151
|
The Antifungal and Antibiofilm Activities of Caffeine against Candida albicans on Polymethyl Methacrylate Denture Base Material. Biomedicines 2022; 10:biomedicines10092078. [PMID: 36140179 PMCID: PMC9495344 DOI: 10.3390/biomedicines10092078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background: In this study, the effect of pure caffeine was established against Candida albicans (C. albicans) using different microbiological techniques. Methods: Broth microdilution and colony forming units (CFUs) assays were used to detect the minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC). The Live/Dead fluorescent dyes were implemented to determine the yeast viability. Polymethyl methacrylate acrylic resin (PMMA) discs were prepared to evaluate caffeine’s effects against adherent C. albicans using microplate reader, CFUs, and scanning electron microscope (SEM). Results: caffeine’s MIC was detected around 30 mg/mL, while the MFC was considered at 60 mg/mL. In an agar-well diffusion test, the inhibition zones were wider in caffeine groups. The Live/Dead viability test verified caffeine’s antifungal effects. The optical density of the adherent C. albicans on PMMA discs were lower at 620 nm or 410 nm in caffeine groups. CFU count was also reduced by caffeine treatments. SEM revealed the lower adherent C. albicans count in caffeine groups. The effect of caffeine was dose-dependent at which the 60 mg/mL dose demonstrated the most prominent effect. Conclusion: The study reinforced caffeine’s antifungal and antibiofilm properties and suggested it as an additive, or even an alternative, disinfectant solution for fungal biofilms on denture surfaces.
Collapse
|
152
|
Lycosin-II Exhibits Antifungal Activity and Inhibits Dual-Species Biofilm by Candida albicans and Staphylococcus aureus. J Fungi (Basel) 2022; 8:jof8090901. [PMID: 36135626 PMCID: PMC9504746 DOI: 10.3390/jof8090901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
The increase and dissemination of antimicrobial resistance is a global public health issue. To address this, new antimicrobial agents have been developed. Antimicrobial peptides (AMPs) exhibit a wide range of antimicrobial activities against pathogens, including bacteria and fungi. Lycosin-II, isolated from the venom of the spider Lycosa singoriensis, has shown antibacterial activity by disrupting membranes. However, the mode of action of Lycosin-II and its antifungal activity have not been clearly described. Therefore, we confirmed that Lycosin-II showed antifungal activity against Candida albicans (C. albicans). To investigate the mode of action, membrane-related assays were performed, including an evaluation of C. albicans membrane depolarization and membrane integrity after exposure to Lycosin-II. Our results indicated that Lycosin-II damaged the C. albicans membrane. Additionally, Lycosin-II induced oxidative stress through the generation of reactive oxygen species (ROS) in C. albicans. Moreover, Lycosin-II exhibited an inhibitory effect on dual-species biofilm formation by C. albicans and Staphylococcus aureus (S. aureus), which are the most co-isolated fungi and bacteria. These results revealed that Lycosin-II can be utilized against C. albicans and dual-species strain infections.
Collapse
|
153
|
Monteiro DR, Pessan JP. Nanocarriers of antifungal drugs: possible strategies to overcome antimicrobial resistance. Future Microbiol 2022; 17:1199-1202. [PMID: 35984414 DOI: 10.2217/fmb-2022-0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Douglas R Monteiro
- Postgraduate Program in Health Sciences, University of Western São Paulo (UNOESTE), Presidente Prudente/São Paulo, 19050-920, Brazil.,Department of Preventive & Restorative Dentistry, São Paulo State University (Unesp), School of Dentistry, Araçatuba, Araçatuba/São Paulo, 16015-050, Brazil
| | - Juliano P Pessan
- Department of Preventive & Restorative Dentistry, São Paulo State University (Unesp), School of Dentistry, Araçatuba, Araçatuba/São Paulo, 16015-050, Brazil
| |
Collapse
|
154
|
Atiencia-Carrera MB, Cabezas-Mera FS, Vizuete K, Debut A, Tejera E, Machado A. Evaluation of the biofilm life cycle between Candida albicans and Candida tropicalis. Front Cell Infect Microbiol 2022; 12:953168. [PMID: 36061861 PMCID: PMC9433541 DOI: 10.3389/fcimb.2022.953168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Candida tropicalis is an emergent pathogen with a high rate of mortality associated with its biofilm formation. Biofilm formation has important repercussions on the public health system. However, little is still known about its biofilm life cycle. The present study analyzed the biofilm life cycle of Candida albicans and C. tropicalis during various timepoints (24, 48, 72, and 96 h) through biomass assays, colony-forming unit (CFU) counting, and epifluorescence and scanning electron microscopies. Our results showed a significant difference between C. albicans and C. tropicalis biofilms in each biomass and viability assay. All-time samples in the biomass and viability assays confirmed statistical differences between the Candida species through pairwise Wilcoxon tests (p < 0.05). C. albicans demonstrated a lower biomass growth but reached nearly the same level of C. tropicalis biomass at 96 h, while the CFU counting assays exhibited a superior number of viable cells within the C. tropicalis biofilm. Statistical differences were also found between C. albicans and C. tropicalis biofilms from 48- and 72-h microscopies, demonstrating C. tropicalis with a higher number of total cells within biofilms and C. albicans cells with a superior cell area and higher matrix production. Therefore, the present study proved the higher biofilm production of C. tropicalis.
Collapse
Affiliation(s)
- María Belén Atiencia-Carrera
- Universidad San Francisco de Quito (USFQ), Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Quito, Ecuador
| | - Fausto Sebastián Cabezas-Mera
- Universidad San Francisco de Quito (USFQ), Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Quito, Ecuador
| | - Karla Vizuete
- Center of Nanoscience and Nanotechnology, Universidad de las Fuerzas Armadas (ESPE), Sangolquí, Ecuador
| | - Alexis Debut
- Center of Nanoscience and Nanotechnology, Universidad de las Fuerzas Armadas (ESPE), Sangolquí, Ecuador
| | - Eduardo Tejera
- Facultad de Ingeniería y Ciencias Agropecuarias Aplicadas, Grupo de Bioquimioinformática, Universidad de Las Américas (UDLA), Quito, Ecuador
- *Correspondence: António Machado, ; Eduardo Tejera,
| | - António Machado
- Universidad San Francisco de Quito (USFQ), Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Quito, Ecuador
- *Correspondence: António Machado, ; Eduardo Tejera,
| |
Collapse
|
155
|
Sahin F, Celik N, Ceylan A, Ruzi M, Onses MS. One-step Green Fabrication of Antimicrobial Surfaces via In Situ Growth of Copper Oxide Nanoparticles. ACS OMEGA 2022; 7:26504-26513. [PMID: 35936466 PMCID: PMC9352341 DOI: 10.1021/acsomega.2c02540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
Microorganisms such as pathogenic bacteria, fungi, and viruses pose a serious threat to human health and society. Surfaces are one of the major pathways for the transmission of infectious diseases. Therefore, imparting antipathogenic properties to these surfaces is significant. Here, we present a rapid, one-step approach for practical fabrication of antimicrobial and antifungal surfaces using an eco-friendly and low-cost reducing agent, the extract of Cedrus libani. Copper oxide nanoparticles were grown in situ on the surface of print paper and fabric in the presence of the copper salt and extract, without the use of any additional chemicals. The morphology and composition of the grown nanoparticles were characterized using field emission scanning electron microscopy, energy-dispersive X-ray spectroscopy, X-ray photoelectron spectroscopy, and X-ray diffraction techniques. The analysis revealed that the grown particles consist of mainly spherical CuO nanoparticles with an average size of ∼14 nm and its clusters with an average size of ∼700 nm. The in situ growth process enables strong bonding of the nanoparticles to the surface, resulting in enhanced durability against wear and tear. Moreover, the fabricated surface shows excellent growth inhibition ability and bactericidal activity against both gram-negative and gram-positive bacteria, Escherichia coli and Staphylococcus aureus, as well as antifungal activity against Candida albicans, a common pathogenic fungus. The ability to grow copper oxide nanoparticles on different surfaces paves the way for a range of applications in wound dressings, masks, and protective medical equipment.
Collapse
Affiliation(s)
- Furkan Sahin
- Nanotechnology
Application and Research Center, ERNAM—Erciyes
University, Kayseri38039, Turkey
| | - Nusret Celik
- Nanotechnology
Application and Research Center, ERNAM—Erciyes
University, Kayseri38039, Turkey
- Department
of Materials Science and Engineering, Erciyes
University, Kayseri38039, Turkey
| | - Ahmet Ceylan
- Faculty
of Pharmacy, Erciyes University, Kayseri38039, Turkey
| | - Mahmut Ruzi
- Nanotechnology
Application and Research Center, ERNAM—Erciyes
University, Kayseri38039, Turkey
| | - M. Serdar Onses
- Nanotechnology
Application and Research Center, ERNAM—Erciyes
University, Kayseri38039, Turkey
- Department
of Materials Science and Engineering, Erciyes
University, Kayseri38039, Turkey
- UNAM-Institute
of Materials Science and Nanotechnology, Bilkent University, Ankara06800, Turkey
| |
Collapse
|
156
|
Jakab Á, Kovács F, Balla N, Tóth Z, Ragyák Á, Sajtos Z, Csillag K, Nagy-Köteles C, Nemes D, Bácskay I, Pócsi I, Majoros L, Kovács ÁT, Kovács R. Physiological and transcriptional profiling of surfactin exerted antifungal effect against Candida albicans. Biomed Pharmacother 2022; 152:113220. [PMID: 35671583 DOI: 10.1016/j.biopha.2022.113220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 11/28/2022] Open
Abstract
Given the risk of Candida albicans overgrowth in the gut, novel complementary therapies should be developed to reduce fungal dominancy. This study highlights the antifungal characteristics of a Bacillus subtilis-derived secondary metabolite, surfactin with high potential against C. albicans. Surfactin inhibited the growth of C. albicans following a 1-hour exposure, in addition to reduced adhesion and morphogenesis. Specifically, surfactin did not affect the level of reactive oxygen species but increased the level of reduced glutathione. Surprisingly, ethanol production was increased following 2 h of surfactin exposure. Surfactin treatment caused a significant reduction in intracellular iron, manganese and zinc content compared to control cells, whereas the level of copper was not affected. Alongside these physiological properties, surfactin also enhanced fluconazole efficacy. To gain detailed insights into the surfactin-related effects on C. albicans, genome-wide gene transcription analysis was performed. Surfactin treatment resulted in 1390 differentially expressed genes according to total transcriptome sequencing (RNA-Seq). Of these, 773 and 617 genes with at least a 1.5-fold increase or decrease in transcription, respectively, were selected for detailed investigation. Several genes involved in morphogenesis or related to metabolism (e.g., glycolysis, ethanol and fatty acid biosynthesis) were down-regulated. Moreover, surfactin decreased the expression of ERG1, ERG3, ERG9, ERG10 and ERG11 involved in ergosterol synthesis, whereas genes associated with ribosome biogenesis and iron metabolism and drug transport-related genes were up-regulated. Our data demonstrate that surfactin significantly influences the physiology and gene transcription of C. albicans, and could contribute to the development of a novel innovative complementary therapy.
Collapse
Affiliation(s)
- Ágnes Jakab
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary; Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Noémi Balla
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Zoltán Tóth
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Ágota Ragyák
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary; Department of Inorganic and Analytical Chemistry, Agilent Atomic Spectroscopy Partner Laboratory, University of Debrecen, Debrecen, Hungary
| | - Zsófi Sajtos
- Department of Inorganic and Analytical Chemistry, Agilent Atomic Spectroscopy Partner Laboratory, University of Debrecen, Debrecen, Hungary
| | - Kinga Csillag
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Csaba Nagy-Köteles
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Dániel Nemes
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Ildikó Bácskay
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - István Pócsi
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - László Majoros
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ákos T Kovács
- Bacterial Interactions and Evolution Group, DTU Bioengineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Renátó Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
157
|
Wang S, Wang P, Liu J, Yang C, Wang Q, Su M, Wei M, Gu L. Antibiofilm Activity of Essential Fatty Acids Against Candida albicans from Vulvovaginal Candidiasis and Bloodstream Infections. Infect Drug Resist 2022; 15:4181-4193. [PMID: 35946033 PMCID: PMC9357398 DOI: 10.2147/idr.s373991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/28/2022] [Indexed: 12/20/2022] Open
Abstract
Purpose Methods Results Conclusion
Collapse
Affiliation(s)
- Shuai Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Peng Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jun Liu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Chunxia Yang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Qiangyi Wang
- Department of Clinical Laboratory, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mingze Su
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Ming Wei
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Ming Wei; Li Gu, Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, People’s Republic of China, Tel +86-10-85231513, Email ;
| | - Li Gu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
158
|
Marena GD, Ramos MADS, Carvalho GC, de Lima LC, Nascimento ALCSD, Sábio RM, Rodero CF, Spósito L, Bauab TM, Chorilli M. Development and characterization of an amphotericin B - loaded nanoemulsion applied to Candida auris biofilms control. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
159
|
Wang S, Zhang Y, Mandlaa, Sun Z, Chen Z. Properties and mechanism of the antimicrobial peptide APT produced by Lactobacillus ALAC-4. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
160
|
Gómez-Casanova N, Torres-Cano A, Elias-Rodriguez AX, Lozano T, Ortega P, Gómez R, Pérez-Serrano J, Copa-Patiño JL, Heredero-Bermejo I. Inhibition of Candida glabrata Biofilm by Combined Effect of Dendritic Compounds and Amphotericin. Pharmaceutics 2022; 14:pharmaceutics14081604. [PMID: 36015230 PMCID: PMC9416558 DOI: 10.3390/pharmaceutics14081604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
In the last decade, Candida glabrata has become an important emerging opportunistic pathogen not only because of the increase in nosocomial infections frequency but also because of its ability to form biofilms and its innate resistance to commercial antifungals. These characteristics make this pathogen a major problem in hospital settings, including problems regarding equipment, and in immunosuppressed patients, who are at high risk for candidemia. Therefore, there is an urgent need for the development of and search for new antifungal drugs. In this study, the efficacy of two dendritic wedges with 4-phenyl butyric acid (PBA) at the focal point and cationic charges on the surface ArCO2G2(SNMe3I)4 (1) and ArCO2G3(SNMe3I)8 (2) was studied against C. glabrata strain to inhibit the formation of biofilms and eliminate established biofilm. For this, MBIC (minimum biofilm inhibitory concentration), MBDC (minimum biofilm damaging concentrations), as well as MFCB (minimum fungicidal concentration in biofilm) and MBEC (minimum biofilm eradicating concentration) were determined. In addition, different combinations of dendrons and amphotericin B were tested to study possible synergistic effects. On the other hand, cytotoxicity studies were performed. C. glabrata cells and biofilm structure were visualized by confocal microscopy. ArCO2G2(SNMe3I)4 (1) and ArCO2G3(SNMe3I)8 (2) dendrons showed both an MBIC of 8 mg/L and a MBDC of 32 mg/L and 64 mg/L, respectively. These dendrons managed to eradicate the entirety of an established biofilm. In combination with the antifungal amphotericin, it was possible to prevent the generation of biofilms and eradicate established biofilms at lower concentrations than those required individually for each compound at these conditions.
Collapse
Affiliation(s)
- Natalia Gómez-Casanova
- Department of Biomedicine and Biotechnology, Faculty of Pharmacy, University of Alcalá, 28871 Alcalá de Henares, Spain; (N.G.-C.); (A.T.-C.); (A.X.E.-R.); (J.P.-S.); (J.L.C.-P.)
| | - Alba Torres-Cano
- Department of Biomedicine and Biotechnology, Faculty of Pharmacy, University of Alcalá, 28871 Alcalá de Henares, Spain; (N.G.-C.); (A.T.-C.); (A.X.E.-R.); (J.P.-S.); (J.L.C.-P.)
| | - Alba Xiaohe Elias-Rodriguez
- Department of Biomedicine and Biotechnology, Faculty of Pharmacy, University of Alcalá, 28871 Alcalá de Henares, Spain; (N.G.-C.); (A.T.-C.); (A.X.E.-R.); (J.P.-S.); (J.L.C.-P.)
| | - Tania Lozano
- Department of Organic and Inorganic Chemistry, Faculty of Pharmacy, Research Institute in Chemistry “Andrés M. del Río” (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain; (T.L.); (P.O.); (R.G.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain and Institute “Ramón y Cajal” for Health Research (IRYCIS), 28029 Madrid, Spain
| | - Paula Ortega
- Department of Organic and Inorganic Chemistry, Faculty of Pharmacy, Research Institute in Chemistry “Andrés M. del Río” (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain; (T.L.); (P.O.); (R.G.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain and Institute “Ramón y Cajal” for Health Research (IRYCIS), 28029 Madrid, Spain
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry, Faculty of Pharmacy, Research Institute in Chemistry “Andrés M. del Río” (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain; (T.L.); (P.O.); (R.G.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain and Institute “Ramón y Cajal” for Health Research (IRYCIS), 28029 Madrid, Spain
| | - Jorge Pérez-Serrano
- Department of Biomedicine and Biotechnology, Faculty of Pharmacy, University of Alcalá, 28871 Alcalá de Henares, Spain; (N.G.-C.); (A.T.-C.); (A.X.E.-R.); (J.P.-S.); (J.L.C.-P.)
| | - José Luis Copa-Patiño
- Department of Biomedicine and Biotechnology, Faculty of Pharmacy, University of Alcalá, 28871 Alcalá de Henares, Spain; (N.G.-C.); (A.T.-C.); (A.X.E.-R.); (J.P.-S.); (J.L.C.-P.)
| | - Irene Heredero-Bermejo
- Department of Biomedicine and Biotechnology, Faculty of Pharmacy, University of Alcalá, 28871 Alcalá de Henares, Spain; (N.G.-C.); (A.T.-C.); (A.X.E.-R.); (J.P.-S.); (J.L.C.-P.)
- Correspondence:
| |
Collapse
|
161
|
Bezerra LP, Silva AF, Santos-Oliveira R, Alencar LM, Amaral JL, Neto NA, Silva RG, Belém MO, de Andrade CR, Oliveira JT, Freitas CD, Souza PF. Combined antibiofilm activity of synthetic peptides and antifungal drugs against Candida spp. Future Microbiol 2022; 17:1133-1146. [PMID: 35880557 DOI: 10.2217/fmb-2022-0053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Introduction: Candida krusei and Candida albicans are biofilm-forming drug-resistant yeasts that cause bloodstream infections that can lead to death. Materials & methods: nystatin and itraconazole were combined with two synthetic peptides, PepGAT and PepKAA, to evaluate the synergistic effect against Candida biofilms. Additionally, scanning electron and fluorescence microscopies were employed to understand the mechanism behind the synergistic activity. Results: Peptides enhanced the action of drugs to inhibit the biofilm formation of C. krusei and C. albicans and the degradation of mature biofilms of C. krusei. In combination with antifungal drugs, peptides' mechanism of action involved cell wall and membrane damage and overproduction of reactive oxygen species. Additionally, in combination, the peptides reduced the toxicity of drugs to red blood cells. Conclusion: These results reveal that the synthetic peptides enhanced the antibiofilm activity of drugs, in addition to reducing their toxicity. Thus, these peptides have strong potential as adjuvants and to decrease the toxicity of drugs.
Collapse
Affiliation(s)
- Leandro P Bezerra
- Department of Biochemistry & Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Ayrles Fb Silva
- Department of Physic, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Ralph Santos-Oliveira
- Nanoradiopharmaceuticals & Radiopharmacy, Zona Oeste State University, Brazilian Nuclear Energy Commission, Rio de Janeiro, Rio de Janeiro, 23070200, Brazil
| | - Luciana Mr Alencar
- Department of Physics, Laboratory of Biophysics & Nanosystems, Federal University of Maranhao, São Luís, Maranhão, 65080-805, Brazil
| | - Jackson L Amaral
- Department of Biochemistry & Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil.,Department of Physic, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Nilton As Neto
- Department of Biochemistry & Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Rafael Gg Silva
- Department of Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Mônica O Belém
- Laboratory of Translational Research, Christus University Center, Fortaleza, Ceará, 60192, Brazil
| | - Claudia R de Andrade
- Laboratory of Translational Research, Christus University Center, Fortaleza, Ceará, 60192, Brazil
| | - Jose Ta Oliveira
- Department of Biochemistry & Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Cleverson Dt Freitas
- Department of Biochemistry & Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil
| | - Pedro Fn Souza
- Department of Biochemistry & Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, 60451, Brazil.,Drug Research & Development Center, Department of Physiology & Pharmacology, Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil
| |
Collapse
|
162
|
Xie Y, Hua H, Zhou P. Magnolol as a potent antifungal agent inhibits Candida albicans virulence factors via the PKC and Cek1 MAPK signaling pathways. Front Cell Infect Microbiol 2022; 12:935322. [PMID: 35937692 PMCID: PMC9355038 DOI: 10.3389/fcimb.2022.935322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Magnolol, a lignin compound extracted from Magnolia officinalis Cortex, has been found to have prominent antifungal effects against Candida albicans. However, the specific mechanism still remains unclear. Therefore, this study aimed to further explore the inhibition mechanism of magnolol against Candida albicans virulence factors and the related signaling pathways. By an XTT reduction assay, a hyphal formation assay, confocal laser scanning microscopy, transmission electron microscopy, a calcofluor white staining assay, and a cell wall β-glucan quantitative detection assay, we evaluated the inhibitory effects of magnolol against the adhesion, hyphal formation, biofilm viability, biofilm spatial structure, and cell wall ultrastructure of Candida albicans. Moreover, by RNA sequencing and qRT-PCR, we confirmed the effects of magnolol in inhibiting the gene expression of Candida albicans virulence factors and the related signaling pathways. The results revealed that the adhesion and hyphal formation of Candida albicans were inhibited significantly by magnolol. The viability and spatial structures of Candida albicans biofilms were further weakened. Candida albicans ultrastructure showed partial thinning of cell walls and even rupture, with cytoplasmic leakage. The cell wall intergrity and β-glucan content were also radically reduced. Moreover, magnolol caused significant inhibition of the expression of Candida albicans adhesion, invasion, hyphal formation, biofilm formation, β-1,3-glucan synthesis, and hydrolase secretion-related genes, including ALS1, ALS3, EFG1, EAP1, FKS1, FKS2, PLB2, and SAP2. Furthermore, the PKC pathway-related genes (RHO1, PKC1, BCK1, MKK2, MKC1) and Cek1 pathway-related genes (CDC42, CST20, STE11, HST7, CEK1) were also significantly downregulated, indicating that the inhibition of magnolol against Candida albicans virulence factors might be related to PKC and Cek1 MAPK signaling pathways. In conclusion, the findings of this study confirmed the inhibition mechanism of magnolol against Candida albicans virulence factors, which might be related to PKC and Cek1 MAPK pathways, thus laying the theoretical foundation for its clinical antifungal applications.
Collapse
Affiliation(s)
| | - Hong Hua
- *Correspondence: Peiru Zhou, ; Hong Hua,
| | - Peiru Zhou
- *Correspondence: Peiru Zhou, ; Hong Hua,
| |
Collapse
|
163
|
Molecular Mapping of Antifungal Mechanisms Accessing Biomaterials and New Agents to Target Oral Candidiasis. Int J Mol Sci 2022; 23:ijms23147520. [PMID: 35886869 PMCID: PMC9320712 DOI: 10.3390/ijms23147520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Oral candidiasis has a high rate of development, especially in immunocompromised patients. Immunosuppressive and cytotoxic therapies in hospitalized HIV and cancer patients are known to induce the poor management of adverse reactions, where local and systemic candidiasis become highly resistant to conventional antifungal therapy. The development of oral candidiasis is triggered by several mechanisms that determine oral epithelium imbalances, resulting in poor local defense and a delayed immune system response. As a result, pathogenic fungi colonies disseminate and form resistant biofilms, promoting serious challenges in initiating a proper therapeutic protocol. Hence, this study of the literature aimed to discuss possibilities and new trends through antifungal therapy for buccal drug administration. A large number of studies explored the antifungal activity of new agents or synergic components that may enhance the effect of classic drugs. It was of significant interest to find connections between smart biomaterials and their activity, to find molecular responses and mechanisms that can conquer the multidrug resistance of fungi strains, and to transpose them into a molecular map. Overall, attention is focused on the nanocolloids domain, nanoparticles, nanocomposite synthesis, and the design of polymeric platforms to satisfy sustained antifungal activity and high biocompatibility with the oral mucosa.
Collapse
|
164
|
Flavonoid-Rich Fractions of Bauhinia holophylla Leaves Inhibit Candida albicans Biofilm Formation and Hyphae Growth. PLANTS 2022; 11:plants11141796. [PMID: 35890430 PMCID: PMC9322443 DOI: 10.3390/plants11141796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
This study evaluated the effect of the extract and fractions of Bauhinia holophylla on Candida albicans planktonic growth, biofilm formation, mature biofilm, and hyphae growth. Three C. albicans strains (SC5314, ATCC 18804, and ATCC 10231) were tested. The crude extract and the fractions were obtained by exhaustive percolation and liquid–liquid partition, respectively. Phytochemical analyses of B. holophylla extract and fractions were performed using high-performance liquid chromatography coupled with a diode-array detector and mass spectrometry (HPLC-DAD-MS). A microdilution assay was used to evaluate the effect of the B. holophylla extract and fractions on C. albicans planktonic growth, and crystal violet staining was used to measure the total biomass of the biofilm. Hyphae growth was analyzed using light microscopy. Thirteen flavonoids were identified, with a predominance of the flavonol-3-O-glycoside type based on quercetin, myricetin, and kaempferol. Flavonoid-rich fractions of B. holophylla leaves displayed antifungal activity and inhibited both biofilm formation and hyphae growth in all the tested strains, but were not effective on C. albicans planktonic growth and mature biofilm. This study indicates that flavonoid-rich fractions from B. holophylla leaves interfere with the virulence of Candida species and support the use of Bauhinia spp. in folk medicine to treat infections.
Collapse
|
165
|
Citrus Essential Oils: a Treasure Trove of Antibiofilm Agent. Appl Biochem Biotechnol 2022; 194:4625-4638. [PMID: 35779176 DOI: 10.1007/s12010-022-04033-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 11/02/2022]
Abstract
Biofilms are groups of adherent cell communities that cohere to the biotic and abiotic surfaces with the help of extracellular polymeric substances (EPS). EPS allow bacteria to form a biofilm that facilitates their binding to biotic and abiotic surfaces and provides resistance to the host immune responses and to antibiotics. There are efforts that have led to the development of natural compounds that can overcome this biofilm-mediated resistance. Essential oils (EOs) are a unique mixture of compounds that plays a key role in preventing the development of biofilm. The present overview focusses on the role of various types of citrus essential oils in acting against the biofilm, and the antibiofilm properties of natural compounds that may show an avenue to treat the multidrug-resistant bacteria.
Collapse
|
166
|
Rao KH, Paul S, Natarajan K, Ghosh S. N-acetylglucosamine kinase, Hxk1is a multifaceted metabolic enzyme in model pathogenic yeast Candida albicans. Microbiol Res 2022; 263:127146. [DOI: 10.1016/j.micres.2022.127146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
|
167
|
Holzknecht J, Dubrac S, Hedtrich S, Galgóczy L, Marx F. Small, Cationic Antifungal Proteins from Filamentous Fungi Inhibit Candida albicans Growth in 3D Skin Infection Models. Microbiol Spectr 2022; 10:e0029922. [PMID: 35499318 PMCID: PMC9241769 DOI: 10.1128/spectrum.00299-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
The emerging resistance of human-pathogenic fungi to antifungal drugs urges the development of alternative therapeutic strategies. The small, cationic antifungal proteins (AFPs) from filamentous ascomycetes represent promising candidates for next-generation antifungals. These bio-molecules need to be tested for tolerance in the host and efficacy against fungal pathogens before they can be safely applied in humans. Testing of the efficacy and possible adverse effects of new drug candidates in three-dimensional (3D) human-cell based models represents an advantageous alternative to animal experiments. In, this study, as a proof-of-principle, we demonstrate the usefulness of 3D skin infection models for screening new antifungal drug candidates for topical application. We established a cutaneous infection with the opportunistic human-pathogenic yeast Candida albicans in a commercially available 3D full-thickness (FT) skin model to test the curative potential of distinct AFPs from Penicillium chrysogenum (PAFopt, PAFB, and PAFC) and Neosartorya (Aspergillus) fischeri (NFAP2) in vitro. All tested AFPs were comparably well tolerated by the skin models. The infected 3D models exhibited reduced epidermal permeability barriers, allowing C. albicans to colonize the epidermal and dermal layers, and showed increased secretion of the pro-inflammatory cytokine IL-6 and the chemokine IL-8. AFP treatment diminished the fungal burden and penetration depth of C. albicans in the infected models. The epidermal permeability barrier was restored and the secretion of IL-8 was decreased following AFP treatment. In summary, our study proves that the tested AFPs exhibit antifungal potential against cutaneous C. albicans infection in a 3D FT skin model. IMPORTANCE Candida albicans represents one of the most prevalent opportunistic fungal pathogens, causing superficial skin and mucosal infections in humans with certain predisposing health conditions and life-threatening systemic infections in immunosuppressed patients. The emerging drug resistance of this human-pathogenic yeast and the limited number of antifungal drugs for prevention and treatment of infections urgently demands the identification of new antifungal compounds with novel mechanisms of action. Small, cationic antifungal proteins (AFPs) from filamentous fungi represent promising candidates for next-generation antifungals for topical application. These bio-molecules need to be tested for tolerance by the host and efficacy in pathogen clearance prior to being involved in clinical trials. In a proof-of-principle study, we provide evidence for the suitability of 3D human-cell based models as advantageous alternatives to animal experiments. We document the tolerance of specific AFPs and their curative efficacy against cutaneous C. albicans infection in a 3D skin model.
Collapse
Affiliation(s)
- Jeanett Holzknecht
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
168
|
Abdulghani M, Iram R, Chidrawar P, Bhosle K, Kazi R, Patil R, Kharat K, Zore G. Proteomic profile of Candida albicans biofilm. J Proteomics 2022; 265:104661. [PMID: 35728770 DOI: 10.1016/j.jprot.2022.104661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/06/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
Candida albicans biofilms are characterized by structural and cellular heterogeneity that confers antifungal resistance and immune evasion. Despite this, biofilm formation remains poorly understood. In this study, we used proteomic analysis to understand biofilm formation in C. albicans related to morphophysiological and architectural features. LC-MS/MS analysis revealed that 64 proteins were significantly modulated, of which 31 were upregulated and 33 were downregulated. The results indicate that metabolism (25 proteins), gene expression (13 proteins), stress response (7 proteins), and cell wall (5 proteins) composition are modulated. The rate of oxidative phosphorylation (OxPhos) and biosynthesis of UDP-N-acetylglucosamine, vitamin B6, and thiamine increased, while the rate of methionine biosynthesis decreased. There was a significant modification of the cell wall architecture due to higher levels of Sun41, Pir1 and Csh1 and increased glycosylation of proteins. It was observed that C. albicans induces hyphal growth by upregulating the expression of genes involved in cAMP-PKA and MAPK pathways. This study is significant in that it suggests an increase in OxPhos and alteration of cell wall architecture that could be contributing to the recalcitrance of C. albicans cells growing in biofilms. Nevertheless, a deeper investigation is needed to explore it further. SIGNIFICANCE: Candida sps is included in the list of pathogens with potential drug resistance threat due to the increased frequency especially colonization of medical devices, and tissues among the patients, in recent years. Significance of our study is that we are reporting traits like modulation in cell wall composition, amino acid and vitamin biosynthesis and importantly energy generation (OxPhos) etc. These traits could be conferring antifungal resistance, host immune evasion etc. and thus survival, in addition to facilitating biofilm formation. These findings are expected to prime the further studies on devising potent strategy against biofilm growth among the patients.
Collapse
Affiliation(s)
- Mazen Abdulghani
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Rasiqua Iram
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Priti Chidrawar
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Kajal Bhosle
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Rubina Kazi
- Division of Biochemical Sciences, CSIR-NCL, Pune 8, MS, India
| | - Rajendra Patil
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind, Pune 411007, MS, India
| | - Kiran Kharat
- Department of Biotechnology, Deogiri College, Aurangabad, MS, India
| | - Gajanan Zore
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India.
| |
Collapse
|
169
|
Duplan P, Memon MB, Choudhry H, Patterson J. A Rare Case of Candida parapsilosis Lumbar Discitis With Osteomyelitis. Cureus 2022; 14:e25955. [PMID: 35855242 PMCID: PMC9286020 DOI: 10.7759/cureus.25955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2022] [Indexed: 11/06/2022] Open
Abstract
Fungal osteomyelitis is rare and usually seen in immunocompromised patients. We report a case of Candida parapsilosis osteomyelitis in an immunocompetent patient with no prior surgical history. He went for spinal laminectomy with debridement and drainage. Intraoperative culture grew C. parapsilosis, and the patient was treated with fluconazole.
Collapse
|
170
|
D’Auria FD, Casciaro B, De Angelis M, Marcocci ME, Palamara AT, Nencioni L, Mangoni ML. Antifungal Activity of the Frog Skin Peptide Temporin G and Its Effect on Candida albicans Virulence Factors. Int J Mol Sci 2022; 23:ijms23116345. [PMID: 35683025 PMCID: PMC9181532 DOI: 10.3390/ijms23116345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/26/2023] Open
Abstract
The increasing resistance to conventional antifungal drugs is a widespread concern, and a search for new compounds, active against different species of fungi, is demanded. Antimicrobial peptides (AMPs) hold promises in this context. Here we investigated the activity of the frog skin AMP Temporin G (TG) against a panel of fungal strains, by following the Clinical and Laboratory Standards Institute protocols. TG resulted to be active against (i) Candida species and Cryptococcus neoformans, with MIC50 between 4 µM and 64 µM after 24 h of incubation; (ii) dermatophytes with MIC80 ranging from 4 to 32 µM, and (iii) Aspergillus strains with MIC80 of 128 µM. In addition, our tests revealed that TG reduced the metabolic activity of Candida albicans cells, with moderate membrane perturbation, as proven by XTT and Sytox Green assays, respectively. Furthermore, TG was found to be effective against some C. albicans virulence factors; indeed, at 64 µM it was able to inhibit ~90% of yeast-mycelial switching, strongly prevented biofilm formation, and led to a 50% reduction of metabolic activity in mature biofilm cells, and ~30-35% eradication of mature biofilm biomass. Even though further studies are needed to deepen our knowledge of the mechanisms of TG antifungal activity, our results suggest this AMP as an attractive lead compound for treatment of fungal diseases.
Collapse
Affiliation(s)
- Felicia Diodata D’Auria
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
| | - Bruno Casciaro
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy;
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
- Correspondence: (L.N.); (M.L.M.); Tel.: +39-0649914608 (L.N.); +39-0649910838 (M.L.M.)
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: (L.N.); (M.L.M.); Tel.: +39-0649914608 (L.N.); +39-0649910838 (M.L.M.)
| |
Collapse
|
171
|
Kulig K, Karnas E, Woznicka O, Kuleta P, Zuba-Surma E, Pyza E, Osyczka A, Kozik A, Rapala-Kozik M, Karkowska-Kuleta J. Insight Into the Properties and Immunoregulatory Effect of Extracellular Vesicles Produced by Candida glabrata, Candida parapsilosis, and Candida tropicalis Biofilms. Front Cell Infect Microbiol 2022; 12:879237. [PMID: 35734578 PMCID: PMC9207348 DOI: 10.3389/fcimb.2022.879237] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/11/2022] [Indexed: 01/12/2023] Open
Abstract
Currently, non-albicans Candida species, including C. tropicalis, C. glabrata, and C. parapsilosis, are becoming an increasing epidemiological threat, predominantly due to the distinct collection of virulence mechanisms, as well as emerging resistance to antifungal drugs typically used in the treatment of candidiasis. They can produce biofilms that release extracellular vesicles (EVs), which are nanometric spherical structures surrounded by a lipid bilayer, transporting diversified biologically active cargo, that may be involved in intercellular communication, biofilm matrix production, and interaction with the host. In this work, we characterize the size and protein composition of these structures for three species of non-albicans Candida fungi forming biofilm, indicating considerable heterogeneity of the investigated population of fungal EVs. Examination of the influence of EVs on cytokine production by the human monocytic cell line THP-1 differentiated into macrophage-like cells revealed that the tested vesicles have a stimulating effect on the secretion of tumor necrosis factor α and interleukin 8, while they reduce the production of interleukin 10. This may indicate the proinflammatory nature of the effect of EVs produced by these species on the host immune cells. Moreover, it has been indicated that vesicles may be involved in C. tropicalis biofilm resistance to fluconazole and caspofungin. This reveals the important role of EVs not only in the physiology of C. tropicalis, C. glabrata, and C. parapsilosis fungi but also in the pathogenesis of infections associated with the production of fungal biofilm.
Collapse
Affiliation(s)
- Kamila Kulig
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Olga Woznicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Patryk Kuleta
- Department of Molecular Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Artur Osyczka
- Department of Molecular Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- *Correspondence: Justyna Karkowska-Kuleta,
| |
Collapse
|
172
|
Effects of β-lapachone and β-nor-lapachone on multidrug efflux transporters and biofilms of Candida glabrata. Bioorg Med Chem 2022; 63:116749. [PMID: 35436747 DOI: 10.1016/j.bmc.2022.116749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022]
Abstract
Infections caused particularly by Candida glabrata are hard to treat due to the development of antifungal resistance that occurs mainly through the production of efflux pumps and biofilm. Thus, a promising strategy to overcome infections caused by C. glabrata could be to use a substance able to inhibit efflux pumps and eradicate biofilms. Lapachones are natural naphthoquinones that possess a variety of pharmacological properties. Previous studies show that these substances inhibit the growth, virulence factors and efflux pumps of C. albicans. The aim of the present study was to evaluate whether lapachones are able to inhibit efflux pumps related to antifungal resistance in C. glabrata and either prevent biofilm formation or affect mature biofilms. Assays were performed with Saccharomyces cerevisiae strains that overexpress C. glabrata transporters (CgCdr1p and CgCdr2p). One C. glabrata clinical isolate that overexpresses CgCdr1p was also used. Both β-lapachone and β-nor-lapachone affected the growth of S. cerevisiae and C. glabrata when combined to fluconazole, and this action was inhibited by ascorbic acid. Both lapachones stimulated ROS production, inhibited efflux activity, adhesion, biofilm formation and the metabolism of mature biofilms of C. glabrata. Data obtained on the present study point to the potential use of β-lapachone and β-nor-lapachone as antibiofilm agents and adjuvants on the antifungal therapy related to resistant infections caused by C. glabrata.
Collapse
|
173
|
Boahen A, Than LTL, Loke YL, Chew SY. The Antibiofilm Role of Biotics Family in Vaginal Fungal Infections. Front Microbiol 2022; 13:787119. [PMID: 35694318 PMCID: PMC9179178 DOI: 10.3389/fmicb.2022.787119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/25/2022] [Indexed: 11/15/2022] Open
Abstract
“Unity in strength” is a notion that can be exploited to characterize biofilms as they bestow microbes with protection to live freely, escalate their virulence, confer high resistance to therapeutic agents, and provide active grounds for the production of biofilms after dispersal. Naturally, fungal biofilms are inherently resistant to many conventional antifungals, possibly owing to virulence factors as their ammunitions that persistently express amid planktonic transition to matured biofilm state. These ammunitions include the ability to form polymicrobial biofilms, emergence of persister cells post-antifungal treatment and acquisition of resistance genes. One of the major disorders affecting vaginal health is vulvovaginal candidiasis (VVC) and its reoccurrence is termed recurrent VVC (RVVC). It is caused by the Candida species which include Candida albicans and Candida glabrata. The aforementioned Candida species, notably C. albicans is a biofilm producing pathogen and habitually forms part of the vaginal microbiota of healthy women. Latest research has implicated the role of fungal biofilms in VVC, particularly in the setting of treatment failure and RVVC. Consequently, a plethora of studies have advocated the utilization of probiotics in addressing these infections. Specifically, the excreted or released compounds of probiotics which are also known as postbiotics are being actively researched with vast potential to be used as therapeutic options for the treatment and prevention of VVC and RVVC. These potential sources of postbiotics are harnessed due to their proven antifungal and antibiofilm. Hence, this review discusses the role of Candida biofilm formation in VVC and RVVC. In addition, we discuss the application of pro-, pre-, post-, and synbiotics either individually or in combined regimen to counteract the abovementioned problems. A clear understanding of the role of biofilms in VVC and RVVC will provide proper footing for further research in devising novel remedies for prevention and treatment of vaginal fungal infections.
Collapse
|
174
|
Pezzotti G, Kobara M, Nakaya T, Imamura H, Asai T, Miyamoto N, Adachi T, Yamamoto T, Kanamura N, Ohgitani E, Marin E, Zhu W, Nishimura I, Mazda O, Nakata T, Makimura K. Raman Study of Pathogenic Candida auris: Imaging Metabolic Machineries in Reaction to Antifungal Drugs. Front Microbiol 2022; 13:896359. [PMID: 35694304 PMCID: PMC9175029 DOI: 10.3389/fmicb.2022.896359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/25/2022] [Indexed: 12/02/2022] Open
Abstract
The multidrug-resistant Candida auris often defies treatments and presently represents a worldwide public health threat. Currently, the ergosterol-targeting Amphotericin B (AmB) and the DNA/RNA-synthesis inhibitor 5-flucytosine (5-FC) are the two main drugs available for first-line defense against life-threatening Candida auris infections. However, important aspects of their mechanisms of action require further clarification, especially regarding metabolic reactions of yeast cells. Here, we applied Raman spectroscopy empowered with specifically tailored machine-learning algorithms to monitor and to image in situ the susceptibility of two Candida auris clades to different antifungal drugs (LSEM 0643 or JCM15448T, belonging to the East Asian Clade II; and, LSEM 3673 belonging to the South African Clade III). Raman characterizations provided new details on the mechanisms of action against Candida auris Clades II and III, while also unfolding differences in their metabolic reactions to different drugs. AmB treatment induced biofilm formation in both clades, but the formed biofilms showed different structures: a dense and continuous biofilm structure in Clade II, and an extra-cellular matrix with a “fluffy” and discontinuous structure in Clade III. Treatment with 5-FC caused no biofilm formation but yeast-to-hyphal or pseudo-hyphal morphogenesis in both clades. Clade III showed a superior capacity in reducing membrane permeability to the drug through chemically tailoring chitin structure with a high degree of acetylation and fatty acids networks with significantly elongated chains. This study shows the suitability of the in situ Raman method in characterizing susceptibility and stress response of different C. auris clades to antifungal drugs, thus opening a path to identifying novel clinical solutions counteracting the spread of these alarming pathogens.
Collapse
Affiliation(s)
- Giuseppe Pezzotti
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Kyoto, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Orthopedic Surgery, Tokyo Medical University, Tokyo, Japan
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan
- *Correspondence: Giuseppe Pezzotti
| | - Miyuki Kobara
- Division of Pathological Science, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tamaki Nakaya
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Kyoto, Japan
| | - Hayata Imamura
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Kyoto, Japan
| | - Tenma Asai
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Kyoto, Japan
| | - Nao Miyamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eriko Ohgitani
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Elia Marin
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Kyoto, Japan
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Kyoto, Japan
| | - Ichiro Nishimura
- Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, United States
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuo Nakata
- Division of Pathological Science, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Koichi Makimura
- Medical Mycology, Graduate School of Medicine, Teikyo University, Tokyo, Japan
| |
Collapse
|
175
|
Song Y, Wang Z, Long Y, Mao Y, Jiang F, Lu Y. 2-Alkyl-anthraquinones inhibit Candida albicans biofilm via inhibiting the formation of matrix and hyphae. Res Microbiol 2022; 173:103955. [PMID: 35550403 DOI: 10.1016/j.resmic.2022.103955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Candida albicans can form biofilm on biotic and abiotic surfaces of medical implants to cause superficial and systemic infections under specific condition. The formation of hyphae and matrix of C. albicans are considered as probable virulence factors. We assessed the inhibitory activities of 26 anthraquinones against C. albicans biofilm formation, which were substituted by different functional groups including hydroxyl groups, amino groups, carboxyl groups, alkyl groups, and glycoside groups at C1- or C2-position. Among them, anthraquinones without substituents at other positions but only an alkyl group attached to C2-position, namely 2-alkyl-anthraquinones were determined to have significant anti-biofilm activities. Furthermore, 2-ethylanthraquinone can significantly affect genes related to extracellular matrix (PMT6 and IFD6), and hyphal formation (HWP1, ECE1 and EFG1), leading to the disrupted formation of biofilm, by detail transcriptomics analysis. We believed that 2-ethylanthraquinone could inspire more discoveries of anti-biofilm agents against C. albicans.
Collapse
Affiliation(s)
- Yuanyuan Song
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziqi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Yijing Long
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Mao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Jiang
- State Key Laboratory of Natural Medicines, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
176
|
Bankaitis VA, Tripathi A, Chen XR, Igumenova TI. New strategies for combating fungal infections: Inhibiting inositol lipid signaling by targeting Sec14 phosphatidylinositol transfer proteins. Adv Biol Regul 2022; 84:100891. [PMID: 35240534 PMCID: PMC9149032 DOI: 10.1016/j.jbior.2022.100891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 12/13/2022]
Abstract
Virulent fungi represent a particularly difficult problem in the infectious disease arena as these organisms are eukaryotes that share many orthologous activities with their human hosts. The fact that these activities are often catalyzed by conserved proteins places additional demands on development of pharmacological strategies for specifically inhibiting target fungal activities without imposing undesirable secondary effects on the host. While deployment of a limited set of anti-mycotics has to date satisfied the clinical needs for treatment of fungal infections, the recent emergence of multi-drug resistant fungal 'superbugs' now poses a serious global health threat with rapidly diminishing options for treatment. This escalating infectious disease problem emphasizes the urgent need for development of new classes of anti-mycotics. In that regard, Sec14 phosphatidylinositol transfer proteins offer interesting possibilities for interfering with fungal phosphoinositide signaling with exquisite specificity and without targeting the highly conserved lipid kinases responsible for phosphoinositide production. Herein, we review the establishment of proof-of-principle that demonstrates the feasibility of such an approach. We also describe the lead compounds of four chemotypes that directly target fungal Sec14 proteins. The rules that pertain to the mechanism(s) of Sec14 inhibition by validated small molecule inhibitors, and the open questions that remain, are discussed - as are the challenges that face development of next generation Sec14-directed inhibitors.
Collapse
Affiliation(s)
- Vytas A Bankaitis
- Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, 77843-0014, USA; Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA.
| | - Ashutosh Tripathi
- Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, 77843-0014, USA
| | - Xiao-Ru Chen
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA
| | - Tatyana I Igumenova
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA
| |
Collapse
|
177
|
Elmehbad NY, Mohamed NA, Abd El-Ghany NA. Evaluation of the antimicrobial and anti-biofilm activity of novel salicylhydrazido chitosan derivatives impregnated with titanium dioxide nanoparticles. Int J Biol Macromol 2022; 205:719-730. [PMID: 35306018 DOI: 10.1016/j.ijbiomac.2022.03.076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 02/04/2023]
Abstract
Two novel chitosan derivatives were prepared by incorporating salicylhydrazide into chitosan Schiff base (SCsSB) and chitosan (SCs). Two nanocomposites, SCs/TiO2-1% and SCs/TiO2-3%, were also prepared. Their structures were confirmed using elemental analyses, FTIR, XRD, SEM, EDX and TEM. Their antimicrobial and anti-biofilm activities were arranged as: SCs/TiO2-3% > SCs/TiO2-1% > SCs > SCsSB > chitosan. SCs showed minimum inhibitory concentration (MIC) value of 1.95 μg/mL against A. niger which was comparable with that of Amphotericin B. SCs/TiO2-3% showed higher inhibition against S. epidermidis, S. aureus, S. pyogenes, P. aeruginosa and E. coli than Vancomycin. While, it showed comparable inhibition activity to that of Vancomycin against B. subtilis and P. mirabilis. SCs/TiO2-3% showed MIC values equal 0.48 and 0.98 μg/mL corresponded to 0.98 and 1.95 μg/mL of Amphotericin B against C. albicans, A. fumigatus and A. niger, respectively. SCs/TiO2-3% showed much lower minimum biofilm inhibitory concentration (MBIC) values, ranged between 1.95 and 7.81 μg/mL, than those of SCs, ranged from 62.5 to 125 μg/mL. SCs/TiO2-3% was safe on normal human cells. The modifiers and TiO2 nanoparticles incorporated into chitosan in one structure developed its performance. It is approach for attaining appropriate structures which are good competitors for antimicrobial agents.
Collapse
Affiliation(s)
- Noura Y Elmehbad
- Department of Chemistry, Faculty of Science and Arts, Najran University, Najran, Saudi Arabia
| | - Nadia A Mohamed
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia; Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt.
| | - Nahed A Abd El-Ghany
- Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
178
|
The Bacillary Postbiotics, Including 2-Undecanone, Suppress the Virulence of Pathogenic Microorganisms. Pharmaceutics 2022; 14:pharmaceutics14050962. [PMID: 35631548 PMCID: PMC9143114 DOI: 10.3390/pharmaceutics14050962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/11/2022] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
Secreted molecules from probiotic Bacilli have often been considered potential pharmaceuticals to fight infections caused by bacterial or yeast pathogens. In the present study, we investigated the antagonistic potential of secreted probiotic filtrates (hereafter, postbiotics) derived from Lactobacillus plantarum cells against pathogenic microorganisms, such as Escherichia coli, Staphylococcus aureus, and Candida albicans. We found that the postbiotics mitigate the biofilms of the tested pathogens with no notable effect on their planktonic growth. In addition, the postbiotics suppressed some virulence traits, for instance, the dendrite swarming motility of E. coli and yeast-to-hyphal switch in C. albicans. Further assays with an active constituent produced by the L. plantarum cells–2-undecanone revealed two significant findings: (i) 2-undecanone inhibits C. albicans biofilms and hyphae in vitro and in a Caenorhabditis elegans model, and (ii) it interacts specifically with Gln 58 amino acid residue of hyphal wall protein-1 (Hwp-1) in molecular docking analysis. The results suggest the targeted mode of antagonistic action of 2-undecanone against C. albicans biofilm. In total, the findings of the study depict an appealing strategy to use postbiotics, including specific ketone molecules, produced by L. plantarum for developing novel antibiofilm and anti-hyphal pharmaceuticals.
Collapse
|
179
|
Differential Response of Candida Species Morphologies and Isolates to Fluconazole and Boric Acid. Antimicrob Agents Chemother 2022; 66:e0240621. [PMID: 35446135 DOI: 10.1128/aac.02406-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Candida albicans is the most prevalent cause of vulvovaginal candidiasis ("yeast infection" or VVC) and recurrent vulvovaginal candidiasis (RVVC), although the incidence of non-albicans yeast species is increasing. The azole fluconazole is the primary antifungal drug used to treat RVVC, yet isolates from some species have intrinsic resistance to fluconazole, and recurrent infection can occur even with fluconazole-susceptible populations. The second-line broad-spectrum antimicrobial drug, boric acid, is an alternative treatment that has been found to successfully treat complicated VVC infections. Far less is known about how boric acid inhibits growth of yeast isolates in different morphologies compared to fluconazole. We found significant differences in drug resistance and drug tolerance (the ability of a subpopulation to grow slowly in high levels of drug) between C. albicans, Candida glabrata, and Candida parapsilosis isolates, with the specific relationships dependent on both drug and phenotype. Population-level variation for both susceptibility and tolerance was broader for fluconazole than boric acid in all species. Unlike fluconazole, which neither prevented hyphal formation nor disrupted mature biofilms, boric acid inhibited C. albicans hyphal formation and reduced mature biofilm biomass and metabolic activity in all isolates in a dose-dependent manner. Variation in planktonic response did not generally predict biofilm phenotypes for either drug. Overall, our findings illustrate that boric acid is broadly effective at inhibiting growth across many isolates and morphologies, which could explain why it is an effective treatment for RVVC.
Collapse
|
180
|
Negm WA, El-Aasr M, Attia G, Alqahtani MJ, Yassien RI, Abo Kamer A, Elekhnawy E. Promising Antifungal Activity of Encephalartos laurentianus de Wild against Candida albicans Clinical Isolates: In Vitro and In Vivo Effects on Renal Cortex of Adult Albino Rats. J Fungi (Basel) 2022; 8:jof8050426. [PMID: 35628682 PMCID: PMC9144060 DOI: 10.3390/jof8050426] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 02/08/2023] Open
Abstract
Candida albicans can cause various infections, especially in immunocompromised patients. Its ability to develop resistance to the current antifungal drugs as well as its multiple virulence factors have rendered the problem even more complicated. Thus, in the present investigation, we elucidated an in vitro and in vivo antifungal activity of Encephalartos laurentianus methanol extract (ELME) against C. albicans clinical isolates for the first time. A phytochemical identification of 64 compounds was conducted in ELME using LC-MS/MS. Interestingly, ELME exhibited antifungal activity with MIC values that ranged from 32–256 µg/mL. Furthermore, we investigated the antibiofilm activity of ELME against the biofilms formed by C. albicans isolates. ELME displayed antibiofilm activity using a crystal violet assay as it decreased the percentages of cells, moderately and strongly forming biofilms from 62.5% to 25%. Moreover, the antibiofilm impact of ELME was elucidated using SEM and fluorescent microscope. A significant reduction in the biofilm formation by C. albicans isolates was observed. In addition, we observed that ELME resulted in the downregulation of the biofilm-related tested genes (ALS1, BCR1, PLB2, and SAP5) in 37.5% of the isolates using qRT-PCR. Besides, the in vivo antifungal activity of ELME on the kidney tissues of rats infected with C. albicans was investigated using histological and immunohistochemical studies. ELME was found to protect against C. albicans induced renal damage, decrease desmin and inducible nitric oxide synthase, increase alkaline phosphatase, and increase infected rats’ survival rate. Additionally, the cytotoxicity of ELME was elucidated on Human Skin Fibroblast normal cells using MTT assay. ELME had an IC50 of 31.26 µg/mL. Thus, we can conclude that ELME might be a promising future source for antifungal compounds.
Collapse
Affiliation(s)
- Walaa A. Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (M.E.-A.); (G.A.)
- Correspondence: (W.A.N.); (E.E.)
| | - Mona El-Aasr
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (M.E.-A.); (G.A.)
| | - Ghada Attia
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (M.E.-A.); (G.A.)
| | - Moneerah J. Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Rania Ibrahim Yassien
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shebin El-Kom 32511, Egypt;
| | - Amal Abo Kamer
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
- Correspondence: (W.A.N.); (E.E.)
| |
Collapse
|
181
|
Evaluation of Antimicrobial and Anti-Biofilm Formation Activities of Novel Poly(vinyl alcohol) Hydrogels Reinforced with Crosslinked Chitosan and Silver Nano-Particles. Polymers (Basel) 2022; 14:polym14081619. [PMID: 35458369 PMCID: PMC9026349 DOI: 10.3390/polym14081619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Novel hydrogels were prepared by blending chitosan and poly(vinyl alcohol), PVA, then crosslinking the resulting blends using trimellitic anhydride isothiocyanate at a concentration based on chitosan content in the blends. The weight ratios of chitosan: PVA in the blends were 1:3, 1:1, and 3:1 to produce three hydrogels symbolized as H13, H11, and H31, respectively. For a comparison, H10 was also prepared by crosslinking pure chitosan with trimellitic anhydride isothiocyanate. For further modification, three H31/silver nanocomposites (AgNPs) were synthesized using three different concentrations of silver nitrate to obtain H31/AgNPs1%, H31/AgNPs3% and H31/AgNPs5%. The structures of the prepared samples were emphasized using various analytical techniques. PVA has no inhibition activity against the tested microbes and biofilms. The antimicrobial and anti-biofilm formation activities of the investigated samples was arranged as: H31/AgNPs5% ≥ H31/AgNPs3% > H31/AgNPs1% > H10 > H31 > H11 > H13 > chitosan. H31/AgNPs5% and H31/AgNPs3% were more potent than Vancomycin and Amphotericin B against most of the tested microbes. Interestingly, H31 and H31/AgNPs3% were safe on the normal human cells. Consequently, hydrogels resulting from crosslinked blends of chitosan and PVA loaded with AgNPs in the same structure have significantly reinforced the antimicrobial and inhibition activity against the biofilms of PVA.
Collapse
|
182
|
Bharti S, Zakir F, Mirza MA, Aggarwal G. Antifungal biofilm strategies: a less explored area in wound management. Curr Pharm Biotechnol 2022; 23:1497-1513. [PMID: 35410595 DOI: 10.2174/1389201023666220411100214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/03/2021] [Accepted: 01/03/2022] [Indexed: 11/22/2022]
Abstract
Background- The treatment of wound associated infections has always remained a challenge for clinicians with the major deterring factor being microbial biofilms, majorly bacterial or fungal. Biofilm infections are becoming a global concern owing to resistance against antimicrobials. Fungal biofilms are formed by a wide variety of fungal pathogens namely Candida sp., Aspergillus fumigates, Trichosporon sp., Saccharomyces cerevisiae, Cryptococcus neoformans, among others. The rising cases of fungal biofilm resistance add to the burden of wound care. Additionally, with increase in the number of surgical procedures, transplantation and the exponential use of medical devices, fungal bioburden is on the rise. Objectives- The review discusses the methods of biofilm formation and the resistance mechanisms against conventional treatments. The potential of novel delivery strategies and the mechanisms involved therein are highlighted. Further, the prospects of nanotechnology based medical devices to combat fungal biofilm resistance have also been explored. Some of the clinical trials and up-to-date patent technologies to eradicate the biofilms are also mentioned. Conclusion- Due to the many challenges faced in preventing/eradicating biofilms, only a handful of approaches have been able to make it to the market. Fungal biofilms are a fragmentary area which needs further exploration.
Collapse
Affiliation(s)
- Shilpa Bharti
- Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi
| | - Foziyah Zakir
- Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi
| | - Mohd Aamir Mirza
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Geeta Aggarwal
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| |
Collapse
|
183
|
Fraga-Silva TFDC, Munhoz-Alves N, Mimura LAN, de Oliveira LRC, Figueiredo-Godoi LMA, Garcia MT, Oliveira ES, Ishikawa LLW, Zorzella-Pezavento SFG, Bonato VLD, Junqueira JC, Bagagli E, Sartori A. Systemic Infection by Non-albicans Candida Species Affects the Development of a Murine Model of Multiple Sclerosis. J Fungi (Basel) 2022; 8:jof8040386. [PMID: 35448617 PMCID: PMC9032036 DOI: 10.3390/jof8040386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/26/2022] [Accepted: 04/07/2022] [Indexed: 01/08/2023] Open
Abstract
Candidiasis may affect the central nervous system (CNS), and although Candida albicans is predominant, non-albicans Candida species can also be associated with CNS infections. Some studies have suggested that Candida infections could increase the odds of multiple sclerosis (MS) development. In this context, we investigated whether systemic infection by non-albicans Candida species would affect, clinically or immunologically, the severity of experimental autoimmune encephalomyelitis (EAE), which is an animal model used to study MS. For this, a strain of C. glabrata, C. krusei, and C. parapsilosis was selected and characterized using different in vitro and in vivo models. In these analysis, all the strains exhibited the ability to form biofilms, produce proteolytic enzymes, and cause systemic infections in Galleria mellonella, with C. glabrata being the most virulent species. Next, C57BL/6 mice were infected with strains of C. glabrata, C. krusei, or C. parapsilosis, and 3 days later were immunized with myelin oligodendrocyte glycoprotein to develop EAE. Mice from EAE groups previously infected with C. glabrata and C. krusei developed more severe and more prevalent paralysis, while mice from the EAE group infected with C. parapsilosis developed a disease comparable to non-infected EAE mice. Disease aggravation by C. glabrata and C. krusei strains was concomitant to increased IL-17 and IFN-γ production by splenic cells stimulated with fungi-derived antigens and with increased percentage of T lymphocytes and myeloid cells in the CNS. Analysis of interaction with BV-2 microglial cell line also revealed differences among these strains, in which C. krusei was the strongest activator of microglia concerning the expression of MHC II and CD40 and pro-inflammatory cytokine production. Altogether, these results indicated that the three non-albicans Candida strains were similarly able to reach the CNS but distinct in terms of their effect over EAE development. Whereas C. glabrata and C. Krusei aggravated the development of EAE, C. parapsilosis did not affect its severity. Disease worsening was partially associated to virulence factors in C. glabrata and to a strong activation of microglia in C. krusei infection. In conclusion, systemic infections by non-albicans Candida strains exerted influence on the experimental autoimmune encephalomyelitis in both immunological and clinical aspects, emphasizing their possible relevance in MS development.
Collapse
Affiliation(s)
- Thais Fernanda de Campos Fraga-Silva
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
- Correspondence:
| | - Natália Munhoz-Alves
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
| | - Luiza Ayumi Nishiyama Mimura
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
| | | | - Lívia Mara Alves Figueiredo-Godoi
- Institute of Science and Technology, São Paulo State University (UNESP), Sao Jose dos Campos 12245-000, Brazil; (L.M.A.F.-G.); (M.T.G.); (J.C.J.)
| | - Maíra Terra Garcia
- Institute of Science and Technology, São Paulo State University (UNESP), Sao Jose dos Campos 12245-000, Brazil; (L.M.A.F.-G.); (M.T.G.); (J.C.J.)
| | - Evelyn Silva Oliveira
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
| | - Larissa Lumi Watanabe Ishikawa
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
| | - Sofia Fernanda Gonçalves Zorzella-Pezavento
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
| | - Vânia Luiza Deperon Bonato
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto 14049-900, Brazil;
| | - Juliana Campos Junqueira
- Institute of Science and Technology, São Paulo State University (UNESP), Sao Jose dos Campos 12245-000, Brazil; (L.M.A.F.-G.); (M.T.G.); (J.C.J.)
| | - Eduardo Bagagli
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
| | - Alexandrina Sartori
- Department of Chemistry and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (N.M.-A.); (L.A.N.M.); (E.S.O.); (L.L.W.I.); (S.F.G.Z.-P.); (E.B.); (A.S.)
- Postgraduate Program in Tropical Disease, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil;
| |
Collapse
|
184
|
Ahmed HY, Safwat N, Shehata R, Althubaiti EH, Kareem S, Atef A, Qari SH, Aljahani AH, Al-Meshal AS, Youssef M, Sami R. Synthesis of Natural Nano-Hydroxyapatite from Snail Shells and Its Biological Activity: Antimicrobial, Antibiofilm, and Biocompatibility. MEMBRANES 2022; 12:408. [PMID: 35448378 PMCID: PMC9025656 DOI: 10.3390/membranes12040408] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/25/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023]
Abstract
Hydroxyapatite nanoparticles (HAn) have been produced as biomaterial from biowaste, especially snail shells (Atactodea glabrata). It is critical to recycle the waste product in a biomedical application to overcome antibiotic resistance as well as biocompatibility with normal tissues. Moreover, EDX, TEM, and FT-IR analyses have been used to characterize snail shells and HAn. The particle size of HAn is about 15.22 nm. Furthermore, higher inhibitory activity was observed from HAn than the reference compounds against all tested organisms. The synthesized HAn has shown the lowest MIC values of about 7.8, 0.97, 3.9, 0.97, and 25 µg/mL for S. aureus, B. subtilis, K. pneumonia, C. albicans, and E. coli, respectively. In addition, the HAn displayed potent antibiofilm against S. aureus and B. subtilis. According to the MTT, snail shell and HAn had a minor influence on the viability of HFS-4 cells. Consequently, it could be concluded that some components of waste, such as snail shells, have economic value and can be recycled as a source of CaO to produce HAn, which is a promising candidate material for biomedical applications.
Collapse
Affiliation(s)
- Hanaa Y Ahmed
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11787, Egypt
| | - Nesreen Safwat
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11787, Egypt
| | - Reda Shehata
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11787, Egypt
| | - Eman Hillal Althubaiti
- Department of Biotechnology, Faculty of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Sayed Kareem
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11787, Egypt
| | - Ahmed Atef
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11787, Egypt
| | - Sameer H Qari
- Department of Biology, Al-Jumum University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Amani H Aljahani
- Department of Physical Sport Science, College of Education, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Areej Suliman Al-Meshal
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mahmoud Youssef
- Food Science and Technology Department, Faculty of Agriculture, Al-Azhar University, Cairo 11787, Egypt
| | - Rokayya Sami
- Department of Food Science and Nutrition, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
185
|
Nirwati H, Damayanti E, Sholikhah EN, Mutofa M, Widada J. Soil-derived Streptomyces sp. GMR22 producing antibiofilm activity against Candida albicans: bioassay, untargeted LC-HRMS, and gene cluster analysis. Heliyon 2022; 8:e09333. [PMID: 35520628 PMCID: PMC9065622 DOI: 10.1016/j.heliyon.2022.e09333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/04/2022] [Accepted: 04/21/2022] [Indexed: 11/01/2022] Open
Abstract
Biofilm-forming fungi, Candida albicans, are currently a serious problem in infectious disease cases. Soil bacteria Streptomyces sp. GMR22 have a large genome size and antifungal metabolites against C. albicans, but its potential antibiofilm activity is not clearly defined. The aims of this study were to determine the antibiofilm activity of GMR22 against C. albicans, identify the main constituents of active extracts, and investigate the biosynthesis gene clusters encoding the enzymes related to metabolism pathways. Antifungal and antibiofilm measurements were performed using in vitro assays on C. albicans ATCC 10231. Main constituents of active extracts were analyzed using untargeted Liquid Chromatography tandem High-Resolution Mass Spectrometry (LC-HRMS). RAST software was applied to investigate the gene clusters of the biosynthesis pathways based on whole genome sequences. Chloroform extract of GMR22 has antifungal and antibiofilm properties at 13-420 μg/mL with palmitic acid (C16H32O2, 273.27028 Da), a saturated fatty acid as a major constituent (42.74). Streptomyces sp. GMR22 has 53 subsystems related to fatty acids biosynthesis (Fab) FAS II. The Kyoto Encyclopedia of Gene and Genome map of Fab revealed 10 of 21 (47.6%) gene clusters encode enzymes related to Fab. There were six gene clusters encoding the enzymes related to the hexadecenoic acid (palmitic acid) biosynthesis pathways: 6.4.12; FabD, FabH, FabF, FabG, FabI and 1.14.192. Each enzyme was encoded by 3-14 genes. These results confirmed that soil Streptomyces sp. GMR22 bacterium has remarkable biotechnological potential by producing fatty acids which are mostly palmitic acid as an active antibiofilm agent against C. albicans.
Collapse
Affiliation(s)
- Hera Nirwati
- Department of Microbiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Ema Damayanti
- Research Center for Food Technology and Processing, National Research and Innovation Agency, Gunungkidul, 55861, Indonesia
| | - Eti Nurwening Sholikhah
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Mustofa Mutofa
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Jaka Widada
- Department of Microbiology, Faculty of Agriculture, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| |
Collapse
|
186
|
Perrine-Walker F. Caspofungin resistance in Candida albicans: genetic factors and synergistic compounds for combination therapies. Braz J Microbiol 2022; 53:1101-1113. [PMID: 35352319 PMCID: PMC9433586 DOI: 10.1007/s42770-022-00739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/25/2022] [Indexed: 11/25/2022] Open
Abstract
Caspofungin and other echinocandins have been used for the treatment of human infections by the opportunistic yeast pathogen, Candida albicans. There has been an increase in infections by non-albicans Candida species such as Candida glabrata, Candida parapsilosis, Candida tropicalis, Candida krusei, and Candida auris in clinical or hospital settings. This is problematic to public health due to the increasing prevalence of echinocandin resistant species/strains. This review will present a summary on various studies that investigated the inhibitory action of caspofungin on 1,3-β-D-glucan synthesis, on cell wall structure, and biofilm formation of C. albicans. It will highlight some of the issues linked to caspofungin resistance or reduced caspofungin sensitivity in various Candida species and the potential benefits of antimicrobial peptides and other compounds in synergy with caspofungin.
Collapse
Affiliation(s)
- Francine Perrine-Walker
- Department of Biochemistry and Genetics, La Trobe Institute For Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
187
|
Anti-Candida Properties of Gossypium hirsutum L.: Enhancement of Fungal Growth, Biofilm Production and Antifungal Resistance. Pharmaceutics 2022; 14:pharmaceutics14040698. [PMID: 35456532 PMCID: PMC9031239 DOI: 10.3390/pharmaceutics14040698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/12/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
(1) Background: Candida is a genus of yeasts with notable pathogenicity and significant ability to develop antimicrobial resistance. Gossypium hirsutum L., a medicinal plant that is traditionally used due to its antimicrobial properties, has demonstrated significant antifungal activity. Therefore, this study investigated the chemical composition and anti-Candida effects of aqueous (AELG) and hydroethanolic (HELG) extracts obtained from the leaves of this plant. (2) Methods: The extracts were chemically characterized by UPLC–QTOF-MS/MS, and their anti-Candida activities were investigated by analyzing cell viability, biofilm production, morphological transition, and enhancement of antifungal resistance. (3) Results: The UPLC–QTOF-MS/MS analysis revealed the presence of twenty-one compounds in both AELG and HELG, highlighting the predominance of flavonoids. The combination of the extracts with fluconazole significantly reduced its IC50 values against Candida albicans INCQS 40006, Candida tropicalis INCQS 40042, and C. tropicalis URM 4262 strains, indicating enhanced antifungal activity. About biofilm production, significant inhibition was observed only for the AELG-treated C. tropicalis URM 4262 strain in comparison with the untreated control. Accordingly, this extract showed more significant inhibitory effects on the morphological transition of the INCQS 40006 and URM 4387 strains of C. albicans (4) Conclusions: Gossypium hirsutum L. presents promising antifungal effects, that may be potentially linked to the combined activity of chemical constituents identified in its extracts.
Collapse
|
188
|
Kaypetch R, Rudrakanjana P, Churnjitapirom P, Tua-Ngam P, Tonput P, Tantivitayakul P. Geraniol and thymoquinone inhibit Candida spp. biofilm formation on acrylic denture resin without affecting surface roughness or color. J Oral Sci 2022; 64:161-166. [PMID: 35321964 DOI: 10.2334/josnusd.21-0435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
PURPOSE This study was designed to investigate the in vitro effects of geraniol (GE) and thymoquinone (TQ) on Candida biofilms on denture acrylic and any accompanying changes in acrylic surface roughness or color. METHODS The susceptibility of Candida species to GE and TQ was determined using the broth microdilution method and time-kill assay. A minimum biofilm eradication concentration (MBEC) assay was performed using 7-day Candida biofilms grown on denture acrylic. RESULTS The minimum inhibitory concentration (MIC) of GE and TQ for Candida spp. was 256 and 32 µg/mL, respectively. The Candida strain complete kill rates for GE and TQ at 5-fold MIC were determined after 1 h of incubation. At 5-fold MIC, GE and TQ inhibited the preformed biofilm activity (MBEC80) of all Candida strains on denture acrylic by more than 80% after treatment for 3 h. At sub-MIC levels, GE and TQ prevented the development of C. albicans and C. tropicalis hyphae. SEM images demonstrated that GE and TQ damaged the fungal cell membrane and induced cell lysis. On the other hand, GE and TQ at 10-fold MIC did not alter the surface roughness or color of the denture acrylic. CONCLUSION GE and TQ are interesting natural substances that could be developed as promising disinfectants for removable dentures.
Collapse
Affiliation(s)
| | | | | | | | - Pairin Tonput
- Research Office, Faculty of Dentistry, Mahidol University
| | | |
Collapse
|
189
|
Atiencia-Carrera MB, Cabezas-Mera FS, Tejera E, Machado A. Prevalence of biofilms in Candida spp. bloodstream infections: A meta-analysis. PLoS One 2022; 17:e0263522. [PMID: 35113972 PMCID: PMC8812928 DOI: 10.1371/journal.pone.0263522] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/20/2022] [Indexed: 01/10/2023] Open
Abstract
CONTEXT Candida-related infections are nowadays a serious Public Health Problem emerging multidrug-resistant strains. Candida biofilm also leads bloodstream infections to invasive systemic infections. OBJECTIVE The present meta-analysis aimed to analyze Candida biofilm rate, type, and antifungal resistance among hospitalized patients between 1995 and 2020. DATA SOURCES Web of Science, Scopus, PubMed, and Google Scholar databases were searched for English papers using the following medical subject heading terms (MESH): "invasive candidiasis"; "bloodstream infections"; "biofilm formation"; "biofilm-related infections"; "mortality"; and "prevalence". STUDY SELECTION The major inclusion criteria included reporting the rate of biofilm formation and the prevalence of biofilm-related to Candida species, including observational studies (more exactly, cohort, retrospective, and case-control studies). Furthermore, data regarding the mortality rate, the geographical location of the study set, and the use of anti-fungal agents in clinical isolates were also extracted from the studies. DATA EXTRACTION Independent extraction of articles by 2 authors using predefined data fields, including study quality indicators. DATA SYNTHESIS A total of 31 studies from publicly available databases met our inclusion criteria. The biofilm formation in the data set varied greatly from 16 to 100% in blood samples. Most of the studies belonged to Europe (17/31) and Asia (9/31). Forest plot showed a pooled rate of biofilm formation of 80.0% (CI: 67-90), with high heterogeneity (Q = 2567.45, I2 = 98.83, τ2 = 0.150) in random effects model (p < 0.001). The funnel plot and Egger's linear regression test failed to find publication bias (p = 0.896). The mortality rate in Candida-related bloodstream infections was 37.9% of which 70.0% were from biofilm-associated infections. Furthermore, Candida isolates were also characterized in low, intermediate, or high biofilm formers through their level of biofilm mass (crystal violet staining or XTT assays) after a 24h growth. When comparing between countries, statistical differences were obtained (p = 0.0074), showing the lower and higher biofilm prevalence values in Italy and Spain, respectively. The prevalence of low, intermediate, and high biofilms were 36.2, 18.9, and 35.0% (p < 0.0001), respectively. C. tropicalis was the prevalent species in high biofilm formation (67.5%) showing statistically significant differences when compared to other Candida species, except for C. krusei and C. glabrata. Finally, the rates of antifungal resistance to fluconazole, voriconazole, and caspofungin related to biofilm were 70.5, 67.9 and 72.8% (p < 0.001), respectively. CONCLUSIONS Early detection of biofilms and a better characterization of Candida spp. bloodstream infections should be considered, which eventually will help preserve public health resources and ultimately diminish mortality among patients.
Collapse
Affiliation(s)
- María Belén Atiencia-Carrera
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito (USFQ), Diego de Robles y Vía Interoceánica, Campus Cumbayá, Quito, Pichincha, Ecuador
| | - Fausto Sebastián Cabezas-Mera
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito (USFQ), Diego de Robles y Vía Interoceánica, Campus Cumbayá, Quito, Pichincha, Ecuador
| | - Eduardo Tejera
- Facultad de Ingeniería y Ciencias Agropecuarias Aplicadas, Grupo de Bioquimioinformática, Universidad de Las Américas, Quito, Pichincha, Ecuador
- * E-mail: (ET); (AM)
| | - António Machado
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito (USFQ), Diego de Robles y Vía Interoceánica, Campus Cumbayá, Quito, Pichincha, Ecuador
- * E-mail: (ET); (AM)
| |
Collapse
|
190
|
Ahmed N, Mahmood MS, Ullah MA, Araf Y, Rahaman TI, Moin AT, Hosen MJ. COVID-19-Associated Candidiasis: Possible Patho-Mechanism, Predisposing Factors, and Prevention Strategies. Curr Microbiol 2022; 79:127. [PMID: 35287179 PMCID: PMC8918595 DOI: 10.1007/s00284-022-02824-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/23/2022] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is threatening public health. A large number of affected people need to be hospitalized. Immunocompromised patients and ICU-admitted patients are predisposed to further bacterial and fungal infections, making patient outcomes more critical. Among them, COVID-19-associated candidiasis is becoming more widely recognized as a part of severe COVID-19 sequelae. While the molecular pathophysiology is not fully understood, some factors, including a compromised immune system, iron and zinc deficiencies, and nosocomial and iatrogenic transmissions, predispose COVID-19 patients to candidiasis. In this review, we discuss the existing knowledge of the virulence characteristics of Candida spp. and summarize the key concepts in the possible molecular pathogenesis. We analyze the predisposing factors that make COVID-19 patients more susceptible to candidiasis and the preventive measures which will provide valuable insights to guide the effective prevention of candidiasis in COVID-19 patients.
Collapse
Affiliation(s)
- Nafisa Ahmed
- Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Maiesha Samiha Mahmood
- Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Md. Asad Ullah
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Tanjim Ishraq Rahaman
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Mohammad Jakir Hosen
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
191
|
Miranda-Cadena K, Marcos-Arias C, Perez-Rodriguez A, Cabello-Beitia I, Mateo E, Sevillano E, Madariaga L, Quindós G, Eraso E. In vitro and in vivo anti- Candida activity of citral in combination with fluconazole. J Oral Microbiol 2022; 14:2045813. [PMID: 35251524 PMCID: PMC8896188 DOI: 10.1080/20002297.2022.2045813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Background The ability of Candida to develop biofilms on inert surfaces or living tissues favors recalcitrant and chronic candidiasis associated, in many instances, with resistance to current antifungal therapy. Aim The aim of this study was to evaluate the antifungal activity of citral, a phytocompound present in lemongrass essential oil, in monotherapy and combined with fluconazole against azole-resistant Candida planktonic cells and biofilms. The effect of citral combined with fluconazole was also analysed with regard to the expression of fluconazole resistance-associated genes in Candida albicans and the effectiveness of the combination therapy in a Caenorhabditis elegans model of candidiasis. Results Citral reduced biofilm formation at initial stages and the metabolic activity of the mature biofilm. The combination of citral with fluconazole was synergistic, with a significant increase in the survival of C. elegans infected with Candida. RNA analysis revealed a reduction of the expression of the efflux pump encoded by MDR1, leading to a greater effect of fluconazole. Conclusion Citral in monotherapy and in combination with fluconazole could represent an interesting therapy to treat recalcitrant Candida infections associated to biofilms.
Collapse
Affiliation(s)
- Katherine Miranda-Cadena
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Cristina Marcos-Arias
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Aitzol Perez-Rodriguez
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Iván Cabello-Beitia
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Estibaliz Mateo
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Elena Sevillano
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Lucila Madariaga
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Guillermo Quindós
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| | - Elena Eraso
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country, Bilbao, Spain
| |
Collapse
|
192
|
Discovery of polypyridyl iridium(III) complexes as potent agents against resistant Candida albicans. Eur J Med Chem 2022; 233:114250. [DOI: 10.1016/j.ejmech.2022.114250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/15/2022] [Accepted: 02/28/2022] [Indexed: 12/21/2022]
|
193
|
Bellavita R, Maione A, Merlino F, Siciliano A, Dardano P, De Stefano L, Galdiero S, Galdiero E, Grieco P, Falanga A. Antifungal and Antibiofilm Activity of Cyclic Temporin L Peptide Analogues against Albicans and Non-Albicans Candida Species. Pharmaceutics 2022; 14:pharmaceutics14020454. [PMID: 35214187 PMCID: PMC8877061 DOI: 10.3390/pharmaceutics14020454] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 12/13/2022] Open
Abstract
Temporins are one of the largest families of antimicrobial peptides with both anti-inflammatory and antimicrobial activity. Herein, for a panel of cyclic temporin L isoform analogues, the antifungal and antibiofilm activities were determined against representative Candida strains, including C. albicans, C. glabrata, C. auris, C. parapsilosis and C. tropicalis. The outcomes indicated a significant anti-candida activity against planktonic and biofilm growth for four peptides (3, 7, 15 and 16). The absence of toxicity up to high concentrations and survival after infection were assessed in vivo by using Galleria mellonella larvae, and the correlation between conformation and cytotoxicity was investigated by fluorescence assays and circular dichroism (CD). By combining fluorescence spectroscopy, CD, dynamic light scattering, confocal and atomic force microscopy, the mode of action of four analogues was hypothesized. The results pinpointed that peptide 3 emerged as a non-toxic compound showing a potent antibiofilm activity and represents a promising compound for biomedical applications.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department of Pharmacy, School of Medicine, University of Naples ‘Federico II’, Via Domenico Montesano 49, 80131 Naples, Italy; (R.B.); (F.M.); (S.G.)
| | - Angela Maione
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.S.)
| | - Francesco Merlino
- Department of Pharmacy, School of Medicine, University of Naples ‘Federico II’, Via Domenico Montesano 49, 80131 Naples, Italy; (R.B.); (F.M.); (S.G.)
| | - Antonietta Siciliano
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.S.)
| | - Principia Dardano
- Institute of Applied Sciences and Intelligent Systems, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Naples, Italy; (P.D.); (L.D.S.)
| | - Luca De Stefano
- Institute of Applied Sciences and Intelligent Systems, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Naples, Italy; (P.D.); (L.D.S.)
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples ‘Federico II’, Via Domenico Montesano 49, 80131 Naples, Italy; (R.B.); (F.M.); (S.G.)
| | - Emilia Galdiero
- Department of Biology, University of Naples ‘Federico II’, Via Cinthia, 80126 Naples, Italy; (A.M.); (A.S.)
- Correspondence: (E.G.); (P.G.); (A.F.); Tel.: +39-081-679182 (E.G.); +39-081-678620 (P.G.); +39-081-2534503 (A.F.)
| | - Paolo Grieco
- Department of Pharmacy, School of Medicine, University of Naples ‘Federico II’, Via Domenico Montesano 49, 80131 Naples, Italy; (R.B.); (F.M.); (S.G.)
- Correspondence: (E.G.); (P.G.); (A.F.); Tel.: +39-081-679182 (E.G.); +39-081-678620 (P.G.); +39-081-2534503 (A.F.)
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples ‘Federico II’, Via Università 100, 80055 Portici, Italy
- Correspondence: (E.G.); (P.G.); (A.F.); Tel.: +39-081-679182 (E.G.); +39-081-678620 (P.G.); +39-081-2534503 (A.F.)
| |
Collapse
|
194
|
Silva SL, de Oliveira Pereira F, Cordeiro LV, Diniz Neto H, Dos Santos Maia M, da Silva Souza HD, de Athayde-Filho PF, Scotti MT, Scotti L, de Oliveira Lima E. Antifungal activity of 2-Chloro-N-phenylacetamide, docking and molecular dynamics studies against clinical isolates of Candida tropicalis and Candida parapsilosis. J Appl Microbiol 2022; 132:3601-3617. [PMID: 35179275 DOI: 10.1111/jam.15498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/28/2022]
Abstract
AIMS This study evaluated the antifungal, antibiofilm, and molecular docking of 2-Chloro-N-phenylacetamide against clinical isolates of Candida tropicalis and Candida parapsilosis. METHODS AND RESULTS MIC of the test drugs was determined by microdilution. A1Cl obtained MIC values ranging from 16 and 256 μg/mL. Fluconazole MIC ranging from 16 and 512 μg/mL. MIC of A1Cl showed fungicide activity, emphasizing the solid antifungal potential of this drug. An association study was performed with A1Cl and fluconazole (checkerboard), revealing indifference by decreasing. Thus, we conducted this study using A1Cl isolated. In the micromorphological assay, the test drugs reduced the production of virulence structures compared to the control (concentration-dependent effect). A1Cl inhibited in vitro biofilm formation at all concentrations tested (1/4MIC to 8xMIC) (p<0.05) and reduced mature biofilm biomass (p<0.05) against C. tropicalis and C. parapsilosis. In the ex vivo biofilm susceptibility testing (human nails fragments), A1Cl inhibited biofilm formation and reduced mature biofilm biomass (p<0.05) more than 50% at MIC. Fluconazole had a similar effect at 4xMIC. In silico studies suggest that the mechanism of antifungal activity of A1Cl involves the inhibition of the enzyme dihydrofolate reductase rather than geranylgeranyltransferase-I. CONCLUSIONS The results suggest that A1Cl is a promising antifungal agent. Furthermore, this activity is related to attenuation of expression of virulence factors and antibiofilm effects against C. tropicalis and C. parapsilosis. SIGNIFICANCE AND IMPACT OF THE STUDY Our study provides the first evidence that A1Cl, a novel synthetic drug, has fungicidal effects against C. tropicalis and C. parapsilosis. Furthermore, in vitro and ex vivo biofilms assays have demonstrated the potential antibiofilm of A1Cl. The mechanism of action involves inhibiting the enzyme dihydrofolate reductase, which was supported by in silico analyses. Therefore, this potential can be explored as a therapeutic alternative for onychomycosis and, at the same time, contribute to decreasing the resistance of clinical isolates of C. tropicalis and C. parapsilosis.
Collapse
Affiliation(s)
- Shellygton Lima Silva
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Fillipe de Oliveira Pereira
- Biochemistry Laboratory, Academic Unit of Health, Education and Health Center, Federal University of Campina Grande, Cuité, Brazil
| | - Laisa Vilar Cordeiro
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Hermes Diniz Neto
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Mayara Dos Santos Maia
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Helivado Diogenes da Silva Souza
- Bioenergy and Organic Synthesis Research Laboratory, Department of Chemistry, University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Petrônio F de Athayde-Filho
- Bioenergy and Organic Synthesis Research Laboratory, Department of Chemistry, University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Luciana Scotti
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Edeltrudes de Oliveira Lima
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| |
Collapse
|
195
|
Allert S, Schulz D, Kämmer P, Großmann P, Wolf T, Schäuble S, Panagiotou G, Brunke S, Hube B. From environmental adaptation to host survival: Attributes that mediate pathogenicity of Candida auris. Virulence 2022; 13:191-214. [PMID: 35142597 PMCID: PMC8837256 DOI: 10.1080/21505594.2022.2026037] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Candida species are a major cause of invasive fungal infections. While Candida albicans, C. glabrata, C. parapsilosis, and C. tropicalis are the most dominant species causing life-threatening candidiasis, C. auris recently emerged as a new species causing invasive infections with high rates of clinical treatment failures. To mimic initial phases of systemic Candida infections with dissemination via the bloodstream and to elucidate the pathogenic potential of C. auris, we used an ex vivo whole blood infection model. Similar to other clinically relevant Candida spp., C. auris is efficiently killed in human blood, but showed characteristic patterns of immune cell association, survival rates, and cytokine induction. Dual-species transcriptional profiling of C. auris-infected blood revealed a unique C. auris gene expression program during infection, while the host response proofed similar and conserved compared to other Candida species. C. auris-specific responses included adaptation and survival strategies, such as counteracting oxidative burst of immune cells, but also expression of potential virulence factors, (drug) transporters, and cell surface-associated genes. Despite comparable pathogenicity to other Candida species in our model, C. auris-specific transcriptional adaptations as well as its increased stress resistance and long-term environmental survival, likely contribute to the high risk of contamination and distribution in a nosocomial setting. Moreover, infections of neutrophils with pre-starved C. auris cells suggest that environmental preconditioning can have modulatory effects on the early host interaction. In summary, we present novel insights into C. auris pathogenicity, revealing adaptations to human blood and environmental niches distinctive from other Candida species.
Collapse
Affiliation(s)
- Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Daniela Schulz
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Philipp Kämmer
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Peter Großmann
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Thomas Wolf
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Sascha Schäuble
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Gianni Panagiotou
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany.,Department of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany.,Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
196
|
Cid-Chevecich C, Müller-Sepúlveda A, Jara JA, López-Muñoz R, Santander R, Budini M, Escobar A, Quijada R, Criollo A, Díaz-Dosque M, Molina-Berríos A. Origanum vulgare L. essential oil inhibits virulence patterns of Candida spp. and potentiates the effects of fluconazole and nystatin in vitro. BMC Complement Med Ther 2022; 22:39. [PMID: 35139827 PMCID: PMC8827202 DOI: 10.1186/s12906-022-03518-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Background Recurrence and resistance of Candida spp. infections is associated with the ability of these microorganisms to present several virulence patterns such as morphogenesis, adhesion, and biofilm formation. In the search for agents with antivirulence activity, essential oils could represent a strategy to act against biofilms and to potentiate antifungal drugs. Objective To evaluate the antivirulence effect of Origanum vulgare L. essential oil (O-EO) against Candida spp. and to potentiate the effect of fluconazole and nystatin. Methods The effect of O-EO was evaluated on ATCC reference strains of C. albicans and non-albicans Candida species. Minimum inhibitory concentration (MIC) was determined through broth microdilution assay. Adhesion to microplates was determined by crystal violet (CV) assay. An adapted scratch assay in 24-well was used to determine the effect of essential oil on biofilms proliferation. Viability of biofilms was evaluated by MTT reduction assay and through a checkerboard assay we determined if O-EO could act synergistically with fluconazole and nystatin. Results MIC for C. albicans ATCC-90029 and ATCC-10231 was 0.01 mg/L and 0.97 mg/L, respectively. For non-albicans Candida strains MIC values were 2.6 mg/L for C. dubliniensis ATCC-CD36 and 5.3 mg/L for C. krusei ATCC-6258. By using these concentrations, O-EO inhibited morphogenesis, adhesion, and proliferation at least by 50% for the strains assayed. In formed biofilms O-EO decreased viability in ATCC 90029 and ATCC 10231 strains (IC50 7.4 and 2.8 mg/L respectively). Finally, we show that O-EO interacted synergistically with fluconazole and nystatin. Conclusions This study demonstrate that O-EO could be considered to improve the antifungal treatment against Candida spp. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03518-z.
Collapse
Affiliation(s)
- Camila Cid-Chevecich
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Andrea Müller-Sepúlveda
- Institute of Agrifood, Animals and Environmental Sciences, Universidad de O'Higgins, San Fernando, Chile
| | - José Antonio Jara
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Rodrigo López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Rocío Santander
- Department of Environmental Sciences, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Mauricio Budini
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Alejandro Escobar
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Raúl Quijada
- Faculty of Physical and Mathematical Sciences, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile
| | - Mario Díaz-Dosque
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile.
| | - Alfredo Molina-Berríos
- Laboratory of Pharmacology, Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, University of Chile, Olivos 943, Independencia, Santiago, Chile.
| |
Collapse
|
197
|
Le PH, Nguyen DHK, Medina AA, Linklater DP, Loebbe C, Crawford RJ, MacLaughlin S, Ivanova EP. Surface Architecture Influences the Rigidity of Candida albicans Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:567. [PMID: 35159912 PMCID: PMC8840568 DOI: 10.3390/nano12030567] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023]
Abstract
Atomic force microscopy (AFM) was used to investigate the morphology and rigidity of the opportunistic pathogenic yeast, Candida albicans ATCC 10231, during its attachment to surfaces of three levels of nanoscale surface roughness. Non-polished titanium (npTi), polished titanium (pTi), and glass with respective average surface roughness (Sa) values of 389 nm, 14 nm, and 2 nm, kurtosis (Skur) values of 4, 16, and 4, and skewness (Sskw) values of 1, 4, and 1 were used as representative examples of each type of nanoarchitecture. Thus, npTi and glass surfaces exhibited similar Sskw and Skur values but highly disparate Sa. C. albicans cells that had attached to the pTi surfaces exhibited a twofold increase in rigidity of 364 kPa compared to those yeast cells attached to the surfaces of npTi (164 kPa) and glass (185 kPa). The increased rigidity of the C. albicans cells on pTi was accompanied by a distinct round morphology, condensed F-actin distribution, lack of cortical actin patches, and the negligible production of cell-associated polymeric substances; however, an elevated production of loose extracellular polymeric substances (EPS) was observed. The differences in the physical response of C. albicans cells attached to the three surfaces suggested that the surface nanoarchitecture (characterized by skewness and kurtosis), rather than average surface roughness, could directly influence the rigidity of the C. albicans cells. This work contributes to the next-generation design of antifungal surfaces by exploiting surface architecture to control the extent of biofilm formation undertaken by yeast pathogens and highlights the importance of performing a detailed surface roughness characterization in order to identify and discriminate between the surface characteristics that may influence the extent of cell attachment and the subsequent behavior of the attached cells.
Collapse
Affiliation(s)
- Phuc H. Le
- STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia; (P.H.L.); (D.H.K.N.); (A.A.M.); (D.P.L.); (R.J.C.)
- ARC Research Hub for Australian Steel Manufacturing, STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Duy H. K. Nguyen
- STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia; (P.H.L.); (D.H.K.N.); (A.A.M.); (D.P.L.); (R.J.C.)
| | - Arturo Aburto Medina
- STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia; (P.H.L.); (D.H.K.N.); (A.A.M.); (D.P.L.); (R.J.C.)
- ARC Research Hub for Australian Steel Manufacturing, STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Denver P. Linklater
- STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia; (P.H.L.); (D.H.K.N.); (A.A.M.); (D.P.L.); (R.J.C.)
| | | | - Russell J. Crawford
- STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia; (P.H.L.); (D.H.K.N.); (A.A.M.); (D.P.L.); (R.J.C.)
| | | | - Elena P. Ivanova
- STEM College, School of Science, RMIT University, Melbourne, VIC 3000, Australia; (P.H.L.); (D.H.K.N.); (A.A.M.); (D.P.L.); (R.J.C.)
| |
Collapse
|
198
|
Kovács R, Majoros L. Antifungal lock therapy: an eternal promise or an effective alternative therapeutic approach? Lett Appl Microbiol 2022; 74:851-862. [PMID: 35032330 PMCID: PMC9306927 DOI: 10.1111/lam.13653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/02/2022] [Accepted: 01/07/2022] [Indexed: 11/30/2022]
Abstract
Each year, millions of central venous catheter insertions are performed in intensive care units worldwide. The usage of these indwelling devices is associated with a high risk of bacterial and fungal colonization, leading to the development of microbial consortia, namely biofilms. These sessile structures provide fungal cells with resistance to the majority of antifungals, environmental stress and host immune responses. Based on different guidelines, colonized/infected catheters should be removed and changed immediately in the case of Candida‐related central line infections. However, catheter replacement is not feasible for all patient populations. An alternative therapeutic approach may be antifungal lock therapy, which has received high interest, especially in the last decade. This review summarizes the published Candida‐related in vitro, in vivo data and case studies in terms of antifungal lock therapy. The number of clinical studies remains limited and further studies are needed for safe implementation of the antifungal lock therapy into clinical practice.
Collapse
Affiliation(s)
- Renátó Kovács
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Hungary.,Faculty of Pharmacy, University of Debrecen, Hungary
| | - László Majoros
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Hungary
| |
Collapse
|
199
|
Amphotericin B Polymer Nanoparticles Show Efficacy against Candida Species Biofilms. Pathogens 2022; 11:pathogens11010073. [PMID: 35056021 PMCID: PMC8781556 DOI: 10.3390/pathogens11010073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose: Chronic infections of Candida albicans are characterised by the embedding of budding and entwined filamentous fungal cells into biofilms. The biofilms are refractory to many drugs and Candida biofilms are associated with ocular fungal infections. The objective was to test the activity of nanoparticulate amphotericin B (AmB) against Candida biofilms. Methods: AmB was encapsulated in the Molecular Envelope Technology (MET, N-palmitoyl-N-monomethyl-N,N-dimethyl-N,N,N-trimethyl-6-O-glycolchitosan) nanoparticles and tested against Candida biofilms in vitro. Confocal laser scanning microscopy (CLSM) imaging of MET nanoparticles’ penetration into experimental biofilms was carried out and a MET-AmB eye drop formulation was tested for its stability. Results: MET-AmB formulations demonstrated superior activity towards C. albicans biofilms in vitro with the EC50 being ~30 times lower than AmB alone (EC50 MET-AmB = 1.176 μg mL−1, EC50 AmB alone = 29.09 μg mL−1). A similar superior activity was found for Candida glabrata biofilms, where the EC50 was ~10× lower than AmB alone (EC50 MET-AmB = 0.0253 μg mL−1, EC50 AmB alone = 0.289 μg mL−1). CLSM imaging revealed that MET nanoparticles penetrated through the C. albicans biofilm matrix and bound to fungal cells. The activity of MET-AmB was no different from the activity of AmB alone against C. albicans cells in suspension (MET-AmB MIC90 = 0.125 μg mL−1, AmB alone MIC90 = 0.250 μg mL−1). MET-AmB eye drops were stable at room temperature for at least 28 days. Conclusions: These biofilm activity findings raise the possibility that MET-loaded nanoparticles may be used to tackle Candida biofilm infections, such as refractory ocular fungal infections.
Collapse
|
200
|
Tortorano AM, Prigitano A, Morroni G, Brescini L, Barchiesi F. Candidemia: Evolution of Drug Resistance and Novel Therapeutic Approaches. Infect Drug Resist 2022; 14:5543-5553. [PMID: 34984009 PMCID: PMC8702982 DOI: 10.2147/idr.s274872] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022] Open
Abstract
Candidemia and invasive candidiasis are the most common healthcare-associated invasive fungal infections, with a crude mortality rate of 25–50%. Candida albicans remains the most frequent etiology, followed by C. glabrata, C. parapsilosis and C. tropicalis. With the exception of a limited number of species (ie: C. krusei, C. glabrata and rare Candida species), resistance to fluconazole and other triazoles are quite uncommon. However, recently fluconazole-resistant C. parapsilosis, echinocandin-resistant C. glabrata and the multidrug resistant C. auris have emerged. Resistance to amphotericin B is even more rare due to the reduced fitness of resistant isolates. The mechanisms of antifungal resistance in Candida (altered drug-target interactions, reduced cellular drug concentrations, and physical barriers associated with biofilms) are analyzed. The choice of the antifungal therapy for candidemia must take into account several factors such as type of patient, presence of devices, severity of illness, recent exposure to antifungals, local epidemiology, organs involvement, and Candida species. The first-line therapy in non-neutropenic critical patient is an echinocandin switching to fluconazole in clinically stable patients with negative blood cultures and azole susceptible isolate. Similarly, an echinocandin is the drug of choice also in neutropenic patients. The treatment duration is 14 days after the first negative blood culture or longer in cases of organ involvement. An early removal of vascular catheter improves the outcome. The promising results of new antifungal molecules, such as the terpenoid derivative ibrexafungerp, the novel echinocandin with an enhanced half-life rezafungin, oteseconazole and fosmanogepix, representative of new classes of antifungals, are discussed.
Collapse
Affiliation(s)
- Anna Maria Tortorano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milano, Italy
| | - Anna Prigitano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milano, Italy
| | - Gianluca Morroni
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Lucia Brescini
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy.,Clinic of Infectious Diseases, Azienda Ospedaliero Universitaria, Ospedali Riuniti Umberto I-Lancisi-Salesi, Ancona, Italy
| | - Francesco Barchiesi
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy.,Clinic of Infectious Diseases, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| |
Collapse
|