7090763. [PMID: 34575801 PMCID: PMC8466507 DOI: 10.3390/jof7090763] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022] Open
Abstract
Candida auris is an emerging healthcare-associated fungal pathogen that has become a serious global health threat. Current treatment options are limited due to drug resistance. New therapeutic strategies are required to target this organism and its pathogenicity. Plant polyphenols are structurally diverse compounds that present a vast range of biological properties. In the present study, plant-derived molecules ellagic acid (EA) and caffeic acid phenethyl ester (CAPE) were investigated for their antifungal and antivirulence activities against Candida auris. We also tested against C. albicans. The minimum inhibitory concentration (MIC) for EA ranged from 0.125 to 0.25 µg/mL and for CAPE ranged from 1 to 64 µg/mL against drug-resistant C. auris strains. Killing kinetics determined that after 4 h treatment with CAPE, there was a complete reduction of viable C. auris cells compared to fluconazole. Both compounds might act by modifying the fungal cell wall. CAPE significantly reduced the biomass and the metabolic activity of C. auris biofilm and impaired C. auris adhesion to cultured human epithelial cells. Furthermore, both compounds prolonged the survival rate of Galleria mellonella infected by C. auris (p = 0.0088 for EA at 32 mg/kg and p = 0.0028 for CAPE at 4 mg/kg). In addition, EA at 4 μg/mL prolonged the survival of C. albicans-infected Caenorhabditis elegans (p < 0.0001). CAPE was not able to prolong the survival of C. albicans-infected C. elegans. These findings highlight the antifungal and antivirulence effects of EA and CAPE against C. auris, and warrant further investigation as novel antifungal agents against drug-resistant infections.
Collapse
Affiliation(s)
- Fernanda Cristina Possamai Rossatto
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Nagendran Tharmalingam
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
| | - Iliana E. Escobar
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
| | - Pedro Alves d’Azevedo
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Karine Rigon Zimmer
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
- Correspondence: ; Tel.: +1-401-444-7845; Fax: +1-401-444-8179
| |
Collapse
|
52
|
Fungal Biofilms as a Valuable Target for the Discovery of Natural Products That Cope with the Resistance of Medically Important Fungi-Latest Findings. Antibiotics (Basel) 2021; 10:antibiotics10091053. [PMID: 34572635 PMCID: PMC8471798 DOI: 10.3390/antibiotics10091053] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/18/2022] Open
Abstract
The development of new antifungal agents that target biofilms is an urgent need. Natural products, mainly from the plant kingdom, represent an invaluable source of these entities. The present review provides an update (2017-May 2021) on the available information on essential oils, propolis, extracts from plants, algae, lichens and microorganisms, compounds from different natural sources and nanosystems containing natural products with the capacity to in vitro or in vivo modulate fungal biofilms. The search yielded 42 articles; seven involved essential oils, two Brazilian propolis, six plant extracts and one of each, extracts from lichens and algae/cyanobacteria. Twenty articles deal with the antibiofilm effect of pure natural compounds, with 10 of them including studies of the mechanism of action and five dealing with natural compounds included in nanosystems. Thirty-seven manuscripts evaluated Candida spp. biofilms and two tested Fusarium and Cryptococcus spp. Only one manuscript involved Aspergillus fumigatus. From the data presented here, it is clear that the search of natural products with activity against fungal biofilms has been a highly active area of research in recent years. However, it also reveals the necessity of deepening the studies by (i) evaluating the effect of natural products on biofilms formed by the newly emerged and worrisome health-care associated fungi, C. auris, as well as on other non-albicans Candida spp., Cryptococcus sp. and filamentous fungi; (ii) elucidating the mechanisms of action of the most active natural products; (iii) increasing the in vivo testing.
Collapse
|
53
|
Hassan Y, Chew SY, Than LTL. Candida glabrata: Pathogenicity and Resistance Mechanisms for Adaptation and Survival. J Fungi (Basel) 2021; 7:jof7080667. [PMID: 34436206 PMCID: PMC8398317 DOI: 10.3390/jof7080667] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Candida glabrata is a yeast of increasing medical relevance, particularly in critically ill patients. It is the second most isolated Candida species associated with invasive candidiasis (IC) behind C. albicans. The attributed higher incidence is primarily due to an increase in the acquired immunodeficiency syndrome (AIDS) population, cancer, and diabetic patients. The elderly population and the frequent use of indwelling medical devices are also predisposing factors. This work aimed to review various virulence factors that facilitate the survival of pathogenic C. glabrata in IC. The available published research articles related to the pathogenicity of C. glabrata were retrieved and reviewed from four credible databases, mainly Google Scholar, ScienceDirect, PubMed, and Scopus. The articles highlighted many virulence factors associated with pathogenicity in C. glabrata, including adherence to susceptible host surfaces, evading host defences, replicative ageing, and producing hydrolytic enzymes (e.g., phospholipases, proteases, and haemolysins). The factors facilitate infection initiation. Other virulent factors include iron regulation and genetic mutations. Accordingly, biofilm production, tolerance to high-stress environments, resistance to neutrophil killings, and development of resistance to antifungal drugs, notably to fluconazole and other azole derivatives, were reported. The review provided evident pathogenic mechanisms and antifungal resistance associated with C. glabrata in ensuring its sustenance and survival.
Collapse
Affiliation(s)
- Yahaya Hassan
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Bayero University Kano, Kano 700241, Nigeria;
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Shu Yih Chew
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
- Correspondence: ; Tel.: +60-39769-2373
| |
Collapse
|
54
|
Costa-Orlandi CB, Martinez LR, Bila NM, Friedman JM, Friedman AJ, Mendes-Giannini MJS, Nosanchuk JD. Nitric Oxide-Releasing Nanoparticles Are Similar to Efinaconazole in Their Capacity to Eradicate Trichophyton rubrum Biofilms. Front Cell Infect Microbiol 2021; 11:684150. [PMID: 34336712 PMCID: PMC8319823 DOI: 10.3389/fcimb.2021.684150] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Filamentous fungi such as Trichophyton rubrum and T. mentagrophytes, the main causative agents of onychomycosis, have been recognized as biofilm-forming microorganisms. Nitric oxide-releasing nanoparticles (NO-np) are currently in development for the management of superficial and deep bacterial and fungal infections, with documented activity against biofilms. In this context, this work aimed to evaluate, for the first time, the in vitro anti-T. rubrum biofilm potential of NO-np using standard ATCC MYA-4438 and clinical BR1A strains and compare it to commonly used antifungal drugs including fluconazole, terbinafine and efinaconazole. The biofilms formed by the standard strain produced more biomass than those from the clinical strain. NO-np, fluconazole, terbinafine, and efinaconazole inhibited the in vitro growth of planktonic T. rubrum cells. Similarly, NO-np reduced the metabolic activities of clinical strain BR1A preformed biofilms at the highest concentration tested (SMIC50 = 40 mg/mL). Scanning electron and confocal microscopy revealed that NO-np and efinaconazole severely damaged established biofilms for both strains, resulting in collapse of hyphal cell walls and reduced the density, extracellular matrix and thickness of the biofilms. These findings suggest that biofilms should be considered when developing and testing new drugs for the treatment of dermatophytosis. Development of a biofilm phenotype by these fungi may explain the resistance of dermatophytes to some antifungals and why prolonged treatment is usually required for onychomycosis.
Collapse
Affiliation(s)
- Caroline Barcelos Costa-Orlandi
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY, United States.,Deparment of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Brazil
| | - Luis R Martinez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
| | - Níura Madalena Bila
- Deparment of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Brazil.,Department of Para-Clinic, School of Veterinary, Universidade Eduardo Mondlane (UEM), Maputo, Mozambique
| | - Joel M Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Adam J Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Dermatology, George Washington School of Medicine and Health Sciences, Washington, DC, United States.,Department of Medicine, Division of Dermatology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria José S Mendes-Giannini
- Deparment of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Brazil
| | - Joshua D Nosanchuk
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
55
|
Ottaviano E, Baron G, Fumagalli L, Leite J, Colombo EA, Artasensi A, Aldini G, Borghi E. Candida albicans Biofilm Inhibition by Two Vaccinium macrocarpon (Cranberry) Urinary Metabolites: 5-(3',4'-DihydroxyPhenyl)-γ-Valerolactone and 4-Hydroxybenzoic Acid. Microorganisms 2021; 9:microorganisms9071492. [PMID: 34361928 PMCID: PMC8307188 DOI: 10.3390/microorganisms9071492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/22/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022] Open
Abstract
Candida spp. are pathobionts, as they can switch from commensals to pathogens, responsible for a variety of pathological processes. Adhesion to surfaces, morphological switch and biofilm-forming ability are the recognized virulence factors promoting yeast virulence. Sessile lifestyle also favors fungal persistence and antifungal tolerance. In this study, we investigated, in vitro, the efficacy of two urinary cranberry metabolites, 5-(3′,4′-dihydroxy phenyl)-γ-valerolactone (VAL) and 4-hydroxybenzoic acid (4-HBA), in inhibiting C. albicans adhesion and biofilm formation. Both the reference strain SC5314 and clinical isolates were used. We evaluated biomass reduction, by confocal microscopy and crystal violet assay, and the possible mechanisms mediating their inhibitory effects. Both VAL and 4-HBA were able to interfere with the yeast adhesion, by modulating the expression of key genes, HWP1 and ALS3. A significant dose-dependent reduction in biofilm biomass and metabolic activity was also recorded. Our data showed that the two cranberry metabolites VAL and 4-HBA could pave the way for drug development, for targeting the very early phases of biofilm formation and for preventing genitourinary Candida infections.
Collapse
Affiliation(s)
- Emerenziana Ottaviano
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (E.O.); (E.A.C.)
| | - Giovanna Baron
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Laura Fumagalli
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Jessica Leite
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Elisa Adele Colombo
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (E.O.); (E.A.C.)
| | - Angelica Artasensi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (L.F.); (J.L.); (A.A.); (G.A.)
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (E.O.); (E.A.C.)
- Correspondence: ; Tel.: +39-02-50323287
| |
Collapse
|
56
|
Sun PP, Won J, Choo-Kang G, Li S, Chen W, Monroy GL, Chaney EJ, Boppart SA, Eden JG, Nguyen TH. Inactivation and sensitization of Pseudomonas aeruginosa by microplasma jet array for treating otitis media. NPJ Biofilms Microbiomes 2021; 7:48. [PMID: 34078901 PMCID: PMC8172902 DOI: 10.1038/s41522-021-00219-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/28/2021] [Indexed: 02/04/2023] Open
Abstract
Otitis media (OM), known as a middle ear infection, is the leading cause of antibiotic prescriptions for children. With wide-spread use of antibiotics in OM, resistance to antibiotics continues to decrease the efficacy of the treatment. Furthermore, as the presence of a middle ear biofilm has contributed to this reduced susceptibility to antimicrobials, effective interventions are necessary. A miniaturized 3D-printed microplasma jet array has been developed to inactivate Pseudomonas aeruginosa, a common bacterial strain associated with OM. The experiments demonstrate the disruption of planktonic and biofilm P. aeruginosa by long-lived molecular species generated by microplasma, as well as the synergy of combining microplasma treatment with antibiotic therapy. In addition, a middle ear phantom model was developed with an excised rat eardrum to investigate the antimicrobial effects of microplasma on bacteria located behind the eardrum, as in a patient-relevant setup. These results suggest the potential for microplasma as a new treatment paradigm for OM.
Collapse
Affiliation(s)
- Peter P Sun
- Department of Civil and Environmental Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- N. Holonyak, Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jungeun Won
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Gabrielle Choo-Kang
- Department of Civil and Environmental Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Shouyan Li
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Wenyuan Chen
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Guillermo L Monroy
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Eric J Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Stephen A Boppart
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Champaign, IL, USA.
| | - J Gary Eden
- N. Holonyak, Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Thanh H Nguyen
- Department of Civil and Environmental Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Champaign, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
57
|
Cortés I, Cordisco E, Kaufman TS, Sortino MA, Svetaz LA, Bracca ABJ. First total synthesis of chromanone A, preparation of related compounds and evaluation of their antifungal activity against Candida albicans, a biofilm forming agent. RSC Adv 2021; 11:19587-19597. [PMID: 35479228 PMCID: PMC9033609 DOI: 10.1039/d1ra02553h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/24/2021] [Indexed: 11/21/2022] Open
Abstract
A straightforward and convenient approach for the first total syntheses of chromanone A and a related 7-OMe substituted natural product is reported. These unique C-3 substituted 2-hydroxymethyl chromones were recently isolated as fungal metabolites. Chromanone A was synthesized in 25.3% overall yield from the readily available pyrocatechol, whereas the second natural product was prepared in 39.7% global yield. A small library of chromones, including both natural products and some of their synthetic heterocyclic precursors, was evaluated against Candida albicans ATCC 10231, a biofilm forming agent. It was found that 8-methoxy-3-methyl-4-oxo-4H-chromene-2-carbaldehyde, a partially oxidized form of chromanone A, exhibited a minimum inhibitory concentration of 7.8 μg mL-1 and significantly inhibited the yeast's virulence factors, including the adherence to buccal epithelial cells and the secretion of phospholipases, as well as the formation of germ tubes and the generation of the hyphal pseudomycelium. In addition, despite the heterocycle exhibiting non-significant inhibition of the formation of the Candida biofilm, it completely inhibited the growth of C. albicans in preformed biofilms at 62.5 μg mL-1.
Collapse
Affiliation(s)
- Iván Cortés
- Instituto de Química Rosario (IQUIR, CONICET-UNR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Suipacha 531 S2002LRK Rosario Argentina
| | - Estefanía Cordisco
- Area Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Suipacha 531 S2002LRK Rosario Argentina
| | - Teodoro S Kaufman
- Instituto de Química Rosario (IQUIR, CONICET-UNR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Suipacha 531 S2002LRK Rosario Argentina
| | - Maximiliano A Sortino
- Area Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Suipacha 531 S2002LRK Rosario Argentina
| | - Laura A Svetaz
- Area Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Suipacha 531 S2002LRK Rosario Argentina
| | - Andrea B J Bracca
- Instituto de Química Rosario (IQUIR, CONICET-UNR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Suipacha 531 S2002LRK Rosario Argentina
| |
Collapse
|
58
|
Al-Ani E, Hill D, Doudin K. Chlorhexidine Mucoadhesive Buccal Tablets: The Impact of Formulation Design on Drug Delivery and Release Kinetics Using Conventional and Novel Dissolution Methods. Pharmaceuticals (Basel) 2021; 14:ph14060493. [PMID: 34070990 PMCID: PMC8224615 DOI: 10.3390/ph14060493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Oropharyngeal candidiasis (OPC) is a mucosal infection caused by Candida spp., and it is common among the immunocompromised. This condition is mainly treated using oral antifungals. Chlorhexidine (CHD) is a fungicidal and is available as a mouth wash and oral gel. It is used as an adjuvant in the treatment of OPC due to the low residence time of the current formulations. In this study, its activity was tested against C. albicans biofilm and biocompatibility with the HEK293 human cell line. Then, it was formulated as mucoadhesive hydrogel buccal tablets to extend its activity. Different ratios of hydroxypropyl methylcellulose (HPMC), poloxamer 407 (P407), and three different types of polyols were used to prepare the tablets, which were then investigated for their physicochemical properties, ex vivo mucoadhesion, drug release profiles, and the kinetics of drug release. The release was performed using Apparatus I and a controlled flow rate (CFR) method. The results show that CHD is biocompatible and effective against Candida biofilm at a concentration of 20 µg/mL. No drug excipient interaction was observed through differential scanning calorimetry (DSC) and Fourier-transform infrared spectroscopy (FTIR). The increase in P407 and polyol ratios showed a decrease in the swelling index and an increase in CHD in vitro release. The release of CHD from the selected formulations was 86-92%. The results suggest that chlorhexidine tablets are a possible candidate for the treatment of oropharyngeal candidiasis.
Collapse
Affiliation(s)
- Enas Al-Ani
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- Correspondence: (E.A.-A.); (D.H.); Tel.: +44-1902-32-5876 (E.A.-A.)
| | - David Hill
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- School of Biology, Chemistry and Forensic Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- Correspondence: (E.A.-A.); (D.H.); Tel.: +44-1902-32-5876 (E.A.-A.)
| | - Khalid Doudin
- Department of Chemistry, The University of Sheffield, Sheffield S10 2TN, UK;
| |
Collapse
|
59
|
Bila NM, Costa-Orlandi CB, Vaso CO, Bonatti JLC, de Assis LR, Regasini LO, Fontana CR, Fusco-Almeida AM, Mendes-Giannini MJS. 2-Hydroxychalcone as a Potent Compound and Photosensitizer Against Dermatophyte Biofilms. Front Cell Infect Microbiol 2021; 11:679470. [PMID: 34055673 PMCID: PMC8155603 DOI: 10.3389/fcimb.2021.679470] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
Dermatophytes, fungi that cause dermatophytosis, can invade keratinized tissues in humans and animals. The biofilm-forming ability of these fungi was described recently, and it may be correlated with the long treatment period and common recurrences of this mycosis. In this study, we evaluated the anti-dermatophytic and anti-biofilm activity of 2-hydroxychalcone (2-chalcone) in the dark and photodynamic therapy (PDT)-mediated and to determine its mechanism of action. Trichophyton rubrum and Trichophyton mentagrophytes strains were used in the study. The antifungal susceptibility test of planktonic cells, early-stage biofilms, and mature biofilms were performed using colorimetric methods. Topographies were visualized by scanning electron microscopy (SEM). Human skin keratinocyte (HaCat) monolayers were also used in the cytotoxicity assays. The mechanisms of action of 2-chalcone in the dark and under photoexcitation were investigated using confocal microscopy and the quantification of ergosterol, reactive oxygen species (ROS), and death induction by apoptosis/necrosis. All strains, in the planktonic form, were inhibited after treatment with 2-chalcone (minimum inhibitory concentration (MIC) = 7.8-15.6 mg/L), terbinafine (TRB) (MIC = 0.008–0.03 mg/L), and fluconazole (FLZ) (1–512 mg/L). Early-stage biofilm and mature biofilms were inhibited by 2-chalcone at concentrations of 15.6 mg/L and 31.2 mg/L in all tested strains. However, mature biofilms were resistant to all the antifungal drugs tested. When planktonic cells and biofilms (early-stage and mature) were treated with 2-chalcone-mediated PDT, the inhibitory concentrations were reduced by four times (2–7.8 mg/L). SEM images of biofilms treated with 2-chalcone showed cell wall collapse, resulting from a probable extravasation of cytoplasmic content. The toxicity of 2-chalcone in HaCat cells showed higher IC50 values in the dark than under photoexcitation. Further, 2-chalcone targets ergosterol in the cell and promotes the generation of ROS, resulting in cell death by apoptosis and necrosis. Overall, 2-chalcone-mediated PDT is a promising and safe drug candidate against dermatophytes, particularly in anti-biofilm treatment.
Collapse
Affiliation(s)
- Níura Madalena Bila
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil.,Department of Para-Clinic, School of Veterinary, Universidade Eduardo Mondlane (UEM), Maputo, Mozambique
| | - Caroline Barcelos Costa-Orlandi
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Carolina Orlando Vaso
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Jean Lucas Carvalho Bonatti
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Letícia Ribeiro de Assis
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, Universidade Estadual Paulista (UNESP), Sao Jose do Rio Preto, Brazil
| | - Luís Octavio Regasini
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, Universidade Estadual Paulista (UNESP), Sao Jose do Rio Preto, Brazil
| | - Carla Raquel Fontana
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Ana Marisa Fusco-Almeida
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | | |
Collapse
|
60
|
Veeraraghavan VP, Periadurai ND, Karunakaran T, Hussain S, Surapaneni KM, Jiao X. Green synthesis of silver nanoparticles from aqueous extract of Scutellaria barbata and coating on the cotton fabric for antimicrobial applications and wound healing activity in fibroblast cells (L929). Saudi J Biol Sci 2021; 28:3633-3640. [PMID: 34220213 PMCID: PMC8241602 DOI: 10.1016/j.sjbs.2021.05.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 11/28/2022] Open
Abstract
Scutellaria barbata is a perennial herb which was vastly prescribed in Chinese medicine to treat inflammations, infections and it is also used a detoxifying agent. We synthesized silver nanoparticles with Scutellaria barbata extract and characterized the nanoparticles with UV–Vis spectroscopic analysis, TEM, AFM, FTIR and XRD. The biofilm inhibiting property of synthesized silver nanoparticles were examined with XTT reduction assay and the antimicrobial property was examined with well diffusion method. The silver nanoparticles were also coated with cotton fabrics and their efficacy against antimicrobials was analyzed to prove its application. The cytotoxic property of synthesized silver nanoparticles was examined with L929 fibroblast cells using MTT assay. Finally we analyzed the wound healing property of synthesized silver nanoparticles with wound scratch assay. The result of our UV–Vis spectroscopic analysis confirms Scutellaria barbata aqueous extract reduced silver ions and synthesized silver nanoparticles. The characterization studies TEM, AFM, FTIR and XRD confirms the synthesized silver nanoparticles are in ideal shape and size to be utilized as a drug. The XTT reduction assay proves silver nanoparticles effectively inhibits the biofilm formation in both resistant and sensitive strains. Antimicrobial sensitivity tests confirms synthesized silver nanoparticles and cotton coated synthesized silver nanoparticles both are effective against gram positive, gram negative and fungal species. Further the results of MTT assay confirms the synthesized silver nanoparticles are non toxic and finally the wound healing potency of the nanoparticles was confirmed with wound scratch assay. Over all our results authentically confirms the silver nanoparticles synthesized with Scutellaria barbata aqueous extract is potent wound healing drug.
Collapse
Affiliation(s)
- Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, Tamil Nadu, India
| | - Nanthini Devi Periadurai
- Departments of Microbiology, Molecular Virology and Hospital Infection Control, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, India
| | - Thiruventhan Karunakaran
- Centre for Drug Research, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia.,School of Chemical Sciences, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| | - Sardar Hussain
- Department of Biotechnology, Government Science College, Chitradurga 577501, Karnataka, India
| | - Krishna Mohan Surapaneni
- Departments of Biochemistry, Molecular Virology, Clinical Skills & Simulation and Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India
| | - Xinsheng Jiao
- Department of Cosmetic, Plastic and Burn Surgery, No. 50, Normal Road, The Fourth People's Hospital of Jinan, Jinan 250031, China
| |
Collapse
|
61
|
Effects of Itraconazole and Micafungin on Aspergillus fumigatus Biofilms. Mycopathologia 2021; 186:387-397. [PMID: 33956291 DOI: 10.1007/s11046-021-00534-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/10/2021] [Indexed: 10/20/2022]
Abstract
Aspergillus fumigatus (A. fumigatus) is the most common airborne opportunistic fungal pathogen. Biofilm formation is one of the main pathogenic mechanisms of A. fumigatus. During the past decades, A. fumigatus azole resistance has become prevalent due to the medical and agricultural use of antifungal drugs and fungicides. Until now, the role of fungal biofilms in azole resistance of A. fumigatus remains unclear. In the present study, we compared biofilm drug susceptibility and biofilm formation under itraconazole of azole-resistant strains, sensitive strains, and standard strains, separately. The biofilm viability and matrix thickness at the early and the late stage were measured by XTT assay and Calcofluor white. Our results showed that the sessile minimum inhibitory concentration of itraconazole, which describing the inhibition of drugs on fungi sessile with biofilm, was much higher than the traditional minimal inhibitory concentration of itraconazole. Additionally, low concentrations of itraconazole inhibited biofilm formation of A. fumigatus strains. Notably, biofilm formation by azole-resistant strains could not be inhibited by high concentrations of itraconazole but could be effectively restrained by low concentrations of micafungin, revealing the efficacy of a cell-wall inhibitor to disrupt A. fumigatus biofilm formation. However, late-stage biofilms of both azole-resistant strains and standard strains were hard to disrupt using itraconazole. We found that itraconazole was effective to prevent A. fumigatus biofilm formation at the early stage. For the treatment of A. fumigatus biofilm, our findings suggest that an early-stage preventive strategy is preferred and micafungin is effective to control the azole-resistant strain infection.
Collapse
|
62
|
Lopes LG, Csonka LA, Castellane JAS, Oliveira AW, de Almeida-Júnior S, Furtado RA, Tararam C, Levy LO, Crivellenti LZ, Moretti ML, Giannini MJSM, Pires RH. Disinfectants in a Hemodialysis Setting: Antifungal Activity Against Aspergillus and Fusarium Planktonic and Biofilm Cells and the Effect of Commercial Peracetic Acid Residual in Mice. Front Cell Infect Microbiol 2021; 11:663741. [PMID: 33996634 PMCID: PMC8116949 DOI: 10.3389/fcimb.2021.663741] [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: 02/03/2021] [Accepted: 03/04/2021] [Indexed: 11/17/2022] Open
Abstract
Aspergillus and Fusarium cause a broad spectrum of infections in humans, mainly in immunocompromised patients. Among these, patients undergoing hemodialysis are highly susceptible to infections, requiring a constant and adequate environmental disinfection program. Nevertheless, monitoring the residual disinfectants can contribute to the morbidity and mortality reduction in these patients. Here, we evaluated the susceptibility of Aspergillus spp. (n=19) and Fusarium spp. (n=13) environmental isolates against disinfectants (acetic acid, citric acid, peracetic acid, sodium hypochlorite, and sodium metabisulphite) at different concentrations and time exposures. Also, we investigated the in vivo toxicity of the peracetic acid residual concentration in mice. Fusarium isolates were identified by F. equiseti, F. oxysporum and F. solani while Aspergillus presented clinically relevant species (A. fumigatus, A. niger and A. terreus) and environmental ones. Against planktonic cells, only two disinfectants (acetic acid and sodium hypochlorite) showed a fungicidal effect on Fusarium spp., while only one (sodium hypochlorite) was effective against Aspergillus spp. Both fungi formed robust in vitro biofilms with large amounts of the extracellular matrix, as evidenced by electron micrographs. Exposure of fungal biofilms to disinfectants showed sensitivity to three (acetic, citric, and peracetic acids), although the concentrations and times of exposure varied according to the fungal genus. Mice exposure to the residual dose of peracetic acid during 60 weeks showed anatomopathological, hematological, and biochemical changes. The implementation of news control measures and those that already exist can help reduce infections, the second cause of death and morbidity in these patients, besides providing safety and well-being to them, a priority of any quality health program.
Collapse
Affiliation(s)
- Leonardo G. Lopes
- Postgraduate Program in Health Promotion, University of Franca, Franca, Brazil
| | - Larissa A. Csonka
- Postgraduate Program in Health Promotion, University of Franca, Franca, Brazil
| | | | | | | | | | - Cibele Tararam
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | | | | | | | - Regina H. Pires
- Postgraduate Program in Health Promotion, University of Franca, Franca, Brazil
| |
Collapse
|
63
|
Luo Y, McAuley DF, Fulton CR, Sá Pessoa J, McMullan R, Lundy FT. Targeting Candida albicans in dual-species biofilms with antifungal treatment reduces Staphylococcus aureus and MRSA in vitro. PLoS One 2021; 16:e0249547. [PMID: 33831044 PMCID: PMC8031443 DOI: 10.1371/journal.pone.0249547] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/20/2021] [Indexed: 11/18/2022] Open
Abstract
Polymicrobial biofilms consisting of fungi and bacteria are frequently formed on endotracheal tubes and may contribute to development of ventilator associated pneumonia (VAP) in critically ill patients. This study aimed to determine the role of early Candida albicans biofilms in supporting dual-species (dual-kingdom) biofilm formation with respiratory pathogens in vitro, and investigated the effect of targeted antifungal treatment on bacterial cells within the biofilms. Dual-species biofilm formation between C. albicans and three respiratory pathogens commonly associated with VAP (Pseudomonas aeruginosa, Escherichia coli and Staphylococcus aureus) was studied using quantitative PCR. It was shown that early C. albicans biofilms enhanced the numbers of E. coli and S. aureus (including methicillin resistant S. aureus; MRSA) but not P. aeruginosa within dual-species biofilms. Transwell assays demonstrated that contact with C. albicans was required for the increased bacterial cell numbers observed. Total Internal Reflection Fluorescence microscopy showed that both wild type and hyphal-deficient C. albicans provided a scaffold for initial bacterial adhesion in dual species biofilms. qPCR results suggested that further maturation of the dual-species biofilm significantly increased bacterial cell numbers, except in the case of E.coli with hyphal-deficient C. albicans (Ca_gcn5Δ/Δ). A targeted preventative approach with liposomal amphotericin (AmBisome®) resulted in significantly decreased numbers of S. aureus in dual-species biofilms, as determined by propidium monoazide-modified qPCR. Similar results were observed when dual-species biofilms consisting of clinical isolates of C. albicans and MRSA were treated with liposomal amphotericin. However, reductions in E. coli numbers were not observed following liposomal amphotericin treatment. We conclude that early C. albicans biofilms have a key supporting role in dual-species biofilms by enhancing bacterial cell numbers during biofilm maturation. In the setting of increasing antibiotic resistance, an important and unexpected consequence of antifungal treatment of dual-species biofilms, is the additional benefit of decreased growth of multi-drug resistant bacteria such as MRSA, which could represent a novel future preventive strategy.
Collapse
Affiliation(s)
- Yu Luo
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
- Belfast Health & Social Care Trust, Regional Intensive Care Unit, Royal Victoria Hospital, Belfast, United Kingdom
| | - Catherine R. Fulton
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Joana Sá Pessoa
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Ronan McMullan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Fionnuala T. Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
64
|
Pan G, Li Y, Che X, Tian D, Han W, Wang Z, Zhao Y, Ren S, Xu Y, Hao G, Guo M, Xiao N, Kong F. New Thio-Compounds and Monoterpenes With Anti-inflammatory Activities From the Fungus Aspergillus sp. CYH26. Front Microbiol 2021; 12:668938. [PMID: 33841388 PMCID: PMC8025228 DOI: 10.3389/fmicb.2021.668938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/03/2021] [Indexed: 01/10/2023] Open
Abstract
Two new thio-compounds named aspergerthinol A and B (1 and 2) and two new monoterpenes named aspergerthinacids A and B (3 and 4) were isolated from the fungus Aspergillus sp. CYH26 from the rhizosphere soil of Cynanchum bungei Decne. The structures of compounds were elucidated by spectroscopic data and quantum NMR and ECD calculations. Compounds 1 and 2 represented a new family of sulfur containing natural products with a 3-methyl-4H-cyclopenta[b]thiophen-4-one skeleton. Compounds 1-4 showed inhibitory activities against nitric oxide (NO) with IC50 values of 38.0, 19.8, 46.3, and 56.6 μM, respectively.
Collapse
Affiliation(s)
- Guojun Pan
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Yanling Li
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Xinyu Che
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agriculture University, Tai'an, China
| | - Dan Tian
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agriculture University, Tai'an, China
| | - Wenjie Han
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Zimin Wang
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Yanfen Zhao
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Shuang Ren
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Yiru Xu
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Gangping Hao
- College of Life Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Mengfei Guo
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, China
| | - Na Xiao
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agriculture University, Tai'an, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fandong Kong
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, China
| |
Collapse
|
65
|
Mamouei Z, Singh S, Lemire B, Gu Y, Alqarihi A, Nabeela S, Li D, Ibrahim A, Uppuluri P. An evolutionarily diverged mitochondrial protein controls biofilm growth and virulence in Candida albicans. PLoS Biol 2021; 19:e3000957. [PMID: 33720927 PMCID: PMC8007014 DOI: 10.1371/journal.pbio.3000957] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/29/2021] [Accepted: 01/29/2021] [Indexed: 11/19/2022] Open
Abstract
A forward genetic screening approach identified orf19.2500 as a gene controlling Candida albicans biofilm dispersal and biofilm detachment. Three-dimensional (3D) protein modeling and bioinformatics revealed that orf19.2500 is a conserved mitochondrial protein, structurally similar to, but functionally diverged from, the squalene/phytoene synthases family. The C. albicans orf19.2500 is distinguished by 3 evolutionarily acquired stretches of amino acid inserts, absent from all other eukaryotes except a small number of ascomycete fungi. Biochemical assays showed that orf19.2500 is required for the assembly and activity of the NADH ubiquinone oxidoreductase Complex I (CI) of the respiratory electron transport chain (ETC) and was thereby named NDU1. NDU1 is essential for respiration and growth on alternative carbon sources, important for immune evasion, required for virulence in a mouse model of hematogenously disseminated candidiasis, and for potentiating resistance to antifungal drugs. Our study is the first report on a protein that sets the Candida-like fungi phylogenetically apart from all other eukaryotes, based solely on evolutionary "gain" of new amino acid inserts that are also the functional hub of the protein.
Collapse
Affiliation(s)
- Zeinab Mamouei
- David Geffen School of Medicine, University of California (UCLA), Los Angeles, California, United States of America
| | - Shakti Singh
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Bernard Lemire
- Department of Biochemistry, University of Alberta, Alberta, Canada
| | - Yiyou Gu
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Abdullah Alqarihi
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Sunna Nabeela
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Ashraf Ibrahim
- David Geffen School of Medicine, University of California (UCLA), Los Angeles, California, United States of America
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Priya Uppuluri
- David Geffen School of Medicine, University of California (UCLA), Los Angeles, California, United States of America
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- * E-mail:
| |
Collapse
|
66
|
Albayaty YN, Thomas N, Ramírez-García PD, Davis TP, Quinn JF, Whittaker MR, Prestidge CA. Polymeric micelles with anti-virulence activity against Candida albicans in a single- and dual-species biofilm. Drug Deliv Transl Res 2021; 11:1586-1597. [PMID: 33713317 DOI: 10.1007/s13346-021-00943-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
Abstract
Infections caused by fungal biofilms with rapidly evolving resistance against the available antifungal agents are difficult to manage. These difficulties demand new strategies for effective eradication of biofilms from both biological and inert surfaces. In this study, polymeric micelles comprised of di-block polymer, poly-(ethylene glycol) methyl ether methacrylate and poly 2-(N,N-diethylamino) ethyl methacrylate polymer, P(PEGMA-b-DEAEMA), were observed to exhibit remarkable inhibitory effects on hyphal growth of Candida albicans (C. albicans) and C. tropicalis, thus preventing biofilm formation and removing existing biofilms. P(PEGMA-b-DEAEMA) micelles showed biofilm removal efficacy of > 40% and a 1.4-log reduction in cell viability of C. albicans in its single-species biofilms. In addition, micelles alone promoted high removal percentage in a mixed biofilm of C. albicans and C. tropicalis (~ 70%) and remarkably reduced cell viability of both strains. Co-delivery of fluconazole (Flu) and amphotericin B (AmB) with micelles showed synergistic effects on C. albicans biofilms (3-log reduction for AmB and 2.2-log reduction for Flu). Similar effects were noted on C. albicans planktonic cells when treated with the micellar system combined with AmB but not with Flu. Moreover, micelle-drug combinations showed an enhancement in the antibiofilm activity of Flu and AmB against dual-species biofilms. Furthermore, in vivo studies using Caenorhabditis elegans nematodes revealed no obvious toxicity of the micelles. Targeting morphologic transitions provides a new strategy for defeating fungal biofilms of polymorphic resistance strains and can be potentially used in counteracting Candida virulence.
Collapse
Affiliation(s)
- Yassamin N Albayaty
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
- Basil Hetzel Institute for Translational Health Research, Woodville South, Woodville, SA, 5011, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia
| | - Nicky Thomas
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
- Basil Hetzel Institute for Translational Health Research, Woodville South, Woodville, SA, 5011, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia
| | - Paulina D Ramírez-García
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC, 3052, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC, 3052, Australia
| | - John F Quinn
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC, 3052, Australia
- Department of Chemical Engineering, Faculty of Engineering, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Michael R Whittaker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC, 3052, Australia
| | - Clive A Prestidge
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Australia.
| |
Collapse
|
67
|
Fulgêncio DLA, da Costa RA, Guilhelmelli F, Silva CMDS, Ortega DB, de Araujo TF, Silva PS, Silva-Pereira I, Albuquerque P, Barreto CC. In vitro antifungal activity of pelgipeptins against human pathogenic fungi and Candida albicans biofilms. AIMS Microbiol 2021; 7:28-39. [PMID: 33659767 PMCID: PMC7921374 DOI: 10.3934/microbiol.2021003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Systemic mycoses have become a major cause of morbidity and mortality, particularly among immunocompromised hosts and long-term hospitalized patients. Conventional antifungal agents are limited because of not only their costs and toxicity but also the rise of resistant strains. Lipopeptides from Paenibacillus species exhibit antimicrobial activity against a wide range of human and plant bacterial pathogens. However, the antifungal potential of these compounds against important human pathogens has not yet been fully evaluated, except for Candida albicans. Paenibacillus elgii produces a family of lipopeptides named pelgipeptins, which are synthesized by a non-ribosomal pathway, such as polymyxin. The present study aimed to evaluate the activity of pelgipeptins produced by P. elgii AC13 against Cryptococcus neoformans, Paracoccidioides brasiliensis, and Candida spp. Pelgipeptins were purified from P. elgii AC13 cultures and characterized by high-performance liquid chromatography (HPLC) and mass spectrometry (MALDI-TOF MS). The in vitro antifugal activity of pelgipeptins was evaluated against C. neoformans H99, P. brasiliensis PB18, C. albicans SC 5314, Candida glabrata ATCC 90030, and C. albicans biofilms. Furthermore, the minimal inhibitory concentration (MIC) was determined according to the CLSI microdilution method. Fluconazole and amphotericin B were also used as a positive control. Pelgipeptins A to D inhibited the formation and development of C. albicans biofilms and presented activity against all tested microorganisms. The minimum inhibitory concentration values ranged from 4 to 64 µg/mL, which are in the same range as fluconazole MICs. These results highlight the potential of pelgipeptins not only as antimicrobials against pathogenic fungi that cause systemic mycoses but also as coating agents to prevent biofilm formation on medical devices.
Collapse
Affiliation(s)
| | - Rosiane Andrade da Costa
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil
| | - Fernanda Guilhelmelli
- Laboratory of Molecular Biology, Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Calliandra Maria de Souza Silva
- Laboratory of Molecular Biology, Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Daniel Barros Ortega
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil
| | - Thiago Fellipe de Araujo
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil
| | - Philippe Spezia Silva
- Laboratory of Molecular Biology, Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Ildinete Silva-Pereira
- Laboratory of Molecular Biology, Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Patrícia Albuquerque
- Laboratory of Molecular Biology, Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Faculty of Ceilandia, University of Brasília, Brasília, Brazil
| | - Cristine Chaves Barreto
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil
| |
Collapse
|
68
|
Zuo X, Liu Y, Cai X, Zhan L, Hu K. Association of different Candida species with catheter-related candidemia, and the potential antifungal treatments against their adhesion properties and biofilm-forming capabilities. J Clin Lab Anal 2021; 35:e23738. [PMID: 33608902 PMCID: PMC8059721 DOI: 10.1002/jcla.23738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 01/12/2023] Open
Abstract
Background To compare the adhesion properties and biofilm‐forming capabilities of 27 Candida isolates obtained from catheter‐related candidemia patients and to evaluate the inhibitory effects of antifungal agents on different Candida species. Material and Methods Seven C. albicans, six C. parapsilosis, five C. guilliermondii, five C. tropicalis, and four C. glabrata clinical isolates were investigated. We quantified the adherence of these Candida species by flow cytometric method and evaluated the formation of biofilms by XTT reduction and crystal violet methods. Actions of micafungin (MF), fluconazole (FZ), and N‐acetylcysteine (NAC) on the adhesion and biofilm formation of different Candida species were determined. Results Non‐albicans Candida species were demonstrated to have stronger adhesion abilities compared with C. albicans. The biofilm‐forming capabilities of different Candida species were varied considerably, and the degree of biofilm formation might be affected by different assay approaches. Interestingly, C. parapsilosis displayed the highest biofilm formation abilities, while C. glabrata exhibited the lowest total biomass and metabolic activity. Furthermore, the inhibitory activities of MF, FZ, and NAC on fungal adhesion and biofilm formation were evaluated, and the results indicated that MF could reduce the adhesion ability and biofilm metabolism more significantly (p < 0.05), and its antifungal activity was elevated in a dose‐dependent manner. Conclusion Non‐albicans Candida species, especially C. guilliermondii, C. tropicalis, and C. parapsilosis, exhibited higher adhesion ability in catheter‐related candidemia patients. However, these Candida species had varied biofilm‐forming capabilities. MF tended to have stronger inhibitory effects against both adhesion and biofilm formation of different Candida species.
Collapse
Affiliation(s)
- Xiao‐shu Zuo
- Department of Critical Care MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yanan Liu
- Department of Critical Care MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xuan Cai
- Department of Clinical LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Liying Zhan
- Department of Critical Care MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ke Hu
- Department of Respiratory and Critical Care MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
69
|
The Antifungal and Synergistic Effect of Bisphosphonates in Cryptococcus. Antimicrob Agents Chemother 2021; 65:AAC.01753-20. [PMID: 33139289 DOI: 10.1128/aac.01753-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/24/2020] [Indexed: 01/04/2023] Open
Abstract
New treatment strategies are required for cryptococcosis, a leading mycosis in HIV-AIDS patients. Following the identification of Cryptococcus proteins differentially expressed in response to fluconazole, we targeted farnesyl pryrophosphate synthetase (FPPS), an enzyme in the squalene biosynthesis pathway, using nitrogenous bisphosphonates. We hypothesized that these would disrupt squalene synthesis and thereby produce synergy with fluconazole, which acts on a downstream pathway that requires squalene. The susceptibilities of 39 clinical isolates from 6 different species of Cryptococcus were assessed for bisphosphonates and fluconazole, used both independently and in combination. Effective fluconazole-bisphosphonate combinations were then assessed for fungicidal activity, efficacy against biofilms, and ability to resolve cryptococcosis in an invertebrate model. The nitrogenous bisphosphonates risedronate, alendronate, and zoledronate were antifungal against all strains tested. Zoledronate was the most effective (geometric mean MIC = 113.03 mg/liter; risedronate = 378.49 mg/liter; alendronate = 158.4 mg/liter) and was broadly synergistic when combined with fluconazole, with a fractional inhibitory concentration index (FICI) of ≤0.5 in 92% of isolates. Fluconazole and zoledronate in combination were fungicidal in a time-kill assay, inhibited Cryptococcus biofilms, prevented the development of fluconazole resistance, and resolved infection in a nematode model. Supplementation with squalene eliminated bisphosphonate-mediated synergy, demonstrating that synergy was due to the inhibition of squalene biosynthesis. This study demonstrates the utility of targeting squalene synthesis for improving the efficacy of azole-based antifungal drugs and suggests bisphosphonates are promising lead compounds for further antifungal development.
Collapse
|
70
|
Barreto JO, do Nascimento FBSA, Fonseca SGDC, Serpa Sampaio L, da Silva CR, de Andrade Neto JB, Júnior HVN, Regis RR. Microbiological evaluation of an experimental denture cleanser containing essential oil of Lippia sidoides. BIOFOULING 2021; 37:117-130. [PMID: 33593175 DOI: 10.1080/08927014.2021.1885649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
The antimicrobial activity of an experimental solution containing essential oil of Lippia sidoides for denture cleaning was evaluated by (1) minimum inhibitory (MIC) and fungicidal/bactericidal concentration (MFC/MBC) tests against Candida albicans, Staphylococcus aureus, and Pseudomona aeruginosa; (2) the metabolic activity of C. albicans biofilm formed on flat-bottom microplates and denture base specimens based on the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT); and (3) scanning electron microscopy, to evaluate the fungal biofilm morphology. The solution showed antimicrobial action against the pathogens tested (C. albicans - MIC and MFC: 19.53 µg ml-1, S. aureus - MIC and MBC: 78.12 µg ml-1, P. aeruginosa - MIC: 625 µg ml-1, MBC: 2,500 µg ml-1), reduced the metabolic activity of C. albicans biofilm up to 97%, and caused cell wall damage at low concentrations (195.3-390.6 µg ml-1) and in short time periods (20 min). Therefore, the experimental solution has the potential to be used as an alternative in the prevention and treatment of denture-induced infections.
Collapse
Affiliation(s)
- Joel Oliveira Barreto
- Department of Restorative Dentistry, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
| | - Francisca Bruna Stefany Aires do Nascimento
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, Brazil
| | | | - Letícia Serpa Sampaio
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, Brazil
| | - Cecília Rocha da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, Brazil
| | - João Batista de Andrade Neto
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, Brazil
| | - Hélio Vitoriano Nobre Júnior
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, Brazil
| | - Rômulo Rocha Regis
- Department of Restorative Dentistry, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
71
|
Stevanović NL, Aleksic I, Kljun J, Skaro Bogojevic S, Veselinovic A, Nikodinovic-Runic J, Turel I, Djuran MI, Glišić BĐ. Copper(II) and Zinc(II) Complexes with the Clinically Used Fluconazole: Comparison of Antifungal Activity and Therapeutic Potential. Pharmaceuticals (Basel) 2020; 14:24. [PMID: 33396681 PMCID: PMC7823955 DOI: 10.3390/ph14010024] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 01/18/2023] Open
Abstract
Copper(II) and zinc(II) complexes with clinically used antifungal drug fluconazole (fcz), {[CuCl2(fcz)2].5H2O}n, 1, and {[ZnCl2(fcz)2]·2C2H5OH}n, 2, were prepared and characterized by spectroscopic and crystallographic methods. The polymeric structure of the complexes comprises four fluconazole molecules monodentately coordinated via the triazole nitrogen and two chlorido ligands. With respect to fluconazole, complex 2 showed significantly higher antifungal activity against Candida krusei and Candida parapsilosis. All tested compounds reduced the total amount of ergosterol at subinhibitory concentrations, indicating that the mode of activity of fluconazole was retained within the complexes, which was corroborated via molecular docking with cytochrome P450 sterol 14α-demethylase (CYP51) as a target. Electrostatic, steric and internal energy interactions between the complexes and enzyme showed that 2 has higher binding potency to this target. Both complexes showed strong inhibition of C. albicans filamentation and biofilm formation at subinhibitory concentrations, with 2 being able to reduce the adherence of C. albicans to A549 cells in vitro. Complex 2 was able to reduce pyocyanin production in Pseudomonas aeruginosa between 10% and 25% and to inhibit its biofilm formation by 20% in comparison to the untreated control. These results suggest that complex 2 may be further examined in the mixed Candida-P. aeruginosa infections.
Collapse
Affiliation(s)
- Nevena Lj. Stevanović
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia;
| | - Ivana Aleksic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (I.A.); (S.S.B.)
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia;
| | - Sanja Skaro Bogojevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (I.A.); (S.S.B.)
| | - Aleksandar Veselinovic
- Department of Chemistry, Faculty of Medicine, University of Niš, Blvd. Dr Zorana Djindjica 81, 18108 Niš, Serbia;
| | - Jasmina Nikodinovic-Runic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (I.A.); (S.S.B.)
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia;
| | - Miloš I. Djuran
- Department of Chemical and Biological Sciences, Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000 Belgrade, Serbia
| | - Biljana Đ. Glišić
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia;
| |
Collapse
|
72
|
Wall G, Chen E, Hull MV, Lopez-Ribot JL. Screening the CALIBR ReFRAME Library in Search for Inhibitors of Candida auris Biofilm Formation. Front Cell Infect Microbiol 2020; 10:597931. [PMID: 33324579 PMCID: PMC7723901 DOI: 10.3389/fcimb.2020.597931] [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] [Received: 08/22/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Candida auris is an emerging yeast which, since its first isolation about a decade ago, has spread rapidly and triggered major infectious outbreaks in health care facilities around the world. C. auris strains often display resistance to clinically-used antifungal agents, contributing to high mortality rates. Thus, there is an urgent need for new antifungals to contain the spread of this emerging multi-drug resistant pathogen and to improve patient outcomes. However, the timeline for the development of a new antifungal agent typically exceeds 10‑15 years. Thus, repurposing of current drugs could significantly accelerate the development and eventual deployment of novel therapies for the treatment of C. auris infections. Toward this end, in this study we have profiled a library of known drugs encompassing approximately 12,000 clinical-stage or FDA-approved small molecules in search for known molecules with antifungal activity against C. auris; more specifically, those capable of inhibiting C. auris biofilm formation. From this library, 100 compounds displaying antifungal activity were identified in the initial screen, including 26 compounds for which a dose-response relationship with biofilm-inhibitory activity against C. auris could be confirmed. Of these, five were identified as the most interesting potential repositionable candidates. Due to their known pharmacological and human safety profiles, identification of such compounds should allow for their accelerated preclinical and clinical development for the treatment of C. auris infections.
Collapse
Affiliation(s)
- Gina Wall
- Department of Biology and The South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Emily Chen
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, United States
| | - Mitchell V Hull
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, United States
| | - Jose L Lopez-Ribot
- Department of Biology and The South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
73
|
The Transcription Factor SomA Synchronously Regulates Biofilm Formation and Cell Wall Homeostasis in Aspergillus fumigatus. mBio 2020; 11:mBio.02329-20. [PMID: 33173002 PMCID: PMC7667024 DOI: 10.1128/mbio.02329-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The cell wall is essential for fungal viability and is absent from human hosts; thus, drugs disrupting cell wall biosynthesis have gained more attention. Caspofungin is a member of a new class of clinically approved echinocandin drugs to treat invasive aspergillosis by blocking β-1,3-glucan synthase, thus damaging the fungal cell wall. Here, we demonstrate that caspofungin and other cell wall stressors can induce galactosaminogalactan (GAG)-dependent biofilm formation in the human pathogen Aspergillus fumigatus. We further identified SomA as a master transcription factor playing a dual role in both biofilm formation and cell wall homeostasis. SomA plays this dual role by direct binding to a conserved motif upstream of GAG biosynthetic genes and genes involved in cell wall stress sensors, chitin synthases, and β-1,3-glucan synthase. Collectively, these findings reveal a transcriptional control pathway that integrates biofilm formation and cell wall homeostasis and suggest SomA as an attractive target for antifungal drug development. Polysaccharides are key components of both the fungal cell wall and biofilm matrix. Despite having distinct assembly and regulation pathways, matrix exopolysaccharide and cell wall polysaccharides share common substrates and intermediates in their biosynthetic pathways. It is not clear, however, if the biosynthetic pathways governing the production of these polysaccharides are cooperatively regulated. Here, we demonstrate that cell wall stress promotes production of the exopolysaccharide galactosaminogalactan (GAG)-depend biofilm formation in the major fungal pathogen of humans Aspergillus fumigatus and that the transcription factor SomA plays a crucial role in mediating this process. A core set of SomA target genes were identified by transcriptome sequencing and chromatin immunoprecipitation coupled to sequencing (ChIP-Seq). We identified a novel SomA-binding site in the promoter regions of GAG biosynthetic genes agd3 and ega3, as well as its regulators medA and stuA. Strikingly, this SomA-binding site was also found in the upstream regions of genes encoding the cell wall stress sensors, chitin synthases, and β-1,3-glucan synthase. Thus, SomA plays a direct regulation of both GAG and cell wall polysaccharide biosynthesis. Consistent with these findings, SomA is required for the maintenance of normal cell wall architecture and compositions in addition to its function in biofilm development. Moreover, SomA was found to globally regulate glucose uptake and utilization, as well as amino sugar and nucleotide sugar metabolism, which provides precursors for polysaccharide synthesis. Collectively, our work provides insight into fungal adaptive mechanisms in response to cell wall stress where biofilm formation and cell wall homeostasis were synchronously regulated.
Collapse
|
74
|
Wei L, Li W, Entcheva E, Li Z. Microfluidics-enabled 96-well perfusion system for high-throughput tissue engineering and long-term all-optical electrophysiology. LAB ON A CHIP 2020; 20:4031-4042. [PMID: 32996969 PMCID: PMC7680692 DOI: 10.1039/d0lc00615g] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
This work demonstrates a novel high-throughput (HT) microfluidics-enabled uninterrupted perfusion system (HT-μUPS) and validates its use with chronic all-optical electrophysiology in human excitable cells. HT-μUPS consists of a soft multichannel microfluidic plate cover which could button on a commercial HT 96-well plate. Herein, we demonstrate the manufacturing process of the system and its usages in acute and chronic all-optical electrophysiological studies of human induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CM) and engineered excitable (spiking HEK) cells. HT-μUPS perfusion maintained functional voltage and calcium responses in iPSC-CM and spiking HEK cells under spontaneous conditions and under optogenetic pacing. Long-term culture with HT-μUPS improved cell viability and optogenetically-tracked calcium responses in spiking HEK cells. The simplicity of this design and its compatibility with HT all-optical electrophysiology can empower cell-based assays for personalized medicine using patient-derived cells.
Collapse
Affiliation(s)
- Lai Wei
- Department of Biomedical Engineering, The George Washington University, Washington DC, USA
- These authors contributed equally: Lai Wei, Weizhen Li
| | - Weizhen Li
- Department of Biomedical Engineering, The George Washington University, Washington DC, USA
- These authors contributed equally: Lai Wei, Weizhen Li
| | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington DC, USA
- ,
| | - Zhenyu Li
- Department of Biomedical Engineering, The George Washington University, Washington DC, USA
- ,
| |
Collapse
|
75
|
Antibiofilm Activity on Candida albicans and Mechanism of Action on Biomembrane Models of the Antimicrobial Peptide Ctn[15-34]. Int J Mol Sci 2020; 21:ijms21218339. [PMID: 33172206 PMCID: PMC7664368 DOI: 10.3390/ijms21218339] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
Ctn[15–34], the C-terminal fragment of crotalicidin, an antimicrobial peptide from the South American rattlesnake Crotalus durissus terrificus venom, displays remarkable anti-infective and anti-proliferative activities. Herein, its activity on Candida albicans biofilms and its interaction with the cytoplasmic membrane of the fungal cell and with a biomembrane model in vitro was investigated. A standard C. albicans strain and a fluconazole-resistant clinical isolate were exposed to the peptide at its minimum inhibitory concentration (MIC) (10 µM) and up to 100 × MIC to inhibit biofilm formation and its eradication. A viability test using XTT and fluorescent dyes, confocal laser scanning microscopy, and atomic force microscopy (AFM) were used to observe the antibiofilm effect. To evaluate the importance of membrane composition on Ctn[15–34] activity, C. albicans protoplasts were also tested. Fluorescence assays using di-8-ANEPPS, dynamic light scattering, and zeta potential measurements using liposomes, protoplasts, and C. albicans cells indicated a direct mechanism of action that was dependent on membrane interaction and disruption. Overall, Ctn[15–34] showed to be an effective antifungal peptide, displaying antibiofilm activity and, importantly, interacting with and disrupting fungal plasma membrane.
Collapse
|
76
|
Rodriguez-de la Noval C, Ruiz Mendoza S, de Souza Gonçalves D, da Silva Ferreira M, Honorato L, Peralta JM, Nimrichter L, Guimarães AJ. Protective Efficacy of Lectin-Fc(IgG) Fusion Proteins In Vitro and in a Pulmonary Aspergillosis In Vivo Model. J Fungi (Basel) 2020; 6:jof6040250. [PMID: 33120893 PMCID: PMC7712007 DOI: 10.3390/jof6040250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
Aspergillosis cases by Aspergillus fumigatus have increased, along with fungal resistance to antifungals, urging the development of new therapies. Passive immunization targeting common fungal antigens, such as chitin and β-glucans, are promising and would eliminate the need of species-level diagnosis, thereby expediting the therapeutic intervention. However, these polysaccharides are poorly immunogenic. To overcome this drawback, we developed the lectin-Fc(IgG) fusion proteins, Dectin1-Fc(IgG2a), Dectin1-Fc(IgG2b) and wheat germ agglutinin (WGA)-Fc(IgG2a), based on their affinity to β-1,3-glucan and chitooligomers, respectively. The WGA-Fc(IgG2a) previously demonstrated antifungal activity against Histoplasma capsulatum, Cryptococcus neoformans and Candida albicans. In the present work, we evaluated the antifungal properties of these lectin-Fc(s) against A. fumigatus. Lectin-Fc(IgG)(s) bound in a dose-dependent manner to germinating conidia and this binding increased upon conidia germination. Both lectin-Fc(IgG)(s) displayed in vitro antifungal effects, such as inhibition of conidia germination, a reduced length of germ tubes and a diminished biofilm formation. Lectin-Fc(IgG)(s) also enhanced complement deposition on conidia and macrophage effector functions, such as increased phagocytosis and killing of fungi. Finally, administration of the Dectin-1-Fc(IgG2b) and WGA-Fc(IgG2a) protected mice infected with A. fumigatus, with a 20% survival and a doubled life-span of the infected mice, which was correlated to a fungal burden reduction in lungs and brains of treated animals. These results confirm the potential of lectin-Fc(IgGs)(s) as a broad-spectrum antifungal therapeutic.
Collapse
Affiliation(s)
- Claudia Rodriguez-de la Noval
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói 24020-141, RJ, Brazil; (C.R.-d.l.N.); (S.R.M.); (D.d.S.G.); (M.d.S.F.)
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.H.); (L.N.)
| | - Susana Ruiz Mendoza
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói 24020-141, RJ, Brazil; (C.R.-d.l.N.); (S.R.M.); (D.d.S.G.); (M.d.S.F.)
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Diego de Souza Gonçalves
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói 24020-141, RJ, Brazil; (C.R.-d.l.N.); (S.R.M.); (D.d.S.G.); (M.d.S.F.)
- Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil
| | - Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói 24020-141, RJ, Brazil; (C.R.-d.l.N.); (S.R.M.); (D.d.S.G.); (M.d.S.F.)
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.H.); (L.N.)
| | - José Mauro Peralta
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil
| | - Leonardo Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.H.); (L.N.)
| | - Allan J. Guimarães
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói 24020-141, RJ, Brazil; (C.R.-d.l.N.); (S.R.M.); (D.d.S.G.); (M.d.S.F.)
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas (PPGMPA), Instituto Biomédico, Universidade Federal Fluminense, Rua Professor Hernani Pires de Melo 101, São Domingos, Niterói 24210-130, RJ, Brazil
- Correspondence: ; Tel.: +55-21-2629-2410
| |
Collapse
|
77
|
Sun Y, Gao L, Yuan M, Yuan L, Yang J, Zeng T. In vitro and in vivo Study of Antifungal Effect of Pyrvinium Pamoate Alone and in Combination With Azoles Against Exophiala dermatitidis. Front Cell Infect Microbiol 2020; 10:576975. [PMID: 33194816 PMCID: PMC7649562 DOI: 10.3389/fcimb.2020.576975] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Infections of Exophiala dermatitidis are often chronic and recalcitrant. Combination therapies with novel compounds and azoles could be an effective solution. Previously, we have demonstrated that pyrvinium pamoate exerted antifungal activity alone and favorable synergy with azoles against planktonic E. dermatitidis. Herein, the underlying antifungal mode of action were investigated. Pyrvinium alone showed sessile MIC50 (SMIC50) of 8->16 μg/ml against E. dermatitidis biofilms. However, synergism of PP with itraconazole, voriconazole, and posaconazole were observed against 16 (88.9%), 9 (50%), and 13 (72.2%) strains of E. dermatitidis biofilms. In accordance with in vitro susceptibilities, pyrvinium alone at concentration of 2 μg/ml resulted in significant growth restriction of planktonic E. dermatitidis. Pyrvinium alone resulted in reduction of biofilm formation. Higher concentration of pyrvinium was associate with more progressive reduction of biofilm mass. The in vivo activity of pyrvinium alone and combined with azoles was evaluated using Galleria mellonella model. Pyrvinium alone significantly improved the survival rate of larvae (P < 0.0001). The combination of pyrvinium and voriconazole or posaconazole acted synergistically in vivo (P < 0.05). Fungal burden determination revealed significant reduction of numbers of colony forming unit (CFU) in larvae treated with pyrvinium-itraconazole and pyrvinium-posaconazole compared to itraconazole or posaconazole alone group, respectively. The effect of pyrvinium on apoptosis, expression of TOR and HSP90, and drug efflux reversal were evaluated by PI/Annexin V staining, Real-Time Quantitative PCR and Rhodamine 6G assay, respectively. Pyrvinium alone or combined with azoles significantly (P < 0.05) increased late apoptosis or necrosis of E. dermatitidis cells. Pyrvinium combined with posaconazole significantly decreased the expression of TOR and Hsp90 compared to posaconazole alone group (P < 0.05). Pyrvinium resulted in significant (P < 0.05) decrease of the efflux of Rhodamine 6G. These findings suggested pyrvinium could be a promising synergist with azoles. The underlying mechanisms could be explained by inducing apoptosis/necrosis, inhibition of drug efflux pumps, and signaling pathways related with stress response and growth control.
Collapse
Affiliation(s)
- Yi Sun
- Department of Dermatology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Lujuan Gao
- Department of Dermatology, Zhongshan Hospital Fudan University (Xiamen Branch), Xiamen, China
- Department of Dermatology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Mingzhu Yuan
- Department of Clinical Medicine, Yangtze University, Jingzhou, China
| | - Lu Yuan
- Department of Pathology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Ji Yang
- Department of Dermatology, Zhongshan Hospital Fudan University (Xiamen Branch), Xiamen, China
- Department of Dermatology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Tongxiang Zeng
- Department of Dermatology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| |
Collapse
|
78
|
Sumiyoshi M, Miyazaki T, Makau JN, Mizuta S, Tanaka Y, Ishikawa T, Makimura K, Hirayama T, Takazono T, Saijo T, Yamaguchi H, Shimamura S, Yamamoto K, Imamura Y, Sakamoto N, Obase Y, Izumikawa K, Yanagihara K, Kohno S, Mukae H. Novel and potent antimicrobial effects of caspofungin on drug-resistant Candida and bacteria. Sci Rep 2020; 10:17745. [PMID: 33082485 PMCID: PMC7576149 DOI: 10.1038/s41598-020-74749-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Echinocandins, including caspofungin, micafungin, and anidulafungin, are first-line antifungal agents for the treatment of invasive candidiasis. They exhibit fungicidal activity by inhibiting the synthesis of β-1,3-D-glucan, an essential component of the fungal cell wall. However, they are active only against proliferating fungal cells and unable to completely eradicate fungal cells even after a 24 h drug exposure in standard time-kill assays. Surprisingly, we found that caspofungin, when dissolved in low ionic solutions, had rapid and potent antimicrobial activities against multidrug-resistant (MDR) Candida and bacteria cells even in non-growth conditions. This effect was not observed in 0.9% NaCl or other ion-containing solutions and was not exerted by other echinocandins. Furthermore, caspofungin dissolved in low ionic solutions drastically reduced mature biofilm cells of MDR Candida auris in only 5 min, as well as Candida-bacterial polymicrobial biofilms in a catheter-lock therapy model. Caspofungin displayed ion concentration-dependent conformational changes and intracellular accumulation with increased reactive oxygen species production, indicating a novel mechanism of action in low ionic conditions. Importantly, caspofungin dissolved in 5% glucose water did not exhibit increased toxicity to human cells. This study facilitates the development of new therapeutic strategies in the management of catheter-related biofilm infections.
Collapse
Affiliation(s)
- Makoto Sumiyoshi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Taiga Miyazaki
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Juliann Nzembi Makau
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima, 890-0065, Japan
| | - Koichi Makimura
- Medical Mycology, Graduate School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Tatsuro Hirayama
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Tomomi Saijo
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Hiroyuki Yamaguchi
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shintaro Shimamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kazuko Yamamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yoshifumi Imamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Noriho Sakamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yasushi Obase
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shigeru Kohno
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
79
|
Vazquez-Munoz R, Arellano-Jimenez MJ, Lopez-Ribot JL. Bismuth nanoparticles obtained by a facile synthesis method exhibit antimicrobial activity against Staphylococcus aureus and Candida albicans. BMC Biomed Eng 2020; 2:11. [PMID: 33073175 PMCID: PMC7558697 DOI: 10.1186/s42490-020-00044-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/08/2020] [Indexed: 01/08/2023] Open
Abstract
Background Bismuth compounds are known for their activity against multiple microorganisms; yet, the antibiotic properties of bismuth nanoparticles (BiNPs) remain poorly explored. The objective of this work is to further the research of BiNPs for nanomedicine-related applications. Stable Polyvinylpyrrolidone (PVP)-coated BiNPs were produced by a chemical reduction process, in less than 30 min. Results We produced stable, small, spheroid PVP-coated BiNPs with a crystalline organization. The PVP-BiNPs showed potent antibacterial activity against the pathogenic bacterium Staphylococcus aureus and antifungal activity against the opportunistic pathogenic yeast Candida albicans, both under planktonic and biofilm growing conditions. Conclusions Our results indicate that BiNPs represent promising antimicrobial nanomaterials, and this facile synthetic method may allow for further investigation of their activity against a variety of pathogenic microorganisms.
Collapse
|
80
|
Roisin L, Melloul E, Woerther PL, Royer G, Decousser JW, Guillot J, Dannaoui E, Botterel F. Modulated Response of Aspergillus fumigatus and Stenotrophomonas maltophilia to Antimicrobial Agents in Polymicrobial Biofilm. Front Cell Infect Microbiol 2020; 10:574028. [PMID: 33123497 PMCID: PMC7573239 DOI: 10.3389/fcimb.2020.574028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction: The complexity of biofilms constitutes a therapeutic challenge and the antimicrobial susceptibility of fungal-bacterial biofilms remains poorly studied. The filamentous fungus Aspergillus fumigatus (Af) and the Gram-negative bacillus Stenotrophomonas maltophilia (Sm) can form biofilms and can be co-isolated from the airways of cystic fibrosis (CF) patients. We previously developed an in vitro biofilm model which highlighted the antibiosis effect of Sm on Af, which was dependent on the bacterial fitness. The aim of the present study was to investigate the in vitro susceptibility of Af and Sm in mono- or polymicrobial biofilms to five antimicrobial agents alone and in two-drug combinations. Methods: Af and Sm clinical reference strains and two strains from CF sputa were tested through a planktonic and biofilm approaches. Af, Sm, or Af-Sm susceptibilities to amphotericin B (AMB), itraconazole (ITC), voriconazole (VRC), levofloxacin (LVX), and rifampicin (RFN) were evaluated by conventional planktonic techniques, crystal violet, XTT, qPCR, and viable plate count. Results: Af planktonic cells and biofilms in formation were more susceptible to AMB, ITC, and VRC than Af mature biofilms. Af mature biofilms were susceptible to AMB, but not to ITC and VRC. Based on viable plate count, a lower concentration of LVX and RFN was required to reduce Sm cell numbers on biofilms in formation compared with mature biofilms. The antibiosis effect of Sm on Af growth was more pronounced for the association of CF strains that exhibited a higher fitness than the reference strains. In Af-Sm biofilms, the fungal susceptibility to AMB was increased compared with Af biofilms. In contrast, the bacterial susceptibility to LVX decreased in Af-Sm biofilms and was fungal biomass-dependent. The combination of AMB (64 μg/mL) with LVX or RFN (4 μg/mL) was efficient to impair Af and Sm growth in the polymicrobial biofilm. Conclusion: Sm increased the Af susceptibility to AMB, whereas Af protected Sm from LVX. Interactions between Af and Sm within biofilms modulate susceptibility to antimicrobial agents, opening the way to new antimicrobial strategies in CF patients.
Collapse
Affiliation(s)
- Lolita Roisin
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France
| | - Elise Melloul
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France
| | - Paul-Louis Woerther
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Bactériologie-Hygiène, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Guilhem Royer
- Unité de Bactériologie-Hygiène, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France.,LABGeM, Génomique Métabolique, CEA, Genoscope, Institut François Jacob, Université d'Evry, Université Paris-Saclay, CNRS, Evry, France
| | - Jean-Winoc Decousser
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Bactériologie-Hygiène, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Jacques Guillot
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Parasitologie-Mycologie, Ecole nationale vétérinaire d'Alfort, Maisons-Alfort, France
| | - Eric Dannaoui
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Parasitologie-Mycologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, AP-HP, Université Paris-Descartes, Paris, France
| | - Françoise Botterel
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Parasitologie-Mycologie, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
81
|
Wijesinghe GK, Maia FC, de Oliveira TR, de Feiria SNB, Joia F, Barbosa JP, Boni GC, Sardi JDCO, Rosalen PL, Höfling JF. Effect of Cinnamomum verum leaf essential oil on virulence factors of Candida species and determination of the in-vivo toxicity with Galleria mellonella model. Mem Inst Oswaldo Cruz 2020; 115:e200349. [PMID: 32997002 PMCID: PMC7523505 DOI: 10.1590/0074-02760200349] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/25/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Essential oils (EO) extracted from Cinnamomum verum has been used as an antimicrobial agents for centuries. The effects of C. verum leaf oil against virulence of microorganisms is not well studied yet. OBJECTIVES This study evaluates the effect of C. verum leaf oil against three virulence factors of Candida albicans, C. tropicalis and C. dubliniensis and its in-vivo toxicity. METHODS Chemical composition of EO was determined using gas chromatography-mass spectrometry (GC-MS). Minimum inhibitory concentration (MIC) was determined using clinical and laboratory standards institute (CLSI) M27-A3 broth microdilution. Effect of EO on initial adhesion was quantified using XTT assay after allowing Candida cells to adhere to the polystyrene surface for 2 h. Biofilm formation of Candida in the presence of EO was quantified using XTT viability assay. Efficacy on reduction of germ tube formation was evaluated using standard protocol. Visualisation of biofilm formation and progression under the EO treatment were done using scanning electron microscope (SEM) and Time lapses microscope respectively. In-vivo toxicity of EO was determined using Galleria mellonella larvae. Chlorhexidine digluconate: positive control. RESULTS Eugenol was the main compound of EO. MIC was 1.0 mg/mL. 50% reduction in initial adhesion was achieved by C. albicans, C. tropicalis and C. dubliniensis with 1.0, > 2.0 and 0.34 mg/mL respectively. 0.5 and 1.0 mg/mL significantly inhibit the germ tube formation. MBIC50 for forming biofilms were ≤ 0.35 mg/mL. 1.0 mg/mL prevent biofilm progression of Candida. SEM images exhibited cell wall damages, cellular shrinkages and decreased hyphal formation. No lethal effect was noted with in-vivo experiment model at any concentration tested. CONCLUSION C. verum leaf oil acts against virulence factors of Candida and does not show any toxicity.
Collapse
Affiliation(s)
- Gayan Kanchana Wijesinghe
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Flávia Camila Maia
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Thaís Rossini de Oliveira
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Simone N Busato de Feiria
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Felipe Joia
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Janaina Priscila Barbosa
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Giovana Cláudia Boni
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| | - Janaina de Cássia Orlandi Sardi
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Farmacologia, Anestesiologia e Terapêutica, Departamento de Ciências Fisiológicas, Campinas, SP, Brasil
| | - Pedro Luiz Rosalen
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Farmacologia, Anestesiologia e Terapêutica, Departamento de Ciências Fisiológicas, Campinas, SP, Brasil
| | - José Francisco Höfling
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Área de Microbiologia e Imunologia, Departamento de Diagnóstico Oral, Campinas SP, Brasil
| |
Collapse
|
82
|
Garcia LM, Costa-Orlandi CB, Bila NM, Vaso CO, Gonçalves LNC, Fusco-Almeida AM, Mendes-Giannini MJS. A Two-Way Road: Antagonistic Interaction Between Dual-Species Biofilms Formed by Candida albicans/Candida parapsilosis and Trichophyton rubrum. Front Microbiol 2020; 11:1980. [PMID: 33013741 PMCID: PMC7499241 DOI: 10.3389/fmicb.2020.01980] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Dermatomycoses include superficial fungal infections of the skin and its appendages. Trichophyton rubrum, Candida albicans, and Candida parapsilosis are some of the most prevalent species that cause dermatomycoses. Several studies show a variable predominance of Candida spp. in relation to dermatophytes, especially in onychomycosis and the possibility of isolating both from the same site. The ability of dermatophytes to form biofilms recently been explored and there is currently no evidence on the involvement of these filamentous fungi in multi-species biofilms. Thus, this study aims to investigate the probable dual-species interaction between T. rubrum and C. albicans and T. rubrum and C. parapsilosis biofilms, considering variable formation conditions, as well as the susceptibility of these dual-species biofilms against terbinafine and efinaconazole. Three conditions of formation of dual-species biofilms were tested: (a) the suspensions of T. rubrum and Candida albicans or C. parapsilosis placed together; (b) suspensions of C. albicans and C. parapsilosis added the pre-adhesion of T. rubrum biofilms; (c) after the maturation of T. rubrum sessile cells. In the first and second conditions, the quantification of metabolic activities, biomass, and polysaccharide materials of mixed biofilms tended to resemble Candida monospecies biofilms. In the third condition, the profiles were modified after the addition of Candida, suggesting that T. rubrum biofilms served as substrate for the development of Candida biofilms. Scanning electron microscopy showed Candida predominance, however, numerous blastoconidia were noted, most evident in the conditions under which Candida was added after the pre-adhesion and maturation of T. rubrum biofilms. Despite the predominance of Candida, the presence of T. rubrum appears to inhibit C. albicans filamentation and C. parapsilosis development, confirming an antagonistic interaction. Fungal burden assays performed when the biofilms were formed together confirmed Candida predominance, as well as susceptibility to antifungals. Further studies will be needed to identify the components of the Candida and T. rubrum biofilm supernatants responsible for inhibiting dermatophyte growth and C. albicans filamentation.
Collapse
Affiliation(s)
- Letícia Morais Garcia
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Caroline Barcelos Costa-Orlandi
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Níura Madalena Bila
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,School of Veterinary, Eduardo Mondlane University, Maputo, Mozambique
| | - Carolina Orlando Vaso
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Larissa Naiara Carvalho Gonçalves
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Ana Marisa Fusco-Almeida
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Maria José Soares Mendes-Giannini
- Clinical Mycology Laboratory, Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
83
|
Guo L, Zhang L, Yang Q, Xu B, Fu X, Liu M, Li Z, Zhang S, Xie Z. Antibacterial and Cytotoxic Bridged and Ring Cleavage Angucyclinones From a Marine Streptomyces sp. Front Chem 2020; 8:586. [PMID: 32850626 PMCID: PMC7417440 DOI: 10.3389/fchem.2020.00586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/08/2020] [Indexed: 11/21/2022] Open
Abstract
Chemical investigation of a marine-derived Streptomyces sp. KCB-132, cultivated in liquid ISP2 medium, had led to the discovery of three C-ring cleavage angucyclinone N-heterocycles, pratensilins A–C, with a novel spiro indolinone-naphthofuran skeleton. Addition of 50 μM LaCl3 to the same medium and subsequent chemical analysis of this strain returned a new member of this rare class, pratensilin D (1), along with two new angucyclinone derivatives, featuring ether-bridged (2) and A-ring cleavage (3) structural properties. Their structures and absolute configurations were assigned by spectroscopic analysis, single-crystal X-ray diffractions, and equivalent circulating density (ECD) calculations. (+)- and (–)-1, a pair of enantiomeric nitrogen-containing angucyclinones, exhibited different strengths of antibacterial and cytotoxic activities.
Collapse
Affiliation(s)
- Lin Guo
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Lu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Qiaoli Yang
- College of Life Sciences, Yantai University, Yantai, China
| | - Bo Xu
- College of Life Sciences, Yantai University, Yantai, China
| | - Xinzhen Fu
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Ming Liu
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhi Li
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Shumin Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zeping Xie
- School of Pharmacy, Binzhou Medical University, Yantai, China
| |
Collapse
|
84
|
Andrade G, Orlando HCS, Scorzoni L, Pedroso RS, Abrão F, Carvalho MTM, Veneziani RCS, Ambrósio SR, Bastos JK, Mendes-Giannini MJS, Martins CHG, Pires RH. Brazilian Copaifera Species: Antifungal Activity against Clinically Relevant Candida Species, Cellular Target, and In Vivo Toxicity. J Fungi (Basel) 2020; 6:jof6030153. [PMID: 32872100 PMCID: PMC7560146 DOI: 10.3390/jof6030153] [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] [Received: 07/30/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Plants belonging to the genus Copaifera are widely used in Brazil due to their antimicrobial properties, among others. The re-emergence of classic fungal diseases as a consequence of antifungal resistance to available drugs has stimulated the search for plant-based compounds with antifungal activity, especially against Candida. The Candida-infected Caenorhabditis elegans model was used to evaluate the in vitro antifungal potential of Copaifera leaf extracts and trunk oleoresins against Candida species. The Copaifera leaf extracts exhibited good antifungal activity against all Candida species, with MIC values ranging from 5.86 to 93.75 µg/mL. Both the Copaifera paupera and Copaifera reticulata leaf extracts at 46.87 µg/mL inhibited Candida glabrata biofilm formation and showed no toxicity to C. elegans. The survival of C. glabrata-infected nematodes increased at all the tested extract concentrations. Exposure to Copaifera leaf extracts markedly increased C. glabrata cell vacuolization and cell membrane damage. Therefore, Copaifera leaf extracts are potential candidates for the development of new and safe antifungal agents.
Collapse
Affiliation(s)
- Géssica Andrade
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
| | - Haniel Chadwick Silva Orlando
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
| | - Liliana Scorzoni
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14801-902, Brazil; (L.S.); (M.J.S.M.-G.)
- Science and Technology Institute of São José dos Campos (ICT), São Paulo State University (UNESP), São José dos Campos 12245-000, Brazil
| | - Reginaldo Santos Pedroso
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
- Health Technical School (ESTES), Federal University of Uberlandia, Uberlandia 38400-732, Brazil
| | - Fariza Abrão
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
| | - Marco Túlio Menezes Carvalho
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
| | - Rodrigo Cassio Sola Veneziani
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
| | - Sérgio Ricardo Ambrósio
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
| | - Jairo Kenupp Bastos
- Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirão Preto 14040-903, Brazil;
| | | | - Carlos Henrique Gomes Martins
- Institute of Biomedical Sciences (ICBIM), Federal University of Uberlandia, Uberlandia 38400-902, Brazil
- Correspondence: (C.H.G.M.); (R.H.P.); Tel.: +55-(34)-3225-8670 (C.H.G.M.); +55-(16)-3711-8945 (R.H.P.)
| | - Regina Helena Pires
- University of Franca, Franca 14404-600, Brazil; (G.A.); (H.C.S.O.); (R.S.P.); (F.A.); (M.T.M.C.); (R.C.S.V.); (S.R.A.)
- Correspondence: (C.H.G.M.); (R.H.P.); Tel.: +55-(34)-3225-8670 (C.H.G.M.); +55-(16)-3711-8945 (R.H.P.)
| |
Collapse
|
85
|
Costa ACBP, Back-Brito GN, Mayer FL, Hube B, Wilson D. Candida albicans Mrv8, is involved in epithelial damage and biofilm formation. FEMS Yeast Res 2020; 20:5862582. [PMID: 32584995 PMCID: PMC7343537 DOI: 10.1093/femsyr/foaa033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/24/2020] [Indexed: 12/25/2022] Open
Abstract
Candida albicans is the most common human fungal pathogen that can cause superficial and deep-seated infections in susceptible individuals. Despite its medical importance, the vast majority of C. albicans genes remain of unknown function. Here, we report a role for the lineage-specific gene, MRV8, in host pathogen interactions, mycelial microcolony maturation and biofilm formation. In silico analysis indicated that MRV8 encodes a four-pass transmembrane protein unique to the closely related pathogens C. albicans and Candida dubliniensis. Deletion of MRV8 did not affect C. albicans adherence to, or initial invasion into human oral epithelia, but inhibited mycelial development and strongly reduced epithelial damage. mrv8Δ/Δ cells exhibited a media-dependent defect in biofilm formation and mutant biofilm metabolic activity was enhanced by cyclosporin A. mrv8Δ/Δ biofilms were more tolerant to treatment with caspofungin, but not to fluconazole or amphotericin B. Co-stimulation with calcium chloride and calcofluor white rescued biofilm growth in the presence of caspofungin, and this rescue-effect was Mrv8-dependent. Together, our data demonstrate an important role for a lineage-specific gene (MRV8) in C. albicans biofilm formation, drug tolerance and host–pathogen interactions.
Collapse
Affiliation(s)
- Anna Carolina Borges Pereira Costa
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology (ICT); São José dos Campos, Brazil.,Department of Microbial Pathogenicity Mechanisms, Hans-Knoell-Institute, Jena, Germany.,Aberdeen Fungal Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Aberdeen, United Kingdom
| | - Graziella Nuernberg Back-Brito
- Department of Biosciences and Oral Diagnosis, São Paulo State University (Unesp), Institute of Science and Technology (ICT); São José dos Campos, Brazil
| | - François L Mayer
- Department of Microbial Pathogenicity Mechanisms, Hans-Knoell-Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans-Knoell-Institute, Jena, Germany.,Friedrich Schiller University, Jena, Germany
| | - Duncan Wilson
- Department of Microbial Pathogenicity Mechanisms, Hans-Knoell-Institute, Jena, Germany.,Aberdeen Fungal Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Aberdeen, United Kingdom.,Medical Research Council Centre for Medical Mycology, School of Biosciences, University of Exeter, Stocker Rd, Exeter EX4 4QD, Exeter, United Kingdom
| |
Collapse
|
86
|
Vazquez-Munoz R, Lopez FD, Lopez-Ribot JL. Bismuth Nanoantibiotics Display Anticandidal Activity and Disrupt the Biofilm and Cell Morphology of the Emergent Pathogenic Yeast Candida auris. Antibiotics (Basel) 2020; 9:E461. [PMID: 32751405 PMCID: PMC7460268 DOI: 10.3390/antibiotics9080461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 01/08/2023] Open
Abstract
Candida auris is an emergent multidrug-resistant pathogenic yeast, which forms biofilms resistant to antifungals, sanitizing procedures, and harsh environmental conditions. Antimicrobial nanomaterials represent an alternative to reduce the spread of pathogens-including yeasts-regardless of their drug-resistant profile. Here we have assessed the antimicrobial activity of easy-to-synthesize bismuth nanoparticles (BiNPs) against the emergent multidrug-resistant yeast Candida auris, under both planktonic and biofilm growing conditions. Additionally, we have examined the effect of these BiNPs on cell morphology and biofilm structure. Under planktonic conditions, BiNPs MIC values ranged from 1 to 4 µg mL-1 against multiple C. auris strains tested, including representatives of all different clades. Regarding the inhibition of biofilm formation, the calculated BiNPs IC50 values ranged from 5.1 to 113.1 µg mL-1. Scanning electron microscopy (SEM) observations indicated that BiNPs disrupted the C. auris cell morphology and the structure of the biofilms. In conclusion, BiNPs displayed strong antifungal activity against all strains of C. auris under planktonic conditions, but moderate activity against biofilm growth. BiNPs may potentially contribute to reducing the spread of C. auris strains at healthcare facilities, as sanitizers and future potential treatments. More research on the antimicrobial activity of BiNPs is warranted.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of Biology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Fernando D. Lopez
- School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Jose L. Lopez-Ribot
- Department of Biology, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
87
|
Vazquez-Munoz R, Lopez FD, Lopez-Ribot JL. Silver Nanoantibiotics Display Strong Antifungal Activity Against the Emergent Multidrug-Resistant Yeast Candida auris Under Both Planktonic and Biofilm Growing Conditions. Front Microbiol 2020; 11:1673. [PMID: 32849347 PMCID: PMC7399222 DOI: 10.3389/fmicb.2020.01673] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022] Open
Abstract
Candida auris is an emergent multidrug-resistant pathogenic yeast with an unprecedented ability for a fungal organism to easily spread between patients in clinical settings, leading to major outbreaks in healthcare facilities. The formation of biofilms by C. auris contributes to infection and its environmental persistence. Most antifungals and sanitizing procedures are not effective against C. auris, but antimicrobial nanomaterials could represent a viable alternative to combat the infections caused by this emerging pathogen. We have previously described an easy and inexpensive method to synthesize silver nanoparticles (AgNPs) in non-specialized laboratories. Here, we have assessed the antimicrobial activity of the resulting AgNPs on C. auris planktonic and biofilm growth phases. AgNPs displayed a strong antimicrobial activity against all the stages of all C. auris strains tested, representative of four different clades. Under planktonic conditions, minimal inhibitory concentration (MIC) values of AgNPs against the different strains were <0.5 μg ml−1; whereas calculated IC50 values for inhibition of biofilms formation were <2 μg ml−1 for all, but one of the C. auris strains tested. AgNPs were also active against preformed biofilms formed by all different C. auris strains, with IC50 values ranging from 1.2 to 6.2 μg ml−1. Overall, our results indicate potent activity of AgNPs against strains of C. auris, both under planktonic and biofilm growing conditions, and indicate that AgNPs may contribute to the control of infections caused by this emerging nosocomial threat.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Fernando D Lopez
- Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Jose L Lopez-Ribot
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
88
|
Muñoz JE, Ramirez LM, Dias LDS, Rivas LA, Ramos LS, Santos ALS, Taborda CP, Parra-Giraldo CM. Pathogenicity Levels of Colombian Strains of Candida auris and Brazilian Strains of Candida haemulonii Species Complex in Both Murine and Galleria mellonella Experimental Models. J Fungi (Basel) 2020; 6:jof6030104. [PMID: 32664191 PMCID: PMC7558079 DOI: 10.3390/jof6030104] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/04/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
Candida auris and Candida haemulonii complex (C. haemulonii, C. haemulonii var. vulnera and C. duobushaemulonii) are phylogenetically related species that share some physiological features and habits. In the present study, we compared the virulence of these yeast species using two different experimental models: (i) Galleria mellonella larvae to evaluate the survival rate, fungal burden, histopathology and phagocytosis index and (ii) BALB/c mice to evaluate the survival. In addition, the fungal capacity to form biofilm over an inert surface was analyzed. Our results showed that in both experimental models, the animal survival rate was lower when infected with C. auris strains than the C. haemulonii species complex. The hemocytes of G. mellonella showed a significantly reduced ability to phagocytize the most virulent strains forming the C. haemulonii species complex. Interestingly, for C. auris, it was impossible to measure the phagocytosis index due to a general lysis of the hemocytes. Moreover, it was observed a greater capability of biofilm formation by C. auris compared to C. haemulonii species complex. In conclusion, we observed that C. auris and C. haemulonii complex have different levels of pathogenicity in the experimental models employed in the present study.
Collapse
Affiliation(s)
- Julián E. Muñoz
- MICROS Group, Medicine Traslacional Institute, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, D.C. 111221, Colombia;
| | - Laura M. Ramirez
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C. 110231, Colombia; (L.M.R.); (L.A.R.)
| | - Lucas dos Santos Dias
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo, SP 05508-060, Brazil; (L.d.S.D.); (C.P.T.)
| | - Laura A. Rivas
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C. 110231, Colombia; (L.M.R.); (L.A.R.)
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.S.R.); (A.L.S.S.)
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.S.R.); (A.L.S.S.)
| | - Carlos P. Taborda
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo, SP 05508-060, Brazil; (L.d.S.D.); (C.P.T.)
- Laboratory of Medical Mycology-LIM53/IMTSP, University of São Paulo (USP), São Paulo, SP 05508-060, Brazil
| | - Claudia M. Parra-Giraldo
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C. 110231, Colombia; (L.M.R.); (L.A.R.)
- Correspondence: ; Tel.: +57-1-3208320 (ext. 4305)
| |
Collapse
|
89
|
Chermebilaenes A and B, New Bioactive Meroterpenoids from Co-Cultures of Marine-Derived Isolates of Penicillium bilaiae MA-267 and Penicillium chermesinum EN-480. Mar Drugs 2020; 18:md18070339. [PMID: 32605151 PMCID: PMC7401264 DOI: 10.3390/md18070339] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
The co-cultivation of two or more different microbial strains in one culture vessel was supposed to be a viable experimental approach for enhancing the diversity of the compounds produced. Two new meroterpenoid derivatives, chermebilaenes A (1) and B (2), together with three known sesquiterpenoids, sesquicaranoic acid B (3), cyclonerodiol (4) and bisabol-l-on-13-säuremethylester (5), were characterized from a co-culture of the marine-derived fungal isolates of Penicillium bilaiae MA-267 and Penicillium chermesinum EN-480. Neither fungus produced these compounds when cultured alone under the same conditions. Compound 1 represents an unprecedented acorane-type sesquiterpene hybridized with an octadecadienoic acid skeleton. The structures were elucidated on the basis of spectroscopic analysis, and the absolute configurations were assumed on the basis of acidic hydrolysis combined with modified Mosher’s method and electronic circular dichroism (ECD) calculations. Compound 1 showed potent inhibitory activities against Ceratobasidium cornigerum and Edwardsiella tarda.
Collapse
|
90
|
Almeida Furquim de Camargo B, Soares Silva DE, Noronha da Silva A, Campos DL, Machado Ribeiro TR, Mieli MJ, Borges Teixeira Zanatta M, Bento da Silva P, Pavan FR, Gallina Moreira C, Resende FA, Menegário AA, Chorilli M, Vieira de Godoy Netto A, Bauab TM. New Silver(I) Coordination Compound Loaded into Polymeric Nanoparticles as a Strategy to Improve In Vitro Anti-Helicobacter pylori Activity. Mol Pharm 2020; 17:2287-2298. [DOI: 10.1021/acs.molpharmaceut.9b01264] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | | | - Anderson Noronha da Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| | - Débora Leite Campos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| | | | - Maria Júlia Mieli
- Department of Biological Sciences and Health, University of Araraquara, Araraquara 14801-340, São Paulo, Brazil
| | | | - Patrícia Bento da Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| | - Fernando Rogerio Pavan
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| | - Cristiano Gallina Moreira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| | - Flávia Aparecida Resende
- Department of Biological Sciences and Health, University of Araraquara, Araraquara 14801-340, São Paulo, Brazil
| | - Amauri Antônio Menegário
- Environmental Studies Center, São Paulo State University (UNESP), Rio Claro 13506-900, São Paulo, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| | | | - Taís Maria Bauab
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, São Paulo, Brazil
| |
Collapse
|
91
|
Snyder SS, Gleaton JW, Kirui D, Chen W, Millenbaugh NJ. Antifungal Activity of Synthetic Scorpion Venom-Derived Peptide Analogues Against Candida albicans. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10084-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AbstractFungal infections are becoming a serious problem due to their high morbidity and mortality combined with the rise in drug resistance and dearth of new antimycotic drugs. The scorpion venom-derived peptide BmKn2, and its synthetic analogue Kn2–7, were previously observed to have antibacterial activity. These peptides and their d-amino acid analogues (dBmKn2 and dKn2–7) were tested for antifungal activity against drug resistant and clinical isolates of Candida albicans. In planktonic susceptibility studies, dKn2–7 had greater activity than the other three peptides against 6 out of 7 fungal strains, with no apparent correlation between drug resistance and minimum fungicidal concentrations (MFCs). Time kill experiments demonstrated that the fungicidal activity of dKn2–7 began within the first hour and killing rates were dose dependent at ≥ 1 × MFC. Against biofilms, the d-analogues were the most effective, while the l-analogues had low efficacy in most strains even at 10 times the planktonic MFC. Stability testing suggests that this increased efficacy of the d-analogues may be due to increased resistance to protease degradation compared to the l-analogues. Peptides were also assessed for mammalian cell toxicity. BmKn2 and dBmKn2 induced significant hemolysis at levels similar to their MFCs, whereas Kn2–7 and dKn2–7 caused hemolysis at 4–16 times their MFCs. The 50% inhibitory concentration (IC50) for dKn2–7 against murine fibroblasts was greater than or equal to the planktonic MFCs and biofilm IC50s for dKn2–7 in all C. albicans strains tested. These results support the potential for dKn2–7 to be further investigated as a novel antifungal therapeutic.
Collapse
|
92
|
Costa-Orlandi CB, Serafim-Pinto A, da Silva PB, Bila NM, Bonatti JLDC, Scorzoni L, Singulani JDL, Dos Santos CT, Nazaré AC, Chorilli M, Regasini LO, Fusco-Almeida AM, Mendes-Giannini MJS. Incorporation of Nonyl 3,4-Dihydroxybenzoate Into Nanostructured Lipid Systems: Effective Alternative for Maintaining Anti-Dermatophytic and Antibiofilm Activities and Reducing Toxicity at High Concentrations. Front Microbiol 2020; 11:1154. [PMID: 32582096 PMCID: PMC7290161 DOI: 10.3389/fmicb.2020.01154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Dermatophytosis is the most common mycosis worldwide, affecting approximately 20 to 25% of the population, regardless of gender, race, color, and age. Most antifungal agents used for the treatment of dermatophytosis belong to the azole and allylamine classes. Dermatophytes are reported to be resistant to most commercial drugs, especially microbial biofilms, in addition to their considerable toxicity. It should be emphasized the importance of looking for new molecules with reduced toxicity, as well as new targets and mechanisms of action. This work aims to incorporate nonyl 3,4-dihydroxybenzoate, a potent fungicide compound against planktonic cells and dermatophyte biofilms in nanostructured lipid systems (NLS), in order to reduce toxicity in high concentrations, improve its solubility and maintain its effectiveness. The compound was incorporated into NLS constituted by cholesterol, mixture of polyoxyethylene (23) lauryl ether (Brij®98) and soybean phosphatidylcholine (Epikuron® 200)], 2: 1 ratio and PBS (phosphate-buffered saline). The characterization of the incorporation was performed. Susceptibility tests were conducted according to document M38-A2 by CLSI (2008). The toxicity of the NLS compound was evaluated in HaCaT cell lines by the sulforhodamine B method and in alternative models Caenorhabditis elegans and zebrafish. Finally, its efficacy was evaluated against the mature Trichophyton rubrum and Trichophyton mentagrophytes biofilms. NLS and nonyl 3,4-dihydroxybenzoate loaded into NLS displayed sizes ranging from 137.8 ± 1.815 to 167.9 ± 4.070 nm; the polydispersity index (PDI) varying from 0.331 ± 0.020 to 0.377 ± 0.004 and zeta potential ranging from −1.46 ± 0.157 to −4.63 ± 0.398 mV, respectively. Polarized light microscopy results confirmed the formation of NLS of the microemulsion type. Nonyl incorporated into NLS showed minimum inhibitory concentration (MIC) values, ranging from 2 to 15.6 mg/L. The toxicity tests presented cell viability higher than 80% in all tested concentrations, as well as, a significantly increased of the survival of Caenorhabditis elegans and zebrafish models. Anti-biofilm tests proved the efficacy of the incorporation. These findings contribute significantly to the search for new antifungals and allow the systemic administration of the compound, since the incorporation can increase the solubility of non-polar compounds, improve bioavailability, effectiveness and reduce toxicity.
Collapse
Affiliation(s)
- Caroline Barcelos Costa-Orlandi
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Aline Serafim-Pinto
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Patrícia Bento da Silva
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Níura Madalena Bila
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil.,Universidade Eduardo Mondlane, School of Veterinary, Maputo, Mozambique
| | - Jean Lucas de Carvalho Bonatti
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Liliana Scorzoni
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Junya de Lacorte Singulani
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Claudia Tavares Dos Santos
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Ana Carolina Nazaré
- Institute of Biosciences, Humanities and Exact Sciences, Department of Chemistry and Environmental Sciences, Universidade Estadual Paulista (UNESP), São José do Rio Preto, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Luis Octávio Regasini
- Institute of Biosciences, Humanities and Exact Sciences, Department of Chemistry and Environmental Sciences, Universidade Estadual Paulista (UNESP), São José do Rio Preto, Brazil
| | - Ana Marisa Fusco-Almeida
- School of Pharmaceutical Sciences, Department of Clinical Analysis, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | | |
Collapse
|
93
|
Bio-Mediated Synthesis of Reduced Graphene Oxide Nanoparticles from Chenopodium album: Their Antimicrobial and Anticancer Activities. NANOMATERIALS 2020; 10:nano10061096. [PMID: 32492878 PMCID: PMC7353263 DOI: 10.3390/nano10061096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023]
Abstract
A novel method of preparing reduced graphene oxide (RGOX) from graphene oxide (GOX) was developed employing vegetable extract, Chenopodium album, as a reducing and stabilizing agent. Chenopodium album is a green leafy vegetable with a low shelf life, fresh leaves of this vegetable are encouraged to be used due to high water content. The previously modified ‘Hummers method’ has been in practice for the preparation of GOX by using precursor graphite powder. In this study, green synthesis of RGOX was functionally verified by employing FTIR and UV-visible spectroscopy, along with SEM and TEM. Our results demonstrated typical morphology of RGOX stacked in layers that appeared as silky, transparent, and rippled. The antibacterial activity was shown by analyzing minimal inhibitory concentration values, agar diffusion assay, fluorescence techniques. It showed enhanced antibacterial activity against Gram-positive and Gram-negative bacteria in comparison to GOX. It has also been shown that the synthesized compound exhibited enhanced antibiofilm activity as compared to its parent compound. The efficacy of RGOX and GOX has been demonstrated on a human breast cancer cell line, which suggested RGOX as a potential anticancer agent.
Collapse
|
94
|
Nagy F, Vitális E, Jakab Á, Borman AM, Forgács L, Tóth Z, Majoros L, Kovács R. In vitro and in vivo Effect of Exogenous Farnesol Exposure Against Candida auris. Front Microbiol 2020; 11:957. [PMID: 32508780 PMCID: PMC7251031 DOI: 10.3389/fmicb.2020.00957] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
The spreading of multidrug-resistant Candida auris is considered as an emerging global health threat. The number of effective therapeutic regimens is strongly limited; therefore, development of novel strategies is needed. Farnesol is a quorum-sensing molecule with a potential antifungal and/or adjuvant effect; it may be a promising candidate in alternative treatment against Candida species including C. auris. To examine the effect of farnesol on C. auris, we performed experiments focusing on growth, biofilm production ability, production of enzymes related to oxidative stress, triazole susceptibility and virulence. Concentrations ranging from 100 to 300 μM farnesol caused a significant growth inhibition against C. auris planktonic cells for 24 h (p < 0.01-0.05). Farnesol treatment showed a concentration dependent inhibition in terms of biofilm forming ability of C. auris; however, it did not inhibit significantly the biofilm development at 24 h. Nevertheless, the metabolic activity of adhered farnesol pre-exposed cells (75 μM) was significantly diminished at 24 h depending on farnesol treatment during biofilm formation (p < 0.001-0.05). Moreover, 300 μM farnesol exerted a marked decrease in metabolic activity against one-day-old biofilms between 2 and 24 h (p < 0.001). Farnesol increased the production of reactive species remarkably, as revealed by 2',7'-dichlorofluorescein (DCF) assay {3.96 ± 0.89 [nmol DCF (OD640)-1] and 23.54 ± 4.51 [nmol DCF (OD640)-1] for untreated cells and farnesol exposed cells, respectively; p < 0.001}. This was in line with increased superoxide dismutase level {85.69 ± 5.42 [munit (mg protein)-1] and 170.11 ± 17.37 [munit (mg protein)-1] for untreated cells and farnesol exposed cells, respectively; p < 0.001}, but the catalase level remained statistically comparable between treated and untreated cells (p > 0.05). Concerning virulence-related enzymes, exposure to 75 μM farnesol did not influence phospholipase or aspartic proteinase activity (p > 0.05). The interaction between fluconazole, itraconazole, voriconazole, posaconazole, isavuconazole and farnesol showed clear synergism (FICI ranges from 0.038 to 0.375) against one-day-old biofilms. Regarding in vivo experiments, daily 75 μM farnesol treatment decreased the fungal burden in an immunocompromised murine model of disseminated candidiasis, especially in case of inocula pre-exposed to farnesol (p < 0.01). In summary, farnesol shows a promising therapeutic or adjuvant potential in traditional or alternative therapies such as catheter lock therapy.
Collapse
Affiliation(s)
- Fruzsina Nagy
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Eszter Vitális
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary.,Hospital Hygiene Ward, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - Ágnes Jakab
- Department of Molecular Biotechnology and Microbiology, Faculty of Science and Technology, Institute of Biotechnology, University of Debrecen, Debrecen, Hungary
| | - Andrew M Borman
- UK National Mycology Reference Laboratory, Public Health England, Bristol, United Kingdom
| | - Lajos Forgács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Zoltán Tóth
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - László Majoros
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Renátó Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
95
|
Wu Y, Wu M, Gao J, Ying C. Antifungal Activity and Mode of Action of Miltefosine Against Clinical Isolates of Candida krusei. Front Microbiol 2020; 11:854. [PMID: 32508766 PMCID: PMC7248313 DOI: 10.3389/fmicb.2020.00854] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Candida krusei attracts attention from medical professionals mainly for its intrinsic resistance to fluconazole and the limited number of drugs available to treat C. krusei vulvovaginal candidiasis. Miltefosine was demonstrated to have good antifungal activity both in vitro and in vivo. Here, we determined the susceptibility profiles of 57 clinical C. krusei isolates from vulvovaginal candidiasis patients and assessed the antifungal activity of miltefosine against C. krusei. All isolates were susceptible to voriconazole and itraconazole, whereas 1.8% of the isolates were of non-wild-type phenotype to amphotericin B. In contrast, miltefosine showed low MICs against all C. krusei isolates with fungicidal activity. The checkerboard assay showed that the synergistic effect of miltefosine in combination with amphotericin B was observed in 25% of the tested planktonic C. krusei isolates and 18.8% of the tested preformed biofilms, whereas miltefosine in combination with fluconazole showed indifferent interaction for all tested planktonic isolates. The presence of sorbitol in the broth microdilution assay did not influence the MIC values of miltefosine against C. krusei, but the presence of ergosterol increased the MIC values. Visible changes in cell content in cells treated with miltefosine were observed. We found that cells treated with miltefosine showed decreased cell viability and chromatin condensation under PI staining, which indicates that miltefosine may induce apoptosis-like cell death in C. krusei. In conclusion, we found miltefosine has a good activity against C. krusei isolates and exerts its fungicidal effect by binding to ergosterol in the cell membrane and inducing apoptosis.
Collapse
Affiliation(s)
- Yongqin Wu
- Department of Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Mengying Wu
- Department of Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jing Gao
- Department of Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chunmei Ying
- Department of Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
96
|
Lara HH, Ixtepan-Turrent L, Yacaman MJ, Lopez-Ribot J. Inhibition of Candida auris Biofilm Formation on Medical and Environmental Surfaces by Silver Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21183-21191. [PMID: 31944650 PMCID: PMC8243355 DOI: 10.1021/acsami.9b20708] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Candida auris is an emerging pathogenic fungus implicated in healthcare-associated outbreaks and causes bloodstream infections associated with high mortality rates. Biofilm formation represents one of the major pathogenetic traits associated with this microorganism. Unlike most other Candida species, C. auris has the ability to survive for weeks on different surfaces. Therefore, there is an urgent need to develop new effective control strategies to combat the threat of C. auris. Advances in nanotechnologies have emerged that carry significant potential impact against Candida biofilms. We obtained pure round silver nanoparticles (AgNPs) (1 to 3 nm in diameter) using a microwave-assisted synthetic approach. When tested against C. auris, our results indicated a potent inhibitory activity both on biofilm formation (half maximal inhibitory concentration (IC50) of 0.06 ppm) and against preformed biofilms (IC50 of 0.48 ppm). Scanning electron microscopy images of AgNP-treated biofilms showed cell wall damage mostly by disruption and distortion of the outer surface of the fungal cell wall. In subsequent experiments AgNPs were used to functionalize medical and environmental surfaces. Silicone elastomers functionalized with AgNPs demonstrated biofilm inhibition (>50%) at relatively low concentrations (2.3 to 0.28 ppm). Bandage dressings loaded with AgNPs inhibited growth of C. auris biofilms by more than 80% (2.3 to 0.017 ppm). Also, to demonstrate long-lasting protection, dressings loaded with AgNPs (0.036 ppm) were washed thoroughly with phosphate-buffered saline, maintaining protection against the C. auris growth from cycles 1 to 3 (>80% inhibition) and from cycles 4 to 6 (>50% inhibition). Our results demonstrate the dose-dependent activity of AgNPs against biofilms formed by C. auris on both medical (silicone elastomer) and environmental (bandage fibers) surfaces. The AgNPs-functionalized fibers retain the fungicidal effect even after repeated thorough washes. Overall these results point to the utility of silver nanoparticles to prevent and control infections caused by this emerging pathogenic fungus.
Collapse
Affiliation(s)
- Humberto H. Lara
- Department of Biology and South Texas Center for Emerging Infectious Diseases
| | - Liliana Ixtepan-Turrent
- Departamento de Ciencias Basicas, Division de Ciencia de la Salud, Universidad de Monterrey, San Pedro Garza García, Nuevo León 66238, México
| | - Miguel Jose Yacaman
- Department of Applied Physics and Materials Science, Northern Arizona University, 700 South Osborne Drive, Flagstaff, Arizona 86011, United States
| | - Jose Lopez-Ribot
- Department of Biology and South Texas Center for Emerging Infectious Diseases
| |
Collapse
|
97
|
Vitzilaiou E, Aunsbjerg SD, Mahyudin NA, Knøchel S. Stress Tolerance of Yeasts Dominating Reverse Osmosis Membranes for Whey Water Treatment. Front Microbiol 2020; 11:816. [PMID: 32431679 PMCID: PMC7214788 DOI: 10.3389/fmicb.2020.00816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/06/2020] [Indexed: 11/13/2022] Open
Abstract
Filamentous yeast species belonging to the closely related Saprochaete clavata and Magnusiomyces spicifer were recently found to dominate biofilm communities on the retentate and permeate surface of Reverse Osmosis (RO) membranes used in a whey water treatment system after CIP (Cleaning-In-Place). Microscopy revealed that the two filamentous yeast species can cover extensive areas due to their large cell size and long hyphae formation. Representative strains from these species were here further characterized and displayed similar physiological and biochemical characteristics. Both strains tested were able to grow in twice RO-filtrated permeate water and metabolize the urea present. Little is known about the survival characteristics of these strains. Here, their tolerance toward heat (60, 70, and 80°C) and Ultraviolet light (UV-C) treatment at 255 nm using UV-LED was assessed as well as their ability to form biofilm and withstand cleaning associated stress. According to the heat tolerance experiments, the D60°C of S. clavata and M. spicifer is 16.37 min and 7.24 min, respectively, while a reduction of 3.5 to >4.5 log (CFU/mL) was ensured within 5 min at 70°C. UV-C light at a dose level 10 mJ/cm2 had little effect, while doses of 40 mJ/cm2 and upward ensured a ≥4log reduction in a static laboratory scale set-up. The biofilm forming potential of one filamentous yeast and one budding yeast, Sporopachydermia lactativora, both isolated from the same biofilm, was compared in assays employing flat-bottomed polystyrene microwells and peg lids, respectively. In these systems, employing both nutrient rich as well as nutrient poor media, only the filamentous yeast was able to create biofilm. However, on RO membrane coupons in static systems, both the budding yeast and a filamentous yeast were capable of forming single strain biofilms and when these coupons were exposed to different simulations of CIP treatments both the filamentous and budding yeast survived these. The dominance of these yeasts in some filter systems tested, their capacity to adhere and their tolerance toward relevant stresses as demonstrated here, suggest that these slow growing yeasts are well suited to initiate microbial biofouling on surfaces in low nutrient environments.
Collapse
Affiliation(s)
- Eirini Vitzilaiou
- Laboratory of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Stina D. Aunsbjerg
- Laboratory of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - N. A. Mahyudin
- Department of Food Service and Management, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Susanne Knøchel
- Laboratory of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
98
|
Reddy GKK, Nancharaiah YV. Alkylimidazolium Ionic Liquids as Antifungal Alternatives: Antibiofilm Activity Against Candida albicans and Underlying Mechanism of Action. Front Microbiol 2020; 11:730. [PMID: 32373105 PMCID: PMC7186398 DOI: 10.3389/fmicb.2020.00730] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/27/2020] [Indexed: 01/12/2023] Open
Abstract
Candida albicans is an opportunistic pathogen causes fungal infections that range from common skin infections to persistent infections through biofilm formation on tissues, implants and life threatening systemic infections. New antifungal agents or therapeutic methods are desired due to high incidence of infections and emergence of drug-resistant strains. The present study aimed to evaluate (i) the antifungal and antibiofilm activity of 1-alklyl-3-methyl imidazolium ionic liquids ([CnMIM]+[X]-, n = 4, 12 and 16) against Candida albicans ATCC 10231 and two clinical C. albicans strains and (ii) the mechanism of action of promising antifungal ionic liquid on C. albicans. Two of the tested compounds were identified as more effective in preventing growth and biofilm formation. These ionic liquid compounds with -dodecyl and -hexadecyl alkyl groups effectively prevented biofilm formation by fluconazole resistant C. albicans 10231 and two other clinical C. albicans strains. Although both the compounds caused viability loss in mature C. albicans biofilms, an ionic liquid with -hexadecyl group ([C16MIM]+[Cl]-) was more effective in dispersing mature biofilms. This promising ionic liquid compound ([C16MIM]+[Cl]-) was chosen for determining the underlying mode of action on C. albicans cells. Light microscopy showed that ionic liquid treatment led to a significant reduction in cell volume and length. Increased cell membrane permeability in the ionic liquid treated C. albicans cells was evident in propidium iodide staining. Leakage of intracellular material was evident in terms of increased absorbance of supernatant and release of potassium and calcium ions into extracellular medium. A decrease in ergosterol content was evident when C. albicans cells were cultured in the presence of antifungal ionic liquid. 2',7'-Dichlorodihydrofluorescein acetate assay revealed reactive oxygen species generation and accumulation in C. albicans cells upon treatment with antifungal ionic liquid. The effect of antifungal ionic liquid on mitochondria was evident by decreased membrane potential (measured by Rhodamine 123 assay) and loss of metabolic activity (measured by MTT assay). This study demonstrated that imidazolium ionic liquid compound exert antifungal and antibiofilm activity by affecting various cellular processes. Thus, imidazolium ionic liquids represent a promising antifungal treatment strategy in lieu of resistance development to common antifungal drugs.
Collapse
Affiliation(s)
- G. Kiran Kumar Reddy
- Biofouling and Biofilm Processes, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, India
- Homi Bhabha National Institute, Mumbai, India
| | - Y. V. Nancharaiah
- Biofouling and Biofilm Processes, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
99
|
Lactoferrin Is Broadly Active against Yeasts and Highly Synergistic with Amphotericin B. Antimicrob Agents Chemother 2020; 64:AAC.02284-19. [PMID: 32094132 PMCID: PMC7179636 DOI: 10.1128/aac.02284-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/15/2020] [Indexed: 12/23/2022] Open
Abstract
Lactoferrin (LF) is a multifunctional milk protein with antimicrobial activity against a range of pathogens. While numerous studies report that LF is active against fungi, there are considerable differences in the level of antifungal activity and the capacity of LF to interact with other drugs. Here we undertook a comprehensive evaluation of the antifungal spectrum of activity of three defined sources of LF across 22 yeast and 24 mold species and assessed its interactions with six widely used antifungal drugs. LF was broadly and consistently active against all yeast species tested (MICs, 8 to 64 μg/ml), with the extent of activity being strongly affected by iron saturation. LF was synergistic with amphotericin B (AMB) against 19 out of 22 yeast species tested, and synergy was unaffected by iron saturation but was affected by the extent of LF digestion. LF-AMB combination therapy significantly prolonged the survival of Galleria mellonella wax moth larvae infected with Candida albicans or Cryptococcus neoformans and decreased the fungal burden 12- to 25-fold. Evidence that LF directly interacts with the fungal cell surface was seen via scanning electron microscopy, which showed pore formation, hyphal thinning, and major cell collapse in response to LF-AMB synergy. Important virulence mechanisms were disrupted by LF-AMB treatment, which significantly prevented biofilms in C. albicans and C. glabrata, inhibited hyphal development in C. albicans, and reduced cell and capsule size and phenotypic diversity in Cryptococcus Our results demonstrate the potential of LF-AMB as an antifungal treatment that is broadly synergistic against important yeast pathogens, with the synergy being attributed to the presence of one or more LF peptides.
Collapse
|
100
|
Shin DS, Rhee KJ, Eom YB. Effect of Probiotic Clostridium butyricum NCTC 7423 Supernatant on Biofilm Formation and Gene Expression of Bacteroides fragilis. J Microbiol Biotechnol 2020; 30:368-377. [PMID: 32066216 PMCID: PMC9728305 DOI: 10.4014/jmb.2001.01027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Enterotoxigenic Bacteroides fragilis (ETBF) is the main pathogen causing severe inflammatory diseases and colorectal cancer. Its biofilm plays a key role in the development of colorectal cancer. The objective of this study was to determine the antagonistic effects of cell-free supernatants (CFS) derived from Clostridium butyricum against the growth and biofilm of ETBF. Our data showed that C. butyricum CFS inhibited the growth of B. fragilis in planktonic culture. In addition, C. butyricum CFS exhibited an antibiofilm effect by inhibiting biofilm development, disassembling preformed biofilms and reducing the metabolic activity of cells in biofilms. Using confocal laser scanning microscopy, we found that C. butyricum CFS significantly suppressed the proteins and extracellular nucleic acids among the basic biofilm components. Furthermore, C. butyricum CFS significantly downregulated the expression of virulence- and efflux pump-related genes including ompA and bmeB3 in B. fragilis. Our findings suggest that C. butyricum can be used as biotherapeutic agent by inhibiting the growth and biofilm of ETBF.
Collapse
Affiliation(s)
- Da-Seul Shin
- Department of Medical Sciences, College of Medical Sciences, Soonchunhyang University, Asan 3538, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju 6493, Republic of Korea
| | - Yong-Bin Eom
- Department of Medical Sciences, College of Medical Sciences, Soonchunhyang University, Asan 3538, Republic of Korea,Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan 158, Republic of Korea,Corresponding author Phone: +82-41-530-3039 Fax: +82-41-530-3085 E-mail:
| |
Collapse
|