1
|
Almeida NLM, Peralta LCF, Pontes FML, Rinaldo D, Porto VC, Lara VS. Anti-Candida activity and biocompatibility of silver nanoparticles associated with denture glaze: a new approach to the management of denture stomatitis. Folia Microbiol (Praha) 2024; 69:1229-1246. [PMID: 38652435 DOI: 10.1007/s12223-024-01161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
The association of silver nanoparticles (AgNps) to sealant agent Palaseal® can be a promising alternative for complete denture wearers who may develop denture stomatitis (DS). The study aimed to evaluate the anti-Candida and biocompatible potential of silver nanoparticles synthesized by three routes associated with denture glaze to prevent and/or treat oral candidiasis. Surface acrylic resin specimens were treated with different associations of glaze with AgNps (VER+AgUV, VER+AgTurk and VER+AgGm). As controls, specimens were treated with glaze+nystatin (VER+Nyst), glaze only (VER) or submerged in PBS (PBS). Afterwards, Candida albicans biofilm was developed for 24 h, 15 d and 30 d. Subsequently, the biofilm was quantified by CFU/mL, XTT assay and confocal laser scanning microscopy. Fibroblasts were submitted to conditioned medium with the same associations for 24, 48 and 72 h and LIVE/DEAD® viability test was carried out. Regardless of the period, there was a significant reduction (p < 0.01) of viable fungal cells load, as well as inhibition of fungal metabolic activity, in specimens treated with glaze+AgNps associations, compared to VER and PBS. The anti-Candida effects of the associations were similar to the VER+Nyst group, with emphasis on VER+AgGm, which showed the highest percentage values of non-viable fungal cells maintained over time. The associations did not prove toxicity to fibroblasts. The AgNps exerted antimicrobial activity against C. albicans biofilms and are biocompatible. The most effective results were achieved with the association of glaze+silver nanoparticles synthesized by the green chemistry method (AgGm), proving to be an innovative alternative in the management of DS.
Collapse
Affiliation(s)
- Nara Lígia Martins Almeida
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, 17012-901, Brazil
| | - Laura Catalí Ferreira Peralta
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru 17012-901, Brazil
| | | | - Daniel Rinaldo
- Department of Chemistry, São Paulo State University (UNESP), Bauru 17033-360, Brazil
| | - Vinicius Carvalho Porto
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru 17012-901, Brazil
| | - Vanessa Soares Lara
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, 17012-901, Brazil.
| |
Collapse
|
2
|
Pezzotti G, Adachi T, Imamura H, Ikegami S, Kitahara R, Yamamoto T, Kanamura N, Zhu W, Ishibashi KI, Okuma K, Mazda O, Komori A, Komatsuzawa H, Makimura K. Raman Spectroscopic Algorithms for Assessing Virulence in Oral Candidiasis: The Fight-or-Flight Response. Int J Mol Sci 2024; 25:11410. [PMID: 39518963 PMCID: PMC11545699 DOI: 10.3390/ijms252111410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
This study aimed to test the effectiveness of Raman spectroscopy in the characterization of the degrees of physiological stress and virulence in clinical swab samples collected from patients affected by oral candidiasis. Raman experiments were conducted on a series of eight isolates, both in an as-collected state and after biofilm purification followed by 3 days of culture. The outputs were matched to optical microscopy observations and the results of conventional chromogenic medium assays. A statistically significant series of ten Raman spectra were collected for each clinical sample, and their averages were examined and interpreted as multiomic snapshots for albicans and non-albicans species. Spectroscopic analyses based on selected Raman parameters previously developed for standard Candida samples revealed an extreme structural complexity for all of the clinical samples, which arose from the concurrent presence of a variety of biofilms and commensal bacteria in the samples, as well as a number of other biochemical circumstances affecting the cells in their physiological stress state. However, three Raman algorithms survived such complexity, which enabled insightful classifications of Candida cells from clinical samples, in terms of their physiological stress and morphogenic state, membrane permeability, and virulence. These three characteristics, in turn, converged into a seemingly "fight or flight" response of the Candida cells. Although yet preliminary, the present study points out criticalities and proposes solutions regarding the potential utility of Raman spectroscopy in fast bedside analyses of surveillance samples.
Collapse
Affiliation(s)
- Giuseppe Pezzotti
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (H.I.); (S.I.); (W.Z.)
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan; (T.A.); (O.M.)
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.)
- Department of Orthopedic Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
- Department of Molecular Science and Nanosystems, Ca’ Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Tetsuya Adachi
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan; (T.A.); (O.M.)
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.)
- Department of Microbiology, Graduate School of Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan;
- Department of Dentistry, Kyoto Prefectural Rehabilitation Hospital for Mentally and Physically Disabled, Naka Ashihara, Joyo, Kyoto 610-0113, Japan
| | - Hayata Imamura
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (H.I.); (S.I.); (W.Z.)
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.)
| | - Saki Ikegami
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (H.I.); (S.I.); (W.Z.)
- Department of Microbiology, Graduate School of Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan;
| | - Ryo Kitahara
- Structural Biology Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Kusatsu 525-8577, Japan;
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.)
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.)
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (H.I.); (S.I.); (W.Z.)
| | - Ken-ichi Ishibashi
- Laboratory of Host Defense and Responses, Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Saitama, Sakado, Saitama 350-0288, Japan;
| | - Kazu Okuma
- Department of Microbiology, Graduate School of Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan;
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan; (T.A.); (O.M.)
| | - Aya Komori
- Medical Mycology, Graduate School of Medicine, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (A.K.); (K.M.)
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
| | - Koichi Makimura
- Medical Mycology, Graduate School of Medicine, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (A.K.); (K.M.)
- Teikyo University Institute of Medical Mycology (TIMM), 359 Otsuka, Hachijoji, Tokyo 192-0395, Japan
| |
Collapse
|
3
|
Corbu VM, Georgescu AM, Marinas IC, Pericleanu R, Mogos DV, Dumbravă AȘ, Marinescu L, Pecete I, Vassu-Dimov T, Czobor Barbu I, Csutak O, Ficai D, Gheorghe-Barbu I. Phenotypic and Genotypic Characterization of Resistance and Virulence Markers in Candida spp. Isolated from Community-Acquired Infections in Bucharest, and the Impact of AgNPs on the Highly Resistant Isolates. J Fungi (Basel) 2024; 10:563. [PMID: 39194889 DOI: 10.3390/jof10080563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND This study aimed to determine, at the phenotypic and molecular levels, resistance and virulence markers in Candida spp. isolated from community-acquired infections in Bucharest outpatients during 2021, and to demonstrate the efficiency of alternative solutions against them based on silver nanoparticles (AgNPs). METHODS A total of 62 Candida spp. strains were isolated from dermatomycoses and identified using chromogenic culture media and MALDI-TOF MS, and then investigated for their antimicrobial resistance and virulence markers (VMs), as well as for metabolic enzymes using enzymatic tests for the expression of soluble virulence factors, their biofilm formation and adherence capacity on HeLa cells, and PCR assays for the detection of virulence markers and the antimicrobial activity of alternative solutions based on AgNPs. RESULTS Of the total of 62 strains, 45.16% were Candida parapsilosis; 29.03% Candida albicans; 9.67% Candida guilliermondii; 3.22% Candida lusitaniae, Candia pararugosa, and Candida tropicalis; and 1.66% Candida kefyr, Candida famata, Candida haemulonii, and Candida metapsilosis. Aesculin hydrolysis, caseinase, and amylase production were detected in the analyzed strains. The strains exhibited different indices of adherence to HeLa cells and were positive in decreasing frequency order for the LIP1, HWP1, and ALS1,3 genes (C. tropicalis/C. albicans). An inhibitory effect on microbial growth, adherence capacity, and on the production of virulence factors was obtained using AgNPs. CONCLUSIONS The obtained results in C. albicans and Candida non-albicans circulating in Bucharest outpatients were characterized by moderate-to-high potential to produce VMs, necessitating epidemiological surveillance measures to minimize the chances of severe invasive infections.
Collapse
Affiliation(s)
- Viorica Maria Corbu
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
- The Research Institute of the University of Bucharest (ICUB), 050095 Bucharest, Romania
| | - Ana-Maria Georgescu
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
| | | | - Radu Pericleanu
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
| | - Denisa Vasilica Mogos
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
| | - Andreea Ștefania Dumbravă
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
- The Research Institute of the University of Bucharest (ICUB), 050095 Bucharest, Romania
| | - Liliana Marinescu
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology Politechnica of Bucharest, 060042 Bucharest, Romania
| | - Ionut Pecete
- Central Reference Synevo-Medicover Laboratory, 021408 Bucharest, Romania
| | - Tatiana Vassu-Dimov
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
| | - Ilda Czobor Barbu
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
- The Research Institute of the University of Bucharest (ICUB), 050095 Bucharest, Romania
| | - Ortansa Csutak
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
- The Research Institute of the University of Bucharest (ICUB), 050095 Bucharest, Romania
| | - Denisa Ficai
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology Politechnica of Bucharest, 060042 Bucharest, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Irina Gheorghe-Barbu
- Faculty of Biology, University of Bucharest, Intrarea Portocalelor No. 1-3, 060101 Bucharest, Romania
- The Research Institute of the University of Bucharest (ICUB), 050095 Bucharest, Romania
| |
Collapse
|
4
|
Mannala GK, Rupp M, Walter N, Scholz KJ, Simon M, Riool M, Alt V. Galleria mellonella as an alternative in vivo model to study implant-associated fungal infections. J Orthop Res 2023; 41:2547-2559. [PMID: 37080929 DOI: 10.1002/jor.25572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/22/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Fungal implant-associated bone infections are rare but difficult to treat and often associated with a poor outcome for patients. Candida species account for approximately 90% of all fungal infections. In vivo biofilm models play a major role to study biofilm development and potential new treatment options; however, there are only a very few in vivo models to study fungi-associated biofilms. Furthermore, mammalian infection models are replaced more and more due to ethical restrictions with other alternative models in basic research. Recently, we developed an insect infection model with Galleria mellonella larvae to study biofilm-associated infections with bacteria. Here, we further expanded the G. mellonella model to study in vivo fungal infections using Candida albicans and Candida krusei. We established a planktonic and biofilm-implant model to test different antifungal medication with amphotericin B, fluconazole, and voriconazole against the two species and assessed the fungal biofilm-load on the implant surface. Planktonic infection with C. albicans and C. krusei showed the killing of the G. mellonella larvae at 5 × 105 colony forming units (CFU). Treatment of larvae with antifungal compounds with amphotericin B and fluconazole showed significant survival improvement against planktonic C. albicans infection, but voriconazole had no effect. Titanium and stainless steel K-wires were preincubated with C. albicans and implanted inside the larvae to induce biofilm infection on the implant surface. The survival analysis revealed significantly reduced survival of the larvae with Candida spp. infection compared to noninfected implants. The treatment with antifungal amphotericin B and fluconazole resulted in a slight and nonsignificant improvement survival of the larvae. The treatment with the antifungal compounds in the biofilm-infection model was not as effective as in the planktonic infection model, which highlights the resistance of fungal biofilms to antifungal compounds like in bacterial biofilms. Scanning electron microscopy (SEM) analysis revealed the formation of a fungal biofilm with hyphae and spores associated with larvae tissue on the implant surface. Thus, our study highlights the use of G. mellonella larvae as alternative in vivo model to study biofilm-associated implant fungal infections and that fungal biofilms exhibit high resistance profiles comparable to bacterial biofilms. The model can be used in the future to test antifungal treatment options for fungal biofilm infections.
Collapse
Affiliation(s)
- Gopala K Mannala
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Markus Rupp
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Nike Walter
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Konstantin J Scholz
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Regensburg, Germany
| | - Michaela Simon
- Institute of Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Martijn Riool
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Volker Alt
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
5
|
Peralta LCF, Almeida NLM, Pontes FML, Rinaldo D, Carneiro CA, Neppelenbroek KH, Lara VS, Porto VC. Silver nanoparticles in denture adhesive: An antimicrobial approach against Candida albicans. J Dent 2023; 131:104445. [PMID: 36773742 DOI: 10.1016/j.jdent.2023.104445] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/21/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE To evaluate the antimicrobial potential of silver nanoparticles (Ag NPs) synthesized using three different routes (ultraviolet light, Turkevich, and green chemistry method using Glycine max extract) associated with COREGA® denture powder adhesive. METHODS Heat-cured acrylic resin specimens were treated with different Ag NPs associated with the adhesive (AD + Ag UV, AD + Ag Turk, and AD + Ag Gm groups). As controls, the specimens were treated with a combination of adhesive and nystatin (AD + Nyst group), only adhesive (AD group), or submerged on the surface of the specimens (PBS group). After the treatments, biofilms of C. albicans developed for 3, 6, and 12 h on the specimen surfaces. The biofilm was quantified using colony-forming units per milliliter, colorimetric assay, and confocal laser scanning microscopy. RESULTS Regardless of the period, we observed an inhibition of fungal load and a reduction in metabolic activity and biofilm mass in the resin specimens treated with the combinations AD/Ag NPs, compared to AD and PBS. The antimicrobial action of the AD + Turk and AD + Ag Gm groups was similar than that for the AD + Nyst group in all periods and viability tests, except for the biofilm mass (12 h). CONCLUSIONS The COREGA® adhesive with Ag NPs, mainly those synthesized using the Turkevich and Glycine max methods, showed excellent antimicrobial activity against C. albicans biofilms, maintained for up to 12 h. CLINICAL SIGNIFICANCE The association of Ag NPs to the adhesive can add preventive or therapeutic effects against denture stomatitis, to this prosthetic material.
Collapse
Affiliation(s)
- Laura Catalí Ferreira Peralta
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil, 17012-901
| | - Nara Ligia Martins Almeida
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil, 17012-901
| | | | - Daniel Rinaldo
- Department of Chemistry, São Paulo State University (UNESP), Bauru, Brazil
| | - Camila Alves Carneiro
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil, 17012-901
| | - Karin Hermana Neppelenbroek
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil, 17012-901
| | - Vanessa Soares Lara
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil, 17012-901
| | - Vinicius Carvalho Porto
- Department of Prosthodontics and Periodontics, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil, 17012-901.
| |
Collapse
|
6
|
Prasad P, Tippana M. Morphogenic plasticity: the pathogenic attribute of Candida albicans. Curr Genet 2023; 69:77-89. [PMID: 36947241 DOI: 10.1007/s00294-023-01263-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 03/23/2023]
Abstract
Candida albicans is a commensal organism of the human gastrointestinal tract and a prevalent opportunistic pathogen. It exhibits different morphogenic forms to survive in different host niches with distinct environmental conditions (pH, temperature, oxidative stress, nutrients, serum, chemicals, radiation, etc.) and genetic factors (transcription factors and genes). The different morphogenic forms of C. albicans are yeast, hyphal, pseudohyphal, white, opaque, and transient gray cells, planktonic and biofilm forms of cells. These forms differ in the parameters like cellular phenotype, colony morphology, adhesion to solid surfaces, gene expression profile, and the virulent traits. Each form is functionally distinct and responds discretely to the host immune system and antifungal drugs. Hence, morphogenic plasticity is the key to virulence. In this review, we address the characteristics, the pathogenic potential of the different morphogenic forms and the conditions required for morphogenic transitions.
Collapse
Affiliation(s)
- Priya Prasad
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, India.
| | - Meena Tippana
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, India
| |
Collapse
|
7
|
Loaiza-Oliva M, Arias-Durango L, Martínez-Pabón MC. The Cytotoxic and Inhibitory Effects of Plant Derivatives on Candida albicans Biofilms: A Scoping Review. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010130. [PMID: 36615324 PMCID: PMC9822484 DOI: 10.3390/molecules28010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Candida albicans infections are related to biofilm formation. The increase in antifungal resistance and their adverse effects have led to the search for therapeutic options as plant derivatives. This scoping review aims to identify the current status of in vitro research on the cytotoxicity and inhibitory effects of plant derivatives on C. albicans biofilms. In this study, PRISMA items were followed. After recognition of the inclusion criteria, full texts were read and disagreements were resolved with a third party. A risk of bias assessment was performed, and information was summarized using Microsoft Office Excel. Thirty-nine papers fulfilling the selection criteria were included. The risk of bias analysis identified most of the studies as low risk. Studies evaluated plant derivatives such as extracts, essential oils, terpenes, alkaloids, flavonoids and polyphenols. Some studies evaluated the inhibition of C. albicans biofilm formation, inhibition on preformed biofilms or both. The derivatives at concentrations greater than or equal to those that have an inhibitory effect on C. albicans biofilms, without showing cytotoxicity, include magnoflorin, ellagic acid, myricetin and eucarobustol from Eucalyptus robusta and, as the works in which these derivatives were studied are of good quality, it is desirable to carry out study in other experimental phases, with methodologies that generate comparable information.
Collapse
|
8
|
Lin P, Zhang J, Xie G, Li J, Guo C, Lin H, Zhang Y. Innate Immune Responses to Sporothrix schenckii: Recognition and Elimination. Mycopathologia 2022; 188:71-86. [PMID: 36329281 DOI: 10.1007/s11046-022-00683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/24/2022] [Indexed: 11/06/2022]
Abstract
Sporothrix schenckii (S. schenckii), a ubiquitous thermally dimorphic fungus, is the etiological agent of sporotrichosis, affecting immunocompromised and immunocompetent individuals. Despite current antifungal regimens, sporotrichosis results in prolonged treatment and significant mortality rates in the immunosuppressed population. The innate immune system forms the host's first and primary line of defense against S. schenckii, which has a bi-layered cell wall structure. Many components act as pathogen-associated molecular patterns (PAMPs) in pathogen-host interactions. PAMPs are recognized by pattern recognition receptors (PRRs) such as toll-like receptors, C-type lectin receptors, and complement receptors, triggering innate immune cells such as neutrophils, macrophages, and dendritic cells to phagocytize or produce mediators, contributing to S. schenckii elimination. The ultrastructure of S. schenckii and pathogen-host interactions, including PRRs and innate immune cells, are summarized in this review, promoting a better understanding of the innate immune response to S. schenckii and aiding in the development of protective and therapeutic strategies to combat sporotrichosis.
Collapse
Affiliation(s)
- Peng Lin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianfeng Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guinan Xie
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junchen Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chenqi Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haiyue Lin
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Yu Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China.
| |
Collapse
|
9
|
Wang Y, Zhou H, Wang W, Duan N, Luo Z, Chai H, Jiang L, Chen Q, Liu J, Hua H, Yan Z, Fan Y, Xu J, Guan X, Wang H, Lu H, Lun W, Fei W, Zhang T, Zhao J, Jia C, Kong H, Shen X, Liu Q, Wang W, Tang G. Efficacy and safety of miconazole muco-adhesive tablet versus itraconazole in oropharyngeal candidiasis: A randomized, multi-centered, double-blind, phase 3 trial. Med Mycol 2022; 60:myac076. [PMID: 36149321 DOI: 10.1093/mmy/myac076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/16/2022] [Accepted: 09/21/2022] [Indexed: 11/14/2022] Open
Abstract
Oropharyngeal candidiasis (OPC) is an opportunistic infection treated with anti-fungal agents. Herein, we evaluate the efficacy and safety of miconazole buccal tablets (MBT) and itraconazole capsules in the localized treatment of patients with OPC. In this multi-centered, double-blinded, phase III trial (CTR20130414), both males and non-pregnant females (≥18 years) with OPC were randomized (1:1) to MBT plus placebo (experimental group) or itraconazole capsules plus placebo (control group). The primary endpoint was clinical cure at the end-of-treatment period [visit 4 (V4)] while secondary endpoints were clinical remission rates, partial remission rates, mycological cure, clinical relapse, and adverse events (AEs). All endpoints were statistically analyzed in both the full analysis set (FAS) and per-protocol (PP) set. A total of 431 (experimental: 216; control: 215) subjects were included. At V4, in the FAS set, the clinical cure was achieved in 68% and 59% patients in experimental and control groups, respectively with a treatment difference of 9% [95% confidence interval (CI): -1,19; P < .001] demonstrating non-inferiority of MBT over itraconazole. At V4, mycological cure rates were 68.2% and 42.0% in the experimental group and control groups (P < .001), respectively in FAS. The relapse rates were 5.4% and 6.6%, respectively, in the experimental and control groups. A total of 210 patients experienced AEs during treatment with 47.7% in the experimental group and 49.8% in the control group with no deaths. This study demonstrated that once-daily treatment with MBT was non-inferior to itraconazole with higher mycological cure rates and was tolerable with mild AE in patients with OPC.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| | - Haiwen Zhou
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| | - Wenmei Wang
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, Jiangsu, China
| | - Ning Duan
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, Jiangsu, China
| | - Zhixiao Luo
- Dental Department, Taihe Hospital, Affiliated Hospital of Hubei, University of Medicine, Shiyan 442099, Hubei, China
| | - Hongbo Chai
- Dental Department, Taihe Hospital, Affiliated Hospital of Hubei, University of Medicine, Shiyan 442099, Hubei, China
| | - Lu Jiang
- Department of Oral Medicine, West China School/Hospital Stomatology Sichuan University, Chengdu 610042, Sichuan, China
| | - Qianming Chen
- Department of Oral Medicine, West China School/Hospital Stomatology Sichuan University, Chengdu 610042, Sichuan, China
| | - Jinli Liu
- Department of Oral Medicine, West China School/Hospital Stomatology Sichuan University, Chengdu 610042, Sichuan, China
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Zhimin Yan
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yuan Fan
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Juanyong Xu
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xiaobing Guan
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Hongjian Wang
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Hongzhou Lu
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Wenhui Lun
- Department of Dermatology and Venereology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Wei Fei
- Department of Stomatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan, China
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Jizhi Zhao
- Department of Stomatology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Chunling Jia
- Department of Oral Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Hui Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Medicine, Department of Operative Dentistry & Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xuemin Shen
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| | - Qing Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Medicine, Department of Operative Dentistry & Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Weizhi Wang
- Department of Oral Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Guoyao Tang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| |
Collapse
|
10
|
Ramakrishnan R, Singh AK, Singh S, Chakravortty D, Das D. Enzymatic Dispersion of Biofilms: An Emerging Biocatalytic Avenue to Combat Biofilm-Mediated Microbial Infections. J Biol Chem 2022; 298:102352. [PMID: 35940306 PMCID: PMC9478923 DOI: 10.1016/j.jbc.2022.102352] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/01/2023] Open
Abstract
Drug resistance by pathogenic microbes has emerged as a matter of great concern to mankind. Microorganisms such as bacteria and fungi employ multiple defense mechanisms against drugs and the host immune system. A major line of microbial defense is the biofilm, which comprises extracellular polymeric substances that are produced by the population of microorganisms. Around 80% of chronic bacterial infections are associated with biofilms. The presence of biofilms can increase the necessity of doses of certain antibiotics up to 1000-fold to combat infection. Thus, there is an urgent need for strategies to eradicate biofilms. Although a few physicochemical methods have been developed to prevent and treat biofilms, these methods have poor efficacy and biocompatibility. In this review, we discuss the existing strategies to combat biofilms and their challenges. Subsequently, we spotlight the potential of enzymes, in particular, polysaccharide degrading enzymes, for biofilm dispersion, which might lead to facile antimicrobial treatment of biofilm-associated infections.
Collapse
Affiliation(s)
- Reshma Ramakrishnan
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Ashish Kumar Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Simran Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Debasis Das
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
11
|
Rashid S, Correia-Mesquita TO, Godoy P, Omran RP, Whiteway M. SAGA Complex Subunits in Candida albicans Differentially Regulate Filamentation, Invasiveness, and Biofilm Formation. Front Cell Infect Microbiol 2022; 12:764711. [PMID: 35350439 PMCID: PMC8957876 DOI: 10.3389/fcimb.2022.764711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/11/2022] [Indexed: 11/24/2022] Open
Abstract
SAGA (Spt-Ada-Gcn5-acetyltransferase) is a highly conserved, multiprotein co-activator complex that consists of five distinct modules. It has two enzymatic functions, a histone acetyltransferase (HAT) and a deubiquitinase (DUB) and plays a central role in processes such as transcription initiation, elongation, protein stability, and telomere maintenance. We analyzed conditional and null mutants of the SAGA complex module components in the fungal pathogen Candida albicans; Ngg1, (the HAT module); Ubp8, (the DUB module); Tra1, (the recruitment module), Spt7, (the architecture module) and Spt8, (the TBP interaction unit), and assessed their roles in a variety of cellular processes. We observed that spt7Δ/Δ and spt8Δ/Δ strains have a filamentous phenotype, and both are highly invasive in yeast growing conditions as compared to the wild type, while ngg1Δ/Δ and ubp8Δ/Δ are in yeast-locked state and non-invasive in both YPD media and filamentous induced conditions compared to wild type. RNA-sequencing-based transcriptional profiling of SAGA mutants reveals upregulation of hyphal specific genes in spt7Δ/Δ and spt8Δ/Δ strains and downregulation of ergosterol metabolism pathway. As well, spt7Δ/Δ and spt8Δ/Δ confer susceptibility to antifungal drugs, to acidic and alkaline pH, to high temperature, and to osmotic, oxidative, cell wall, and DNA damage stresses, indicating that these proteins are important for genotoxic and cellular stress responses. Despite having similar morphological phenotypes (constitutively filamentous and invasive) spt7 and spt8 mutants displayed variation in nuclear distribution where spt7Δ/Δ cells were frequently binucleate and spt8Δ/Δ cells were consistently mononucleate. We also observed that spt7Δ/Δ and spt8Δ/Δ mutants were quickly engulfed by macrophages compared to ngg1Δ/Δ and ubp8Δ/Δ strains. All these findings suggest that the SAGA complex modules can have contrasting functions where loss of Spt7 or Spt8 enhances filamentation and invasiveness while loss of Ngg1 or Ubp8 blocks these processes.
Collapse
Affiliation(s)
| | | | | | | | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
12
|
Application of proper orthogonal decomposition for evaluation of coherent structures and energy contents in microbial biofilms. METHODS IN MICROBIOLOGY 2022; 194:106420. [DOI: 10.1016/j.mimet.2022.106420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022]
|
13
|
Vila T, Kong EF, Montelongo-Jauregui D, Van Dijck P, Shetty AC, McCracken C, Bruno VM, Jabra-Rizk MA. Therapeutic implications of C. albicans-S. aureus mixed biofilm in a murine subcutaneous catheter model of polymicrobial infection. Virulence 2021; 12:835-851. [PMID: 33682623 PMCID: PMC7946022 DOI: 10.1080/21505594.2021.1894834] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Biofilm-associated polymicrobial infections tend to be challenging to treat. Candida albicans and Staphylococcus aureus are leading pathogens due to their ability to form biofilms on medical devices. However, the therapeutic implications of their interactions in a host is largely unexplored. In this study, we used a mouse subcutaneous catheter model for in vivo-grown polymicrobial biofilms to validate our in vitro findings on C. albicans-mediated enhanced S. aureus tolerance to vancomycin in vivo. Comparative assessment of S. aureus recovery from catheters with single- or mixed-species infection demonstrated failure of vancomycin against S. aureus in mice with co-infected catheters. To provide some mechanistic insights, RNA-seq analysis was performed on catheter biofilms to delineate transcriptional modulations during polymicrobial infections. C. albicans induced the activation of the S. aureus biofilm formation network via down-regulation of the lrg operon, repressor of autolysis, and up-regulation of the ica operon and production of polysaccharide intercellular adhesin (PIA), indicating an increase in eDNA production, and extracellular polysaccharide matrix, respectively. Interestingly, virulence factors important for disseminated infections, and superantigen-like proteins were down-regulated during mixed-species infection, whereas capsular polysaccharide genes were up-regulated, signifying a strategy favoring survival, persistence and host immune evasion. In vitro follow-up experiments using DNA enzymatic digestion, lrg operon mutant strains, and confocal scanning microscopy confirmed the role of C. albicans-mediated enhanced eDNA production in mixed-biofilms on S. aureus tolerance to vancomycin. Combined, these findings provide mechanistic insights into the therapeutic implications of interspecies interactions, underscoring the need for novel strategies to overcome limitations of current therapies.
Collapse
Affiliation(s)
- Taissa Vila
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Eric F Kong
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium.,VIB-KU Leuven Center for Microbiology, Flanders, Belgium
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vincent M Bruno
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Lu H, Shrivastava M, Whiteway M, Jiang Y. Candida albicans targets that potentially synergize with fluconazole. Crit Rev Microbiol 2021; 47:323-337. [PMID: 33587857 DOI: 10.1080/1040841x.2021.1884641] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fluconazole has characteristics that make it widely used in the clinical treatment of C. albicans infections. However, fluconazole has only a fungistatic activity in C. albicans, therefore, in the long-term treatment of C. albicans infection with fluconazole, C. albicans has the potential to acquire fluconazole resistance. A promising approach to increase fluconazole's efficacy is identifying potential targets of drugs that can enhance the antifungal effect of fluconazole, or even make the drug fungicidal. In this review, we systematically provide a global overview of potential targets of drugs synergistic with fluconazole in C. albicans, identify new avenues for research on fluconazole potentiation, and highlight the promise of combinatorial strategies with fluconazole in combatting C. albicans infections.
Collapse
Affiliation(s)
- Hui Lu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Pereira R, Dos Santos Fontenelle RO, de Brito EHS, de Morais SM. Biofilm of Candida albicans: formation, regulation and resistance. J Appl Microbiol 2020; 131:11-22. [PMID: 33249681 DOI: 10.1111/jam.14949] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/10/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022]
Abstract
Candida albicans is the most common human fungal pathogen, causing infections that range from mucous membranes to systemic infections. The present article provides an overview of C. albicans, with the production of biofilms produced by this fungus, as well as reporting the classes of antifungals used to fight such infections, together with the resistance mechanisms to these drugs. Candida albicans is highly adaptable, enabling the transition from commensal to pathogen due to a repertoire of virulence factors. Specifically, the ability to change morphology and form biofilms is central to the pathogenesis of C. albicans. Indeed, most infections by this pathogen are associated with the formation of biofilms on surfaces of hosts or medical devices, causing high morbidity and mortality. Significantly, biofilms formed by C. albicans are inherently tolerant to antimicrobial therapy, so the susceptibility of C. albicans biofilms to current therapeutic agents remains low. Therefore, it is difficult to predict which molecules will emerge as new clinical antifungals. The biofilm formation of C. albicans has been causing impacts on susceptibility to antifungals, leading to resistance, which demonstrates the importance of research aimed at the prevention and control of these clinical microbial communities.
Collapse
Affiliation(s)
- R Pereira
- Graduate Program in Biotechnology, Microbiology Laboratory (LABMIC), Vale do Acaraú State University, Sobral, Ceará, Brazil
| | | | - E H S de Brito
- Institute of Health Sciences of University for International Integration of Afro-Brazilian Lusophony, Redenção, Ceará, Brazil
| | - S M de Morais
- Graduate Program in Biotechnology, Laboratory of Chemistry of Natural Products (LQPN), Ceará State University, Fortaleza, Ceará, Brazil
| |
Collapse
|
16
|
Rodríguez-Cerdeira C, Martínez-Herrera E, Carnero-Gregorio M, López-Barcenas A, Fabbrocini G, Fida M, El-Samahy M, González-Cespón JL. Pathogenesis and Clinical Relevance of Candida Biofilms in Vulvovaginal Candidiasis. Front Microbiol 2020; 11:544480. [PMID: 33262741 PMCID: PMC7686049 DOI: 10.3389/fmicb.2020.544480] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/23/2020] [Indexed: 12/30/2022] Open
Abstract
The ability of Candida spp. to form biofilms is crucial for its pathogenicity, and thus, it should be considered an important virulence factor in vulvovaginal candidiasis (VVC) and recurrent VVC (RVVC). Its ability to generate biofilms is multifactorial and is generally believed to depend on the site of infection, species and strain involved, and the microenvironment in which the infection develops. Therefore, both cell surface proteins, such as Hwp1, Als1, and Als2, and the cell wall-related protein, Sun41, play a critical role in the adhesion and virulence of the biofilm. Immunological and pharmacological approaches have identified the NLRP3 inflammasome as a crucial molecular factor contributing to host immunopathology. In this context, we have earlier shown that Candida albicans associated with hyphae-secreted aspartyl proteinases (specifically SAP4-6) contribute to the immunopathology of the disease. Transcriptome profiling has revealed that non-coding transcripts regulate protein synthesis post-transcriptionally, which is important for the growth of Candida spp. Other studies have employed RNA sequencing to identify differences in the 1,245 Candida genes involved in surface and invasive cellular metabolism regulation. In vitro systems allow the simultaneous processing of a large number of samples, making them an ideal screening technique for estimating various physicochemical parameters, testing the activity of antimicrobial agents, and analyzing genes involved in biofilm formation and regulation (in situ) in specific strains. Murine VVC models are used to study C. albicans infection, especially in trials of novel treatments and to understand the cause(s) for resistance to conventional therapeutics. This review on the clinical relevance of Candida biofilms in VVC focuses on important advances in its genomics, transcriptomics, and proteomics. Moreover, recent experiments on the influence of biofilm formation on VVC or RVVC pathogenesis in laboratory animals have been discussed. A clear elucidation of one of the pathogenesis mechanisms employed by Candida biofilms in vulvovaginal candidiasis and its applications in clinical practice represents the most significant contribution of this manuscript.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cerdeira
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Health Research Institute, SERGAS-UVIGO, Vigo, Spain.,Department of Dermatology, Hospital do Meixoeiro and University of Vigo, Vigo, Spain.,European Women's Dermatologic and Venereologic Society, Tui, Spain.,Psychodermatology Task Force of the Ibero-Latin American College of Dermatology (CILAD), Buenos Aires, Argentina
| | - Erick Martínez-Herrera
- Psychodermatology Task Force of the Ibero-Latin American College of Dermatology (CILAD), Buenos Aires, Argentina.,Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Ixtapaluca, Mexico
| | - Miguel Carnero-Gregorio
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Health Research Institute, SERGAS-UVIGO, Vigo, Spain.,Department of Molecular Diagnosis (Array & NGS Division), Institute of Cellular and Molecular Studies, Lugo, Spain
| | - Adriana López-Barcenas
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Psychodermatology Task Force of the Ibero-Latin American College of Dermatology (CILAD), Buenos Aires, Argentina.,Section of Mycology, Department of Dermatology, Manuel Gea González hospital, Mexico City, Mexico
| | - Gabriella Fabbrocini
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Department of Dermatology, University of Naples Federico II, Naples, Italy
| | - Monika Fida
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Department of Dermatology, University of Medicine, Tirana, Tirana, Albania
| | - May El-Samahy
- European Women's Dermatologic and Venereologic Society, Tui, Spain.,Department of Dermatology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - José Luís González-Cespón
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Health Research Institute, SERGAS-UVIGO, Vigo, Spain
| |
Collapse
|
17
|
Abirami G, Alexpandi R, Durgadevi R, Kannappan A, Veera Ravi A. Inhibitory Effect of Morin Against Candida albicans Pathogenicity and Virulence Factor Production: An in vitro and in vivo Approaches. Front Microbiol 2020; 11:561298. [PMID: 33193145 PMCID: PMC7644646 DOI: 10.3389/fmicb.2020.561298] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/28/2020] [Indexed: 01/27/2023] Open
Abstract
Candida albicans is considered an exclusive etiologic agent of candidiasis, a very common fungal infection in human. The expression of virulence factors contributes highly to the pathogenicity of C. albicans. These factors include biofilm formation, yeast-to-hyphal transition, adhesins, aspartyl proteases, and phospholipases secretion. Moreover, resistance development is a critical issue for the therapeutic failure of antifungal agents against systemic candidiasis. To circumvent resistance development, the present study investigated the virulence targeted therapeutic activity of the phyto-bioactive compound morin against C. albicans. Morin is a natural compound commonly found in medicinal plants and widely used in the pharmaceutical and cosmetic products/industries. The present study explicated the significant inhibitory potential of morin against biofilm formation and other virulence factors' production, such as yeast-hyphal formation, phospholipase, and exopolymeric substances, in C. albicans. Further, qPCR analysis confirmed the downregulation of biofilm and virlence-related genes in C. albicans upon morin treatment, which is in correspondence with the in vitro bioassays. Further, the docking analysis revealed that morin shows strong affinity with Hwp-1 protein, which regulates the expression of biofilm and hyphal formation in C. albicans and, thereby, abolishes fungal pathogenicity. Moreover, the anti-infective potential of morin against C. albicans-associated systemic candidiasis is confirmed through an in vivo approach using biomedical model organism zebrafish (Danio rerio). The outcomes of the in vivo study demonstrate that the morin treatment effectively rescues animals from C. albicans infections and extends their survival rate by inhibiting the internal colonization of C. albicans. Histopathology analysis revealed extensive candidiasis-related pathognomonic changes in the gills, intestine, and kidney of animals infected with C. albicans, while no extensive abnormalities were observed in morin-treated animals. The results evidenced that morin has the ability to protect against the pathognomonic effect and histopathological lesions caused by C. albicans infection in zebrafish. Thus, the present study suggests that the utilization of morin could act as a potent therapeutic medication for C. albicans instigated candidiasis.
Collapse
Affiliation(s)
- Gurusamy Abirami
- Department of Biotechnology, School of Biological Sciences, Alagappa University, Karaikudi, India
| | - Rajaiah Alexpandi
- Department of Biotechnology, School of Biological Sciences, Alagappa University, Karaikudi, India
| | - Ravindran Durgadevi
- Department of Biotechnology, School of Biological Sciences, Alagappa University, Karaikudi, India
| | - Arunachalam Kannappan
- Department of Biotechnology, School of Biological Sciences, Alagappa University, Karaikudi, India
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Arumugam Veera Ravi
- Department of Biotechnology, School of Biological Sciences, Alagappa University, Karaikudi, India
| |
Collapse
|
18
|
Effects of sodium citrate on the structure and microbial community composition of an early-stage multispecies biofilm model. Sci Rep 2020; 10:16585. [PMID: 33024198 PMCID: PMC7538881 DOI: 10.1038/s41598-020-73731-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 09/22/2020] [Indexed: 11/30/2022] Open
Abstract
In recent years, most biofilm studies have focused on fundamental investigations using multispecies biofilm models developed preferentially in simulated naturally occurring low-nutrient medium than in artificial nutrient-rich medium. Because biofilm development under low-nutrient growth media is slow, natural media are often supplemented with an additional carbon source to increase the rate of biofilm formation. However, there are knowledge gaps in interpreting the effects of such supplementation on the resulting biofilm in terms of structure and microbial community composition. We investigated the effects of supplementation of a simulated freshwater medium with sodium citrate on the resulting structure, bacterial community composition, and microbial network interactions of an early-stage multispecies biofilm model. Qualitative and quantitative analyses of acquired confocal laser scanning microscopy data confirmed that sodium citrate supplementation distinctly increased biofilm biomass. Sequencing data revealed that the microbial community structure of biofilms grown in sodium citrate-supplemented conditions was characterized with increased relative abundance and dominance of Proteobacteria compared with that of biofilms grown in sodium citrate-free conditions. Our findings suggest that the supplementation of a low-nutrient medium with a carbon source in experiments involving multispecies biofilms may lead to structural and compositional biases of the microbial community, causing changes in biofilm phenotype.
Collapse
|
19
|
Vila T, Sultan AS, Montelongo-Jauregui D, Jabra-Rizk MA. Oral Candidiasis: A Disease of Opportunity. J Fungi (Basel) 2020; 6:jof6010015. [PMID: 31963180 PMCID: PMC7151112 DOI: 10.3390/jof6010015] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Oral candidiasis, commonly referred to as “thrush,” is an opportunistic fungal infection that commonly affects the oral mucosa. The main causative agent, Candida albicans, is a highly versatile commensal organism that is well adapted to its human host; however, changes in the host microenvironment can promote the transition from one of commensalism to pathogen. This transition is heavily reliant on an impressive repertoire of virulence factors, most notably cell surface adhesins, proteolytic enzymes, morphologic switching, and the development of drug resistance. In the oral cavity, the co-adhesion of C. albicans with bacteria is crucial for its persistence, and a wide range of synergistic interactions with various oral species were described to enhance colonization in the host. As a frequent colonizer of the oral mucosa, the host immune response in the oral cavity is oriented toward a more tolerogenic state and, therefore, local innate immune defenses play a central role in maintaining Candida in its commensal state. Specifically, in addition to preventing Candida adherence to epithelial cells, saliva is enriched with anti-candidal peptides, considered to be part of the host innate immunity. The T helper 17 (Th17)-type adaptive immune response is mainly involved in mucosal host defenses, controlling initial growth of Candida and inhibiting subsequent tissue invasion. Animal models, most notably the mouse model of oropharyngeal candidiasis and the rat model of denture stomatitis, are instrumental in our understanding of Candida virulence factors and the factors leading to host susceptibility to infections. Given the continuing rise in development of resistance to the limited number of traditional antifungal agents, novel therapeutic strategies are directed toward identifying bioactive compounds that target pathogenic mechanisms to prevent C. albicans transition from harmless commensal to pathogen.
Collapse
Affiliation(s)
- Taissa Vila
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Ahmed S. Sultan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-706-0508; Fax: +1-410-706-0519
| |
Collapse
|
20
|
Brilhante RSN, de Aguiar FRM, da Silva MLQ, de Oliveira JS, de Camargo ZP, Rodrigues AM, Pereira VS, Serpa R, Castelo-Branco DDSCM, Correia EEM, Pereira-Neto WA, Cordeiro RDA, Rocha MFG, Sidrim JJC. Antifungal susceptibility of Sporothrix schenckii complex biofilms. Med Mycol 2019; 56:297-306. [PMID: 28595275 DOI: 10.1093/mmy/myx043] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 05/10/2017] [Indexed: 01/01/2023] Open
Abstract
Sporotrichosis, caused by species of Sporothrix schenckii complex, is the most prevalent subcutaneous mycosis in many areas of Latin America. The aim of this study was to evaluate the ability of Sporothrix spp. to form biofilms in vitro and to characterize the growth kinetics, morphology, and antifungal susceptibility of biofilms against classical antifungals. We investigated the ability of strains to produce biofilms in vitro and determined the effects of exposure to amphotericin B, itraconazole, caspofungin, ketoconazole, voriconazole, and fluconazole at minimum inhibitory concentration (MIC) against planktonic form and at 10× MIC and 50× MIC on the biomass and metabolic activity of these biofilms. Biofilm structure was analyzed by optical microscopy using Congo-red staining, confocal and scanning electron microscopy. Strains were classified for biofilm-forming ability, through the analysis of absorbance of crystal violet retained by biomass of mature biofilms. We found that all S. brasiliensis (n = 10), S. schenckii sensu stricto (n = 2), S. globosa (n = 2), and S. mexicana (n = 4) strains were strong biofilm-producers. The analyzed biofilms had dense network of hyphae and conidia immersed in extracellular matrix, with presence of water channels. Antifungal drugs at the three tested concentrations showed different effects on biomass and metabolic activity of biofilms. However, the best inhibitory response was observed with 50× MIC of amphotericin B and caspofungin, which reduced these parameters. Furthermore, high drug concentrations, especially amphotericin B and caspofungin, showed antifungal activity against these biofilms, probably because they damaged the architecture and extracellular matrix, allowing diffusion of the drugs.
Collapse
Affiliation(s)
- Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Felipe Rodrigues Magalhães de Aguiar
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Maria Lucilene Queiroz da Silva
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Jonathas Sales de Oliveira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Zoilo Pires de Camargo
- Cellular Biology Division, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Anderson Messias Rodrigues
- Cellular Biology Division, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Vandbergue Santos Pereira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Rosana Serpa
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | | | - Edmilson Emanuel Monteiro Correia
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Waldemiro Aquino Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Rossana de Aguiar Cordeiro
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil.,Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Ceará, Brazil
| |
Collapse
|
21
|
Tulasidas S, Rao P, Bhat S, Manipura R. A study on biofilm production and antifungal drug resistance among Candida species from vulvovaginal and bloodstream infections. Infect Drug Resist 2018; 11:2443-2448. [PMID: 30538510 PMCID: PMC6260174 DOI: 10.2147/idr.s179462] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction Candida species, one among the opportunistic fungi, has become a common pathogen causing vaginal thrush and nosocomial bloodstream infections (BSIs). This study aims to evaluate the prevalence and antifungal susceptibility of various Candida species and slime production by Candida species in BSIs and vulvovaginal candidiasis (VVC). Materials and methods A total of 176 samples were collected for a period of 1 year. Anti-fungal susceptibility testing and biofilm production testing were performed by the Kirby-Bauer method and crystal violet assay, respectively. Results Out of 176 samples, 74 (42%) were from BSIs and 102 (58%) were from VVC. The biofilm production was comparatively high in blood isolates, 55 (74%), than cervical isolates, 45 (44%). Increase in the trends of non-albicans Candida (NAC) species was seen in our setup. Good susceptibility rates were seen among Candida species, 82.38% to voriconazole and an increasing resistance pattern of 26.13% to fluconazole. Conclusion Speciation of Candida becomes important as the prevalence of NAC is increasing. Antifungal susceptibility testing by the disk diffusion method is cost effective and should be adopted in routine testing as there is an increasing azole resistance, especially in invasive NAC infections. In this study, there was no correlation of antifungal drugs with the biofilm production.
Collapse
Affiliation(s)
- Sanyuktha Tulasidas
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Mangalore, Manipal, India
| | - Pooja Rao
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal McGill Center for Infectious Diseases, Mangalore, Manipal, India,
| | - Sevitha Bhat
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal McGill Center for Infectious Diseases, Mangalore, Manipal, India,
| | - Radhakrishna Manipura
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Mangalore, Manipal, India
| |
Collapse
|
22
|
Chong PP, Chin VK, Wong WF, Madhavan P, Yong VC, Looi CY. Transcriptomic and Genomic Approaches for Unravelling Candida albicans Biofilm Formation and Drug Resistance-An Update. Genes (Basel) 2018; 9:genes9110540. [PMID: 30405082 PMCID: PMC6266447 DOI: 10.3390/genes9110540] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 11/17/2022] Open
Abstract
Candida albicans is an opportunistic fungal pathogen, which causes a plethora of superficial, as well as invasive, infections in humans. The ability of this fungus in switching from commensalism to active infection is attributed to its many virulence traits. Biofilm formation is a key process, which allows the fungus to adhere to and proliferate on medically implanted devices as well as host tissue and cause serious life-threatening infections. Biofilms are complex communities of filamentous and yeast cells surrounded by an extracellular matrix that confers an enhanced degree of resistance to antifungal drugs. Moreover, the extensive plasticity of the C. albicans genome has given this versatile fungus the added advantage of microevolution and adaptation to thrive within the unique environmental niches within the host. To combat these challenges in dealing with C. albicans infections, it is imperative that we target specifically the molecular pathways involved in biofilm formation as well as drug resistance. With the advent of the -omics era and whole genome sequencing platforms, novel pathways and genes involved in the pathogenesis of the fungus have been unraveled. Researchers have used a myriad of strategies including transcriptome analysis for C. albicans cells grown in different environments, whole genome sequencing of different strains, functional genomics approaches to identify critical regulatory genes, as well as comparative genomics analysis between C. albicans and its closely related, much less virulent relative, C. dubliniensis, in the quest to increase our understanding of the mechanisms underlying the success of C. albicans as a major fungal pathogen. This review attempts to summarize the most recent advancements in the field of biofilm and antifungal resistance research and offers suggestions for future directions in therapeutics development.
Collapse
Affiliation(s)
- Pei Pei Chong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Malaysia, Subang Jaya, 47500 Selangor, Malaysia.
| | - Voon Kin Chin
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Malaysia, Subang Jaya, 47500 Selangor, Malaysia.
| | - Won Fen Wong
- Department of Microbiology, Faculty of Medicine, University Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Malaysia, Subang Jaya, 47500 Selangor, Malaysia.
| | - Voon Chen Yong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Malaysia, Subang Jaya, 47500 Selangor, Malaysia.
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Malaysia, Subang Jaya, 47500 Selangor, Malaysia.
| |
Collapse
|
23
|
Gulati M, Lohse MB, Ennis CL, Gonzalez RE, Perry AM, Bapat P, Arevalo AV, Rodriguez DL, Nobile CJ. In Vitro Culturing and Screening of Candida albicans Biofilms. ACTA ACUST UNITED AC 2018; 50:e60. [PMID: 29995344 DOI: 10.1002/cpmc.60] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Candida albicans is a normal member of the human microbiota that asymptomatically colonizes healthy individuals, however it is also an opportunistic pathogen that can cause severe infections, especially in immunocompromised individuals. The medical impact of C. albicans depends, in part, on its ability to form biofilms, communities of adhered cells encased in an extracellular matrix. Biofilms can form on both biotic and abiotic surfaces, such as tissues and implanted medical devices. Once formed, biofilms are highly resistant to antifungal agents and the host immune system, and can act as a protected reservoir to seed disseminated infections. Here, we present several in vitro biofilm protocols, including protocols that are optimized for high-throughput screening of mutant libraries and antifungal compounds. We also present protocols to examine specific stages of biofilm development and protocols to evaluate interspecies biofilms that C. albicans forms with interacting microbial partners. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Megha Gulati
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California
| | - Matthew B Lohse
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California.,Department of Biology, BioSynesis, Inc., San Francisco, California
| | - Craig L Ennis
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Ruth E Gonzalez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California
| | - Austin M Perry
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Priyanka Bapat
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Ashley Valle Arevalo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Diana L Rodriguez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California
| |
Collapse
|
24
|
Borges AC, Lima GDMG, Nishime TMC, Gontijo AVL, Kostov KG, Koga-Ito CY. Amplitude-modulated cold atmospheric pressure plasma jet for treatment of oral candidiasis: In vivo study. PLoS One 2018; 13:e0199832. [PMID: 29949638 PMCID: PMC6021106 DOI: 10.1371/journal.pone.0199832] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/14/2018] [Indexed: 01/14/2023] Open
Abstract
The aim of this study was to establish an effective and safe protocol for in vivo oral candidiasis treatment with atmospheric plasma jets. A novel amplitude-modulated cold atmospheric pressure plasma jet (AM-CAPPJ) device, operating with Helium, was tested. In vitro assays with Candida albicans biofilms and Vero cells were performed in order to determine the effective parameters with low cytotoxicity. After the determination of such parameters, the protocol was evaluated in experimentally induced oral candidiasis in mice. AM-CAPPJ could significantly reduce the viability of C. albicans biofilms after 5 minutes of plasma exposure when compared to the non-exposed group (p = 0.0033). After this period of exposure, high viability of Vero cells was maintained (86.33 ± 10.45%). Also, no late effects on these cells were observed after 24 and 48 hours (83.24±15.23% and 88.96±18.65%, respectively). Histological analyses revealed significantly lower occurrence of inflammatory alterations in the AM-CAPPJ group when compared to non-treated and nystatin-treated groups (p < 0.0001). Although no significant differences among the values of CFU/tongue were observed among the non-treated group and the groups treated with AM-CAPPJ or nystatin (p = 0.3201), histological analyses revealed marked reduction in candidal tissue invasion. In conclusion, these results point out to a clinical applicability of this protocol, due to the simultaneous anti-inflammatory and inhibitory effects of AM-CAPPJ with low cytotoxicity.
Collapse
Affiliation(s)
- Aline Chiodi Borges
- Department of Environmental Engineering and Oral Biopathology Graduate Program, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | - Gabriela de Morais Gouvêa Lima
- Department of Environmental Engineering and Oral Biopathology Graduate Program, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | | | - Aline Vidal Lacerda Gontijo
- Department of Environmental Engineering and Oral Biopathology Graduate Program, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | - Konstantin Georgiev Kostov
- Department of Chemistry and Physics, Guaratinguetá Faculty of Engineering, São Paulo State University (UNESP), Guaratinguetá, Brazil
| | - Cristiane Yumi Koga-Ito
- Department of Environmental Engineering and Oral Biopathology Graduate Program, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
- * E-mail:
| |
Collapse
|
25
|
Kim JJ, Reátegui E, Hopke A, Jalali F, Roushan M, Doyle PS, Irimia D. Large-scale patterning of living colloids for dynamic studies of neutrophil-microbe interactions. LAB ON A CHIP 2018; 18:1514-1520. [PMID: 29770423 PMCID: PMC5995581 DOI: 10.1039/c8lc00228b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Neutrophils are the first white blood cells to respond to microbes and to limit their invasion of the body. However, the growth of the microbes into colonies often challenges the neutrophils ability to contain them. To study the interactions between neutrophils and microbial colonies, we designed an assay for arranging microbes in clusters of controlled size (i.e. living colloids). The patterned microbes in the living colloid are mechanically trapped inside the wells and fully accessible to neutrophils. Using the assay, we studied the interactions between human neutrophils and Candida albicans and Staphylococcus aureus, two common human pathogens. We also probed the susceptibility of C. albicans colloids to caspofungin, a common antifungal drug.
Collapse
Affiliation(s)
- Jae Jung Kim
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Garcia-Perez JE, Mathé L, Humblet-Baron S, Braem A, Lagrou K, Van Dijck P, Liston A. A Framework for Understanding the Evasion of Host Immunity by Candida Biofilms. Front Immunol 2018; 9:538. [PMID: 29616035 PMCID: PMC5864854 DOI: 10.3389/fimmu.2018.00538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 03/02/2018] [Indexed: 12/31/2022] Open
Abstract
Candida biofilms are a major cause of nosocomial morbidity and mortality. The mechanism by which Candida biofilms evade the immune system remains unknown. In this perspective, we develop a theoretical framework of the three, not mutually exclusive, models, which could explain biofilm evasion of host immunity. First, biofilms may exhibit properties of immunological silence, preventing immune activation. Second, biofilms may produce immune-deviating factors, converting effective immunity into ineffective immunity. Third, biofilms may resist host immunity, which would otherwise be effective. Using a murine subcutaneous biofilm model, we found that mice infected with biofilms developed sterilizing immunity effective when challenged with yeast form Candida. Despite the induction of effective anti-Candida immunity, no spontaneous clearance of the biofilm was observed. These results support the immune resistance model of biofilm immune evasion and demonstrate an asymmetric relationship between the host and biofilms, with biofilms eliciting effective immune responses yet being resistant to immunological clearance.
Collapse
Affiliation(s)
- Josselyn E Garcia-Perez
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Lotte Mathé
- Center for Microbiology VIB - KU Leuven, Heverlee, Belgium.,Department of Materials Engineering (MTM), KU Leuven, Heverlee, Belgium
| | - Stephanie Humblet-Baron
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Annabel Braem
- Surface and Interface Engineered Materials, Department of Materials Engineering (MTM), KU Leuven, Heverlee, Belgium
| | - Katrien Lagrou
- Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium.,Clinical Department of Laboratory Medicine and National Reference Center for Mycosis, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Van Dijck
- Center for Microbiology VIB - KU Leuven, Heverlee, Belgium.,Department of Materials Engineering (MTM), KU Leuven, Heverlee, Belgium
| | - Adrian Liston
- Laboratory of Translational Immunology, VIB Center for Brain and Disease Research, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Cold atmospheric pressure plasma jet modulates Candida albicans virulence traits. CLINICAL PLASMA MEDICINE 2017. [DOI: 10.1016/j.cpme.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Modulation of Staphylococcus aureus Response to Antimicrobials by the Candida albicans Quorum Sensing Molecule Farnesol. Antimicrob Agents Chemother 2017; 61:AAC.01573-17. [PMID: 28893777 DOI: 10.1128/aac.01573-17] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/04/2017] [Indexed: 12/11/2022] Open
Abstract
In microbial biofilms, microorganisms utilize secreted signaling chemical molecules to coordinate their collective behavior. Farnesol is a quorum sensing molecule secreted by the fungal species Candida albicans and shown to play a central physiological role during fungal biofilm growth. Our pervious in vitro and in vivo studies characterized an intricate interaction between C. albicans and the bacterial pathogen Staphylococcus aureus, as these species coexist in biofilm. In this study, we aimed to investigate the impact of farnesol on S. aureus survival, biofilm formation, and response to antimicrobials. The results demonstrated that in the presence of exogenously supplemented farnesol or farnesol secreted by C. albicans in biofilm, S. aureus exhibited significantly enhanced tolerance to antimicrobials. By using gene expression studies, S. aureus mutant strains, and chemical inhibitors, the mechanism for the enhanced tolerance was attributed to upregulation of drug efflux pumps. Importantly, we showed that sequential exposure of S. aureus to farnesol generated a phenotype of high resistance to antimicrobials. Based on the presence of intracellular reactive oxygen species upon farnesol exposure, we hypothesize that antimicrobial tolerance in S. aureus may be mediated by farnesol-induced oxidative stress triggering the upregulation of efflux pumps, as part of a general stress response system. Hence, in mixed biofilms, C. albicans may influence the pathogenicity of S. aureus through acquisition of a drug-tolerant phenotype, with important therapeutic implications. Understanding interspecies signaling in polymicrobial biofilms and the specific drug resistance responses to secreted molecules may lead to the identification of novel targets for drug development.
Collapse
|
29
|
Abstract
Candida albicans is among the most prevalent fungal species of the human microbiota and asymptomatically colonizes healthy individuals. However, it is also an opportunistic pathogen that can cause severe, and often fatal, bloodstream infections. The medical impact of C. albicans typically depends on its ability to form biofilms, which are closely packed communities of cells that attach to surfaces, such as tissues and implanted medical devices. In this Review, we provide an overview of the processes involved in the formation of C. albicans biofilms and discuss the core transcriptional network that regulates biofilm development. We also consider some of the advantages that biofilms provide to C. albicans in comparison with planktonic growth and explore polymicrobial biofilms that are formed by C. albicans and certain bacterial species.
Collapse
|
30
|
Protocol for Identifying Natural Agents That Selectively Affect Adhesion, Thickness, Architecture, Cellular Phenotypes, Extracellular Matrix, and Human White Blood Cell Impenetrability of Candida albicans Biofilms. Antimicrob Agents Chemother 2017; 61:AAC.01319-17. [PMID: 28893778 DOI: 10.1128/aac.01319-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/04/2017] [Indexed: 11/20/2022] Open
Abstract
In the screening of natural plant extracts for antifungal activity, assessment of their effects on the growth of cells in suspension or in the wells of microtiter plates is expedient. However, microorganisms, including Candida albicans, grow in nature as biofilms, which are organized cellular communities with a complex architecture capable of conditioning their microenvironment, communicating, and excluding low- and high-molecular-weight molecules and white blood cells. Here, a confocal laser scanning microscopy (CLSM) protocol for testing the effects of large numbers of agents on biofilm development is described. The protocol assessed nine parameters from a single z-stack series of CLSM scans for each individual biofilm analyzed. The parameters included adhesion, thickness, formation of a basal yeast cell polylayer, hypha formation, the vertical orientation of hyphae, the hyphal bend point, pseudohypha formation, calcofluor white staining of the extracellular matrix (ECM), and human white blood cell impenetrability. The protocol was applied first to five plant extracts and derivative compounds and then to a collection of 88 previously untested plant extracts. They were found to cause a variety of phenotypic profiles, as was the case for 64 of the 88 extracts (73%). Half of the 46 extracts that did not affect biofilm thickness affected other biofilm parameters. Correlations between specific effects were revealed. The protocol will be useful not only in the screening of chemical libraries but also in the analysis of compounds with known effects and mutations.
Collapse
|
31
|
Mattana S, Alunni Cardinali M, Caponi S, Casagrande Pierantoni D, Corte L, Roscini L, Cardinali G, Fioretto D. High-contrast Brillouin and Raman micro-spectroscopy for simultaneous mechanical and chemical investigation of microbial biofilms. Biophys Chem 2017; 229:123-129. [PMID: 28684254 DOI: 10.1016/j.bpc.2017.06.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/21/2017] [Accepted: 06/21/2017] [Indexed: 12/18/2022]
Abstract
Mechanical mapping with chemical specificity of biological samples is now made possible by joint micro-Brillouin and micro-Raman measurements. In this work, thanks to the unprecedented contrast of a new tandem Fabry-Perot interferometer, we demonstrate simultaneous detection of Brillouin and Raman spectra from different Candida biofilms. Our proof-of-concept study reveals the potential of this label-free joint micro-spectroscopy technique in challenging microbiological issues. In particular, heterogeneous chemo-mechanical maps of Candida biofilms are obtained, without the need for staining or touching the sample. The correlative Raman and Brillouin investigation evidences the role of both extracellular polymeric substances and of hydration water in inducing a marked local softening of the biofilm.
Collapse
Affiliation(s)
- S Mattana
- Dipartimento di Fisica e Geologia, Università di Perugia, Via Pascoli, I-06123 Perugia, Italy.
| | - M Alunni Cardinali
- Dipartimento di Fisica e Geologia, Università di Perugia, Via Pascoli, I-06123 Perugia, Italy
| | - S Caponi
- IOM-CNR c/o Dipartimento di Fisica e Geologia, Università di Perugia, Via Pascoli, I-06123 Perugia, Italy
| | - D Casagrande Pierantoni
- Department of Pharmaceutical Sciences-Microbiology, University of Perugia, Borgo 20 Giugno 74, 06121 Perugia, Italy
| | - L Corte
- Department of Pharmaceutical Sciences-Microbiology, University of Perugia, Borgo 20 Giugno 74, 06121 Perugia, Italy
| | - L Roscini
- Department of Pharmaceutical Sciences-Microbiology, University of Perugia, Borgo 20 Giugno 74, 06121 Perugia, Italy
| | - G Cardinali
- Department of Pharmaceutical Sciences-Microbiology, University of Perugia, Borgo 20 Giugno 74, 06121 Perugia, Italy; CEMIN, Centre of Excellence on Nanostructured Innovative Materials, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - D Fioretto
- Dipartimento di Fisica e Geologia, Università di Perugia, Via Pascoli, I-06123 Perugia, Italy; CEMIN, Centre of Excellence on Nanostructured Innovative Materials, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
32
|
Nguyen Y, Fischer G. Chronic vulvovaginal candidiasis in patients using a levonorgestrel-containing intrauterine device. Australas J Dermatol 2016; 59:e39-e42. [PMID: 27957732 DOI: 10.1111/ajd.12559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/06/2016] [Indexed: 12/01/2022]
Abstract
Chronic vulvovaginal candidiasis is usually responsive to therapy with oral antifungals. We present a case series of 13 patients with this condition who were also using a levonorgestrel intrauterine system (LNG-IUS). All cases responded to ongoing oral fluconazole therapy while the LNG-IUS was in situ. The LNG-IUS was removed in six patients and of these, two experienced clinical improvement with lower fluconazole dosage requirements and three experienced complete resolution of symptoms. One remains on fluconazole 100 mg daily.
Collapse
Affiliation(s)
- Yvonne Nguyen
- Department of Dermatology, Kolling Institute, Northern Sydney Local Health District, Sydney, New South Wales, Australia.,Department of Dermatology, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Gayle Fischer
- Department of Dermatology, Kolling Institute, Northern Sydney Local Health District, Sydney, New South Wales, Australia.,Department of Dermatology, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
33
|
Abstract
Candida albicans is the most common human fungal pathogen causing diseases ranging from mucosal to systemic infections. As a commensal, C. albicans asymptomatically colonizes mucosal surfaces; however, any disruption in the host environment or under conditions of immune dysfunction, C. albicans can proliferate and invade virtually any site in the host. The ability of this highly adaptable fungal species to transition from commensal to pathogen is due to a repertoire of virulence factors. Specifically, the ability to switch morphology and form biofilms are properties central to C. albicans pathogenesis. In fact, the majority of C. albicans infections are associated with biofilm formation on host or abiotic surfaces such as indwelling medical devices, which carry high morbidity and mortality. Significantly, biofilms formed by C. albicans are inherently tolerant to antimicrobial therapy and therefore, the susceptibility of Candida biofilms to the current therapeutic agents remains low. The aim of this review is to provide an overview of C. albicans highlighting some of the diverse biofilm-associated diseases caused by this opportunistic pathogen and the animal models available to study them. Further, the classes of antifungal agents used to combat these resilient infections are discussed along with mechanisms of drug resistance.
Collapse
|
34
|
Modulation of the Substitution Pattern of 5-Aryl-2-Aminoimidazoles Allows Fine-Tuning of Their Antibiofilm Activity Spectrum and Toxicity. Antimicrob Agents Chemother 2016; 60:6483-6497. [PMID: 27550355 PMCID: PMC5075052 DOI: 10.1128/aac.00035-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 08/01/2016] [Indexed: 12/17/2022] Open
Abstract
We previously synthesized several series of compounds, based on the 5-aryl-2-aminoimidazole scaffold, that showed activity preventing the formation of Salmonella enterica serovar Typhimurium and Pseudomonas aeruginosa biofilms. Here, we further studied the activity spectrum of a number of the most active N1- and 2N-substituted 5-aryl-2-aminoimidazoles against a broad panel of biofilms formed by monospecies and mixed species of bacteria and fungi. An N1-substituted compound showed very strong activity against the biofilms formed by Gram-negative and Gram-positive bacteria and the fungus Candida albicans but was previously shown to be toxic against various eukaryotic cell lines. In contrast, 2N-substituted compounds were nontoxic and active against biofilms formed by Gram-negative bacteria and C. albicans but had reduced activity against biofilms formed by Gram-positive bacteria. In an attempt to develop nontoxic compounds with potent activity against biofilms formed by Gram-positive bacteria for application in antibiofilm coatings for medical implants, we synthesized novel compounds with substituents at both the N1 and 2N positions and tested these compounds for antibiofilm activity and toxicity. Interestingly, most of these N1-,2N-disubstituted 5-aryl-2-aminoimidazoles showed very strong activity against biofilms formed by Gram-positive bacteria and C. albicans in various setups with biofilms formed by monospecies and mixed species but lost activity against biofilms formed by Gram-negative bacteria. In light of application of these compounds as anti-infective coatings on orthopedic implants, toxicity against two bone cell lines and the functionality of these cells were tested. The N1-,2N-disubstituted 5-aryl-2-aminoimidazoles in general did not affect the viability of bone cells and even induced calcium deposition. This indicates that modulating the substitution pattern on positions N1 and 2N of the 5-aryl-2-aminoimidazole scaffold allows fine-tuning of both the antibiofilm activity spectrum and toxicity.
Collapse
|
35
|
Commensal Protection of Staphylococcus aureus against Antimicrobials by Candida albicans Biofilm Matrix. mBio 2016; 7:mBio.01365-16. [PMID: 27729510 PMCID: PMC5061872 DOI: 10.1128/mbio.01365-16] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Biofilm-associated polymicrobial infections, particularly those involving fungi and bacteria, are responsible for significant morbidity and mortality and tend to be challenging to treat. Candida albicans and Staphylococcus aureus specifically are considered leading opportunistic fungal and bacterial pathogens, respectively, mainly due to their ability to form biofilms on catheters and indwelling medical devices. However, the impact of mixed-species biofilm growth on therapy remains largely understudied. In this study, we investigated the influence of C. albicans secreted cell wall polysaccharides on the response of S. aureus to antibacterial agents in biofilm. Results demonstrated significantly enhanced tolerance for S. aureus to drugs in the presence of C. albicans or its secreted cell wall polysaccharide material. Fluorescence confocal time-lapse microscopy revealed impairment of drug diffusion through the mixed biofilm matrix. Using C. albicans mutant strains with modulated cell wall polysaccharide expression, exogenous supplementation, and enzymatic degradation, the C. albicans-secreted β-1,3-glucan cell wall component was identified as the key matrix constituent providing the bacteria with enhanced drug tolerance. Further, antibody labeling demonstrated rapid coating of the bacteria by the C. albicans matrix material. Importantly, via its effect on the fungal biofilm matrix, the antifungal caspofungin sensitized the bacteria to the drugs. Understanding such symbiotic interactions with clinical relevance between microbial species in biofilms will greatly aid in overcoming the limitations of current therapies and in defining potential new targets for treating polymicrobial infections. The fungus Candida albicans and the bacterium Staphylococcus aureus are important microbial pathogens responsible for the majority of infections in hospitalized patients and are often coisolated from a host. In this study, we demonstrated that when grown together, the fungus provides the bacterium with enhanced tolerance to antimicrobial drugs. This process was mediated by polysaccharides secreted by the fungal cell into the environment. The biofilm matrix formed by these polysaccharides prevented penetration by the drugs and provided the bacteria with protection. Importantly, we show that by inhibiting the production of the fungal polysaccharides, a specific antifungal agent indirectly sensitized the bacteria to antimicrobials. Understanding the therapeutic implications of the interactions between these two diverse microbial species will aid in overcoming the limitations of current therapies and in defining new targets for treating complex polymicrobial infections.
Collapse
|
36
|
Candida albicans Pathogenesis: Fitting within the Host-Microbe Damage Response Framework. Infect Immun 2016; 84:2724-39. [PMID: 27430274 DOI: 10.1128/iai.00469-16] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Historically, the nature and extent of host damage by a microbe were considered highly dependent on virulence attributes of the microbe. However, it has become clear that disease is a complex outcome which can arise because of pathogen-mediated damage, host-mediated damage, or both, with active participation from the host microbiota. This awareness led to the formulation of the damage response framework (DRF), a revolutionary concept that defined microbial virulence as a function of host immunity. The DRF outlines six classifications of host damage outcomes based on the microbe and the strength of the immune response. In this review, we revisit this concept from the perspective of Candida albicans, a microbial pathogen uniquely adapted to its human host. This fungus commonly colonizes various anatomical sites without causing notable damage. However, depending on environmental conditions, a diverse array of diseases may occur, ranging from mucosal to invasive systemic infections resulting in microbe-mediated and/or host-mediated damage. Remarkably, C. albicans infections can fit into all six DRF classifications, depending on the anatomical site and associated host immune response. Here, we highlight some of these diverse and site-specific diseases and how they fit the DRF classifications, and we describe the animal models available to uncover pathogenic mechanisms and related host immune responses.
Collapse
|
37
|
Abstract
The fungus Candida albicans is a major source of device-associated infection because of its capacity for biofilm formation. It is part of the natural mucosal flora and thus has access to available niches that can lead to infection. In this chapter we discuss the major properties of C. albicans biofilms and the insight that has been gleaned from their genetic determinants. Our specific areas of focus include biofilm structure and development, cell morphology and biofilm formation, biofilm-associated gene expression, the cell surface and adherence, the extracellular matrix, biofilm metabolism, and biofilm drug resistance.
Collapse
|
38
|
Bujdáková H. Management of Candida biofilms: state of knowledge and new options for prevention and eradication. Future Microbiol 2016; 11:235-51. [PMID: 26849383 DOI: 10.2217/fmb.15.139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Biofilms formed by Candida species (spp.) on medical devices represent a potential health risk. The focus of current research is searching for new options for the treatment and prevention of biofilm-associated infections using different approaches including modern nanotechnology. This review summarizes current information concerning the most relevant resistance/tolerance mechanisms to conventional drugs and a role of additional factors contributing to these phenomena in Candida spp. (mostly Candida albicans). Additionally, it provides an information update in prevention and eradication of a Candida biofilm including experiences with 'lock' therapy, potential utilization of small molecules in biomedical applications, and perspectives of using photodynamic inactivation in the control of a Candida biofilm.
Collapse
Affiliation(s)
- Helena Bujdáková
- Comenius University in Bratislava, Faculty of Natural Sciences, Department of Microbiology & Virology, Mlynská dolina, Ilkovičova 6, 842 15 Bratislava, Slovak Republic
| |
Collapse
|
39
|
Candida/Candida biofilms. First description of dual-species Candida albicans/C. rugosa biofilm. Fungal Biol 2016; 120:530-537. [PMID: 27020154 DOI: 10.1016/j.funbio.2016.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/13/2023]
Abstract
Denture liners have physical properties that favour plaque accumulation and colonization by Candida species, irritating oral tissues and causing denture stomatitis. To isolate and determine the incidence of oral Candida species in dental prostheses, oral swabs were collected from the dental prostheses of 66 patients. All the strains were screened for their ability to form biofilms; both monospecies and dual-species combinations were tested. Candida albicans (63 %) was the most frequently isolated microorganism; Candida tropicalis (14 %), Candida glabrata (13 %), Candida rugosa (5 %), Candida parapsilosis (3 %), and Candida krusei (2 %) were also detected. The XTT assay showed that C. albicans SC5314 possessed a biofilm-forming ability significantly higher (p < 0.001) than non-albicans Candida strains, after 6 h 37 °C. The total C. albicans CFU from a dual-species biofilm was less than the total CFU of a monospecies C. albicans biofilm. In contrast to the profuse hyphae verified in monospecies C. albicans biofilms, micrographies showed that the C. albicans/non-albicans Candida biofilms consisted of sparse yeast forms and profuse budding yeast cells that generated a network. These results suggested that C. albicans and the tested Candida species could co-exist in biofilms displaying apparent antagonism. The study provide the first description of C. albicans/C. rugosa mixed biofilm.
Collapse
|
40
|
Gulati M, Nobile CJ. Candida albicans biofilms: development, regulation, and molecular mechanisms. Microbes Infect 2016; 18:310-21. [PMID: 26806384 DOI: 10.1016/j.micinf.2016.01.002] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/15/2016] [Accepted: 01/15/2016] [Indexed: 01/22/2023]
Abstract
A major virulence attribute of Candida albicans is its ability to form biofilms, densely packed communities of cells adhered to a surface. These biofilms are intrinsically resistant to conventional antifungal therapeutics, the host immune system, and other environmental factors, making biofilm-associated infections a significant clinical challenge. Here, we review current knowledge on the development, regulation, and molecular mechanisms of C. albicans biofilms.
Collapse
Affiliation(s)
- Megha Gulati
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, USA
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, USA.
| |
Collapse
|
41
|
Girardot M, Imbert C. Novel strategies against Candida biofilms: interest of synthetic compounds. Future Microbiol 2015; 11:69-79. [PMID: 26673571 DOI: 10.2217/fmb.15.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A biofilm is a consortium of microbial cells that are attached to a substratum or an interface. It should be considered a reservoir that may induce serious infections. Indeed, Candidaspp. biofilms may be involved in the persistence or worsening of some chronic inflammatory diseases as well as in systemic infections, which may lead to high morbidity and mortality rates. New strategies are currently being explored, utilizing several synthetic compounds to prevent or fight these Candida biofilms. This article focuses on active synthetic compounds classified with regards to their modes of action: inhibition of early adherence phase, inhibition or control of biofilm maturation and finally elimination of already formed biofilms. Some of them show promise in fighting biofilm.
Collapse
Affiliation(s)
- Marion Girardot
- EBI, UMR CNRS 7267, Université de Poitiers, Bâtiment D1, 6 rue de la Milétrie, TSA 51115, 86073 POITIERS Cedex 9, France
| | - Christine Imbert
- EBI, UMR CNRS 7267, Université de Poitiers, Bâtiment D1, 6 rue de la Milétrie, TSA 51115, 86073 POITIERS Cedex 9, France
| |
Collapse
|
42
|
Development and In Vivo Evaluation of a Novel Histatin-5 Bioadhesive Hydrogel Formulation against Oral Candidiasis. Antimicrob Agents Chemother 2015; 60:881-9. [PMID: 26596951 DOI: 10.1128/aac.02624-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/18/2015] [Indexed: 01/09/2023] Open
Abstract
Oral candidiasis (OC), caused by the fungal pathogen Candida albicans, is the most common opportunistic infection in HIV(+) individuals and other immunocompromised populations. The dramatic increase in resistance to common antifungals has emphasized the importance of identifying unconventional therapeutic options. Antimicrobial peptides have emerged as promising candidates for therapeutic intervention due to their broad antimicrobial properties and lack of toxicity. Histatin-5 (Hst-5) specifically has exhibited potent anticandidal activity indicating its potential as an antifungal agent. To that end, the goal of this study was to design a biocompatible hydrogel delivery system for Hst-5 application. The bioadhesive hydroxypropyl methylcellulose (HPMC) hydrogel formulation was developed for topical oral application against OC. The new formulation was evaluated in vitro for gel viscosity, Hst-5 release rate from the gel, and killing potency and, more importantly, was tested in vivo in our mouse model of OC. The findings demonstrated a controlled sustained release of Hst-5 from the polymer and rapid killing ability. Based on viable C. albicans counts recovered from tongues of treated and untreated mice, three daily applications of the formulation beginning 1 day postinfection with C. albicans were effective in protection against development of OC. Interestingly, in some cases, Hst-5 was able to clear existing lesions as well as associated tissue inflammation. These findings were confirmed by histopathology analysis of tongue tissue. Coupled with the lack of toxicity as well as anti-inflammatory and wound-healing properties of Hst-5, the findings from this study support the progression and commercial feasibility of using this compound as a novel therapeutic agent.
Collapse
|
43
|
Muzny CA, Schwebke JR. Biofilms: An Underappreciated Mechanism of Treatment Failure and Recurrence in Vaginal Infections. Clin Infect Dis 2015; 61:601-6. [PMID: 25935553 PMCID: PMC4607736 DOI: 10.1093/cid/civ353] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/26/2015] [Indexed: 12/13/2022] Open
Abstract
Biofilms are microbial communities of surface-attached cells embedded in a self-produced extracellular matrix. They are of major medical significance because they decrease susceptibility to antimicrobial agents and enhance the spread of antimicrobial resistance. Biofilm-associated bacterial and fungal microorganisms have increasingly been recognized to play a role in multiple infectious diseases, particularly in their persistence and recurrence. More recently, biofilms have also been implicated in vaginal infections, notably bacterial vaginosis (BV) and vulvovaginal candidiasis (VVC), particularly in the setting of treatment failure and recurrence. The purpose of this review is to discuss the impact of biofilms on the management and treatment of BV and recurrent VVC and highlight the need for additional research and development of novel therapeutics targeting pathogenic vaginal biofilms.
Collapse
Affiliation(s)
- Christina A Muzny
- Division of Infectious Diseases, University of Alabama at Birmingham
| | - Jane R Schwebke
- Division of Infectious Diseases, University of Alabama at Birmingham
| |
Collapse
|
44
|
Chandra J, Mukherjee PK. Candida Biofilms: Development, Architecture, and Resistance. Microbiol Spectr 2015; 3:10.1128/microbiolspec.MB-0020-2015. [PMID: 26350306 PMCID: PMC4566167 DOI: 10.1128/microbiolspec.mb-0020-2015] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Indexed: 12/17/2022] Open
Abstract
Intravascular device-related infections are often associated with biofilms (microbial communities encased within a polysaccharide-rich extracellular matrix) formed by pathogens on the surfaces of these devices. Candida species are the most common fungi isolated from catheter-, denture-, and voice prosthesis-associated infections and also are commonly isolated from contact lens-related infections (e.g., fungal keratitis). These biofilms exhibit decreased susceptibility to most antimicrobial agents, which contributes to the persistence of infection. Recent technological advances have facilitated the development of novel approaches to investigate the formation of biofilms and identify specific markers for biofilms. These studies have provided extensive knowledge of the effect of different variables, including growth time, nutrients, and physiological conditions, on biofilm formation, morphology, and architecture. In this article, we will focus on fungal biofilms (mainly Candida biofilms) and provide an update on the development, architecture, and resistance mechanisms of biofilms.
Collapse
Affiliation(s)
- Jyotsna Chandra
- Center for Medical Mycology and Mycology Reference Laboratory, Department of Dermatology, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, OH 44106
| | - Pranab K Mukherjee
- Center for Medical Mycology and Mycology Reference Laboratory, Department of Dermatology, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
45
|
Kucharíková S, Vande Velde G, Himmelreich U, Van Dijck P. Candida albicans biofilm development on medically-relevant foreign bodies in a mouse subcutaneous model followed by bioluminescence imaging. J Vis Exp 2015:52239. [PMID: 25651138 DOI: 10.3791/52239] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Candida albicans biofilm development on biotic and/or abiotic surfaces represents a specific threat for hospitalized patients. So far, C. albicans biofilms have been studied predominantly in vitro but there is a crucial need for better understanding of this dynamic process under in vivo conditions. We developed an in vivo subcutaneous rat model to study C. albicans biofilm formation. In our model, multiple (up to 9) Candida-infected devices are implanted to the back part of the animal. This gives us a major advantage over the central venous catheter model system as it allows us to study several independent biofilms in one animal. Recently, we adapted this model to study C. albicans biofilm development in BALB/c mice. In this model, mature C. albicans biofilms develop within 48 hr and demonstrate the typical three-dimensional biofilm architecture. The quantification of fungal biofilm is traditionally analyzed post mortem and requires host sacrifice. Because this requires the use of many animals to perform kinetic studies, we applied non-invasive bioluminescence imaging (BLI) to longitudinally follow up in vivo mature C. albicans biofilms developing in our subcutaneous model. C. albicans cells were engineered to express the Gaussia princeps luciferase gene (gLuc) attached to the cell wall. The bioluminescence signal is produced by the luciferase that converts the added substrate coelenterazine into light that can be measured. The BLI signal resembled cell counts obtained from explanted catheters. Non-invasive imaging for quantifying in vivo biofilm formation provides immediate applications for the screening and validation of antifungal drugs under in vivo conditions, as well as for studies based on host-pathogen interactions, hereby contributing to a better understanding of the pathogenesis of catheter-associated infections.
Collapse
Affiliation(s)
- Soňa Kucharíková
- Department of Molecular Microbiology, Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, VIB, KU Leuven
| | | | - Uwe Himmelreich
- Biomedical MRI Unit/ MoSAIC, Department of Imaging & Pathology, KU Leuven
| | - Patrick Van Dijck
- Department of Molecular Microbiology, Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, VIB, KU Leuven;
| |
Collapse
|
46
|
Clinical implications of oral candidiasis: host tissue damage and disseminated bacterial disease. Infect Immun 2014; 83:604-13. [PMID: 25422264 DOI: 10.1128/iai.02843-14] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The clinical significance of polymicrobial interactions, particularly those between commensal species with high pathogenic potential, remains largely understudied. Although the dimorphic fungal species Candida albicans and the bacterium Staphylococcus aureus are common cocolonizers of humans, they are considered leading opportunistic pathogens. Oral candidiasis specifically, characterized by hyphal invasion of oral mucosal tissue, is the most common opportunistic infection in HIV(+) and immunocompromised individuals. In this study, building on our previous findings, a mouse model was developed to investigate whether the onset of oral candidiasis predisposes the host to secondary staphylococcal infection. The findings demonstrated that in mice with oral candidiasis, subsequent exposure to S. aureus resulted in systemic bacterial infection with high morbidity and mortality. Histopathology and scanning electron microscopy of tongue tissue from moribund animals revealed massive C. albicans hyphal invasion coupled with S. aureus deep tissue infiltration. The crucial role of hyphae in the process was demonstrated using a non-hypha-producing and a noninvasive hypha-producing mutant strains of C. albicans. Further, in contrast to previous findings, S. aureus dissemination was aided but not contingent upon the presence of the Als3p hypha-specific adhesion. Importantly, impeding development of mucosal C. albicans infection by administering antifungal fluconazole therapy protected the animals from systemic bacterial disease. The combined findings from this study demonstrate that oral candidiasis may constitute a risk factor for disseminated bacterial disease warranting awareness in terms of therapeutic management of immunocompromised individuals.
Collapse
|
47
|
Borghi E, Romagnoli S, Fuchs BB, Cirasola D, Perdoni F, Tosi D, Braidotti P, Bulfamante G, Morace G, Mylonakis E. Correlation between Candida albicans biofilm formation and invasion of the invertebrate host Galleria mellonella. Future Microbiol 2014; 9:163-73. [PMID: 24571071 DOI: 10.2217/fmb.13.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The aim of our study was to investigate whether biofilm production by Candida albicans clinical isolates could be a hallmark of virulence in vivo. MATERIALS & METHODS Twenty clinical isolates of C. albicans were examined via histological studies on larvae infected with various fungal doses (from 10(3) to 10(5) CFU/larva) of biofilm producer and nonproducer strains. RESULTS The poor prognostic role of infection due to a biofilm-producing isolate was confirmed by the Wald test (hazard ratio: 2.63; 95% CI: 2.03-3.41). Histological examinations at 24 h showed a strong innate immune response, with evidence of melanization for both infection groups. However, at 48 h, we found huge differences in filamentation and tissue invasion capability between biofilm nonproducing and producing isolates, the latter being highly organized into biofilm and invading the larval intestinal tract. Invasion corroborated survival data. CONCLUSION The histological results demonstrate that the production of biofilm could enhance the invasiveness of C. albicans.
Collapse
Affiliation(s)
- Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, via di Rudinì, 8 - 20142 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tsai PW, Chen YT, Yang CY, Chen HF, Tan TS, Lin TW, Hsieh WP, Lan CY. The role of Mss11 in Candida albicans biofilm formation. Mol Genet Genomics 2014; 289:807-19. [PMID: 24752399 DOI: 10.1007/s00438-014-0846-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/22/2014] [Indexed: 01/08/2023]
Abstract
Candida albicans is an opportunistic human pathogen that can form a biofilm on biotic or inert surfaces such as epithelia and clinical devices. In this study, we examine the formation of C. albicans biofilm by establishing a key gene-centered network based on protein-protein interaction (PPI) and gene expression datasets. Starting from C. albicans Cph1 and Efg1, transcription factors associated with morphogenesis of biofilm formation, a network elucidates the complex cellular process and predicts potential unknown components related to biofilm formation. Subsequently, we analyzed the functions of Mss11 among these identified proteins to test the efficiency of the proposed computational approach. MSS11-deleted mutants were compared with a wild-type strain, indicating that the mutant is defective in forming a mature biofilm and partially attenuates the virulence of C. albicans in an infected mouse model. Finally, a DNA microarray analysis was conducted to identify the potential target genes of C. albicans Mss11. The findings of this study clarify complex gene or protein interaction during the biofilm formation process of C. albicans, supporting the application of a systems biology approach to study fungal pathogenesis.
Collapse
Affiliation(s)
- Pei-Wen Tsai
- Institute of Molecular and Cellular Biology, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, 30013, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Candida Biofilm: Clinical Implications of Recent Advances in Research. CURRENT FUNGAL INFECTION REPORTS 2014. [DOI: 10.1007/s12281-014-0176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
50
|
Demuyser L, Jabra-Rizk MA, Van Dijck P. Microbial cell surface proteins and secreted metabolites involved in multispecies biofilms. Pathog Dis 2014; 70:219-30. [PMID: 24376219 DOI: 10.1111/2049-632x.12123] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/16/2013] [Accepted: 12/16/2013] [Indexed: 12/15/2022] Open
Abstract
A considerable number of infectious diseases involve multiple microbial species coexisting and interacting in a host. Only recently however the impact of these polymicrobial diseases has been appreciated and investigated. Often, the causative microbial species are embedded in an extracellular matrix forming biofilms, a form of existence that offers protection against chemotherapeutic agents and host immune defenses. Therefore, recent efforts have focused on developing novel therapeutic strategies targeting biofilm-associated polymicrobial infections, a task that has proved to be challenging. One promising approach to inhibit the development of such complex infections is to impede the interactions between the microbial species via inhibition of adhesion. To that end, studies have focused on identifying specific cell wall adhesins and receptors involved in the interactions between the various bacterial species and the most pathogenic human fungal species Candida albicans. This review highlights the important findings from these studies and describes the available tools and techniques that have provided insights into the role of secreted molecules orchestrating microbial interactions in biofilms. Specifically, we focus on the interactions that take place in oral biofilms and the implications of these interactions on oral health and therapeutic strategies.
Collapse
Affiliation(s)
- Liesbeth Demuyser
- VIB Department of Molecular Microbiology, KU Leuven, Leuven, Belgium; Laboratory of Molecular Cell Biology, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|