101
|
Zeng H, Stadler M, Abraham WR, Müsken M, Schrey H. Inhibitory Effects of the Fungal Pigment Rubiginosin C on Hyphal and Biofilm Formation in Candida albicans and Candida auris. J Fungi (Basel) 2023; 9:726. [PMID: 37504715 PMCID: PMC10381533 DOI: 10.3390/jof9070726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023] Open
Abstract
The two fungal human pathogens, Candida auris and Candida albicans, possess a variety of virulence mechanisms. Among them are the formation of biofilms to protect yeast against harsh conditions through the development of (pseudo)hyphae whilst also facilitating the invasion of host tissues. In recent years, increased rates of antifungal resistance have been associated with C. albicans and C. auris, posing a significant challenge for the effective treatment of fungal infections. In the course of our ongoing search for novel anti-infectives, six selected azaphilones were tested for their cytotoxicity and antimicrobial effects as well as for their inhibitory activity against biofilm and hyphal formation. This study revealed that rubiginosin C, derived from stromata of the ascomycete Hypoxylon rubiginosum, effectively inhibited the formation of biofilms, pseudohyphae, and hyphae in both C. auris and C. albicans without lethal effects. Crystal violet staining assays were utilized to assess the inhibition of biofilm formation, while complementary microscopic techniques, such as confocal laser scanning microscopy, scanning electron microscopy, and optical microscopy, were used to investigate the underlying mechanisms. Rubiginosin C is one of the few substances known to effectively target both biofilm formation and the yeast-to-hyphae transition of C. albicans and C. auris within a concentration range not affecting host cells, making it a promising candidate for therapeutic intervention in the future.
Collapse
Affiliation(s)
- Haoxuan Zeng
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Marc Stadler
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Wolf-Rainer Abraham
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Hedda Schrey
- Department of Microbial Drugs, Helmholtz Centre for Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| |
Collapse
|
102
|
Marcus JE, Ford MB, Sattler LA, Iqbal S, Garner CL, Sobieszczyk MJ, Barsoumian AE. Treatment and outcome of gram-positive bacteremia in patients receiving extracorporeal membrane oxygenation. Heart Lung 2023; 60:15-19. [PMID: 36871407 DOI: 10.1016/j.hrtlng.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND While guidance exists for management of blood stream infections with various invasive devices, there are currently limited data to guide antibiotic selection and duration for bacteremia in patients receiving extracorporeal membrane oxygenation (ECMO). OBJECTIVE To evaluate the treatment and outcomes of thirty-six patients with Staphylococcus aureus and Enterococcus bacteremia on ECMO support. METHODS Blood culture data was retrospectively analyzed from patients with Staphylococcus aureus bacteremia (SAB) or Enterococcus bacteremia who underwent ECMO support between March 2012 and September 2021 at Brooke Army Medical Center. RESULTS Of the 282 patients who received ECMO during this study period, there 25 (9%) patients developed Enterococcus bacteremia and 16 (6%) developed SAB. SAB occurred earlier in ECMO as compared to Enterococcus (median day 2 IQR (1-5) vs. 22 (12-51), p = 0.01). The most common duration of antibiotics was 28 days after clearance for SAB and 14 days after clearance for Enterococcus. 2 (5%) patients underwent cannula exchange with primary bacteremia, and 7 (17%) underwent circuit exchange. 1/3 (33%) patients with SAB and 3/10 (30%) patients with Enterococcus bacteremia who remained cannulated after completion of antibiotics had a second episode of SAB or Enterococcus bacteremia. CONCLUSION This single center case series is the first to describe the specific treatment and outcomes of patients receiving ECMO complicated by SAB and Enterococcus bacteremia. For patients who remain on ECMO after completion of antibiotics, there is a risk of a second episode of Enterococcus bacteremia or SAB.
Collapse
Affiliation(s)
- Joseph E Marcus
- Infectious Disease Service, Department of Medicine, Brooke Army Medical Center 3551 Roger Brooke Drive, Joint Base San Antonio, TX 78234 United States; Department of Medicine, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 United States.
| | - Mary B Ford
- Infectious Disease Service, Department of Medicine, Brooke Army Medical Center 3551 Roger Brooke Drive, Joint Base San Antonio, TX 78234 United States; Department of Medicine, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 United States
| | - Lauren A Sattler
- Department of Medicine, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 United States; Pulmonary and Critical Care Section, Washington University in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110 United States
| | - Sonia Iqbal
- Department of Medicine, Andrews Air Force Base, 1050 West Perimeter Road, Joint Base Andrew AFB, MD 20762 United States
| | - Chelsea L Garner
- Pulmonary and Critical Care Service, Department of Medicine, Brooke Army Medical Center, 3551 Roger Brooke Drive, Joint Base San Antonio, TX 78234 United States
| | - Michal J Sobieszczyk
- Department of Medicine, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 United States; Pulmonary and Critical Care Service, Department of Medicine, Brooke Army Medical Center, 3551 Roger Brooke Drive, Joint Base San Antonio, TX 78234 United States
| | - Alice E Barsoumian
- Infectious Disease Service, Department of Medicine, Brooke Army Medical Center 3551 Roger Brooke Drive, Joint Base San Antonio, TX 78234 United States; Department of Medicine, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 United States
| |
Collapse
|
103
|
Cabezas-Mera FS, Atiencia-Carrera MB, Villacrés-Granda I, Proaño AA, Debut A, Vizuete K, Herrero-Bayo L, Gonzalez-Paramás AM, Giampieri F, Abreu-Naranjo R, Tejera E, Álvarez-Suarez JM, Machado A. Evaluation of the polyphenolic profile of native Ecuadorian stingless bee honeys ( Tribe: Meliponini) and their antibiofilm activity on susceptible and multidrug-resistant pathogens: An exploratory analysis. Curr Res Food Sci 2023; 7:100543. [PMID: 37455680 PMCID: PMC10344713 DOI: 10.1016/j.crfs.2023.100543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/08/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023] Open
Abstract
Biofilms are associated with infections that are resistant to conventional therapies, contributing to the antimicrobial resistance crisis. The need for alternative approaches against biofilms is well-known. Although natural products like stingless bee honeys (tribe: Meliponini) constitute an alternative treatment, much is still unknown. Our main goal was to evaluate the antibiofilm activity of stingless bee honey samples against multidrug-resistant (MDR) pathogens through biomass assays, fluorescence (cell count and viability), and scanning electron (structural composition) microscopy. We analyzed thirty-five honey samples at 15% (v/v) produced by ten different stingless bee species (Cephalotrigona sp., Melipona sp., M. cramptoni, M. fuscopilosa, M. grandis, M. indecisa, M. mimetica, M. nigrifacies, Scaptotrigona problanca, and Tetragonisca angustula) from five provinces of Ecuador (Tungurahua, Pastaza, El Oro, Los Ríos, and Loja) against 24-h biofilms of Staphylococcus aureus, Klebsiella pneumoniae, Candida albicans, and Candida tropicalis. The present honey set belonged to our previous study, where the samples were collected in 2018-2019 and their physicochemical parameters, chemical composition, mineral elements, and minimal inhibitory concentration (MIC) were screened. However, the polyphenolic profile and their antibiofilm activity on susceptible and multidrug-resistant pathogens were still unknown. According to polyphenolic profile of the honey samples, significant differences were observed according to their geographical origin in terms of the qualitative profiles. The five best honey samples (OR24.1, LR34, LO40, LO48, and LO53) belonging to S. problanca, Melipona sp., and M. indecisa were selected for further analysis due to their high biomass reduction values, identification of the stingless bee specimens, and previously reported physicochemical parameters. This subset of honey samples showed a range of 63-80% biofilm inhibition through biomass assays. Fluorescence microscopy (FM) analysis evidenced statistical log reduction in the cell count of honey-treated samples in all pathogens (P <0.05), except for S. aureus ATCC 25923. Concerning cell viability, C. tropicalis, K. pneumoniae ATCC 33495, and K. pneumoniae KPC significantly decreased (P <0.01) by 21.67, 25.69, and 45.62%, respectively. Finally, scanning electron microscopy (SEM) analysis demonstrated structural biofilm disruption through cell morphological parameters (such as area, size, and form). In relation to their polyphenolic profile, medioresinol was only found in the honey of Loja, while scopoletin, kaempferol, and quercetin were only identified in honey of Los Rios, and dihydrocaffeic and dihydroxyphenylacetic acids were only detected in honey of El Oro. All the five honey samples showed dihydrocoumaroylhexose, luteolin, and kaempferol rutinoside. To the authors' best knowledge, this is the first study to analyze stingless bees honey-treated biofilms of susceptible and/or MDR strains of S. aureus, K. pneumoniae, and Candida species.
Collapse
Affiliation(s)
- Fausto Sebastián Cabezas-Mera
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Quito, 170901, Ecuador
| | - María Belén Atiencia-Carrera
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Quito, 170901, Ecuador
| | - Irina Villacrés-Granda
- Programa de Doctorado Interuniversitario en Ciencias de la Salud, Universidad de Sevilla, Sevilla, Spain
- Facultad de Ingeniería y Ciencias Agropecuarias Aplicadas, Grupo de Bioquimioinformática, Universidad de Las Américas (UDLA), De Los Colimes esq, Quito, 170513, Quito, Ecuador
| | - Adrian Alexander Proaño
- Laboratorios de Investigación, Universidad de Las Américas (UDLA), Vía a Nayón, Quito, 170124, Ecuador
| | - Alexis Debut
- Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas ESPE, Sangolquí, 171103, Ecuador
- Centro de Nanociencia y Nanotecnología, Universidad de Las Fuerzas Armadas ESPE, Sangolquí, 171103, Ecuador
| | - Karla Vizuete
- Centro de Nanociencia y Nanotecnología, Universidad de Las Fuerzas Armadas ESPE, Sangolquí, 171103, Ecuador
| | - Lorena Herrero-Bayo
- Grupo de Investigación en Polifenoles (GIP-USAL), Universidad de Salamanca, Campus Miguel de Unamuno, 37008, Salamanca, Spain
| | - Ana M. Gonzalez-Paramás
- Grupo de Investigación en Polifenoles (GIP-USAL), Universidad de Salamanca, Campus Miguel de Unamuno, 37008, Salamanca, Spain
| | - Francesca Giampieri
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, C. Isabel Torres, 21, 39011, Santander, Cantabria, Spain
| | - Reinier Abreu-Naranjo
- Departamento de Ciencias de La Vida, Universidad Estatal Amazónica, Puyo, 160150, Ecuador
| | - Eduardo Tejera
- Facultad de Ingeniería y Ciencias Agropecuarias Aplicadas, Grupo de Bioquimioinformática, Universidad de Las Américas (UDLA), De Los Colimes esq, Quito, 170513, Quito, Ecuador
| | - José M. Álvarez-Suarez
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias e Ingenierías, Departamento de Ingeniería en Alimentos, Calle Diego de Robles y Pampite, Quito, 170901, Ecuador
| | - António Machado
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Quito, 170901, Ecuador
| |
Collapse
|
104
|
Perez-Gonzalez G, Tompsett GA, Mastalerz K, Timko MT, Goodell B. Interaction of oxalate with β-glucan: Implications for the fungal extracellular matrix, and metabolite transport. iScience 2023; 26:106851. [PMID: 37275522 PMCID: PMC10232728 DOI: 10.1016/j.isci.2023.106851] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/28/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
β-glucan is the major component of the extracellular matrix (ECM) of many fungi, including wood degrading fungi. Many of these species also secrete oxalate into the ECM. Our research demonstrates that β-glucan forms a novel, previously unreported, hydrogel at room temperature with oxalate. Oxalate was found to alter the rheometric properties of the β-glucan hydrogels, and modeling showed that β-glucan hydrogen bonds with oxalate in a non-covalent matrix. Change of oxalate concentration also impacted the diffusion of a high-molecular-weight protein through the gels. This finding has relevance to the diffusion of extracellular enzymes into substrates and helps to explain why some types of wood-decay fungi rely on non-enzymatic degradation schemes for carbon cycling. Further, this research has potential impact on the diffusion of metabolites in association with pathogenic/biomedical fungi.
Collapse
Affiliation(s)
| | - Geoffrey A. Tompsett
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA 01609, USA
| | - Kyle Mastalerz
- Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA
| | - Michael T. Timko
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA 01609, USA
| | - Barry Goodell
- Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
105
|
Comitini F, Canonico L, Agarbati A, Ciani M. Biocontrol and Probiotic Function of Non- Saccharomyces Yeasts: New Insights in Agri-Food Industry. Microorganisms 2023; 11:1450. [PMID: 37374952 DOI: 10.3390/microorganisms11061450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Fermented food matrices, including beverages, can be defined as the result of the activity of complex microbial ecosystems where different microorganisms interact according to different biotic and abiotic factors. Certainly, in industrial production, the technological processes aim to control the fermentation to place safe foods on the market. Therefore, if food safety is the essential prerogative, consumers are increasingly oriented towards a healthy and conscious diet driving the production and consequently the applied research towards natural processes. In this regard, the aim to guarantee the safety, quality and diversity of products should be reached limiting or avoiding the addition of antimicrobials or synthetic additives using the biological approach. In this paper, the recent re-evaluation of non-Saccharomyces yeasts (NSYs) has been reviewed in terms of bio-protectant and biocontrol activity with a particular focus on their antimicrobial power using different application modalities including biopackaging, probiotic features and promoting functional aspects. In this review, the authors underline the contribution of NSYs in the food production chain and their role in the technological and fermentative features for their practical and useful use as a biocontrol agent in food preparations.
Collapse
Affiliation(s)
- Francesca Comitini
- Dipartimento di Scienze della Vita e dell'Ambiente (DiSVA), Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Laura Canonico
- Dipartimento di Scienze della Vita e dell'Ambiente (DiSVA), Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Alice Agarbati
- Dipartimento di Scienze della Vita e dell'Ambiente (DiSVA), Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Maurizio Ciani
- Dipartimento di Scienze della Vita e dell'Ambiente (DiSVA), Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| |
Collapse
|
106
|
Soriano A, Honore PM, Puerta-Alcalde P, Garcia-Vidal C, Pagotto A, Gonçalves-Bradley DC, Verweij PE. Invasive candidiasis: current clinical challenges and unmet needs in adult populations. J Antimicrob Chemother 2023:7176280. [PMID: 37220664 DOI: 10.1093/jac/dkad139] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
Invasive candidiasis (IC) is a serious infection caused by several Candida species, and the most common fungal disease in hospitals in high-income countries. Despite overall improvements in health systems and ICU care in the last few decades, as well as the development of different antifungals and microbiological techniques, mortality rates in IC have not substantially improved. The aim of this review is to summarize the main issues underlying the management of adults affected by IC, focusing on specific forms of the infection: IC developed by ICU patients, IC observed in haematological patients, breakthrough candidaemia, sanctuary site candidiasis, intra-abdominal infections and other challenging infections. Several key challenges need to be tackled to improve the clinical management and outcomes of IC patients. These include the lack of global epidemiological data for IC, the limitations of the diagnostic tests and risk scoring tools currently available, the absence of standardized effectiveness outcomes and long-term data for IC, the timing for the initiation of antifungal therapy and the limited recommendations on the optimal step-down therapy from echinocandins to azoles or the total duration of therapy. The availability of new compounds may overcome some of the challenges identified and increase the existing options for management of chronic Candida infections and ambulant patient treatments. However, early identification of patients that require antifungal therapy and treatment of sanctuary site infections remain a challenge and will require further innovations.
Collapse
Affiliation(s)
- Alex Soriano
- Department of Infectious Diseases, Hospital Clinic of Barcelona, IDIBAPS, CIBERINF, University of Barcelona, Barcelona, Spain
| | - Patrick M Honore
- CHU UCL Godinne Namur, UCL Louvain Medical School, Namur, Belgium
| | - Pedro Puerta-Alcalde
- Department of Infectious Diseases, Hospital Clinic of Barcelona, IDIBAPS, CIBERINF, University of Barcelona, Barcelona, Spain
| | - Carolina Garcia-Vidal
- Department of Infectious Diseases, Hospital Clinic of Barcelona, IDIBAPS, CIBERINF, University of Barcelona, Barcelona, Spain
| | | | | | - Paul E Verweij
- Radboudumc-CWZ Center of Expertise for Mycology, Nijmegen, the Netherlands
| |
Collapse
|
107
|
Martins da Silva Filho P, Higor Rocha Mariano P, Lopes Andrade A, Barros Arrais Cruz Lopes J, de Azevedo Pinheiro A, Itala Geronimo de Azevedo M, Carneiro de Medeiros S, Alves de Vasconcelos M, Gonçalvez da Cruz Fonseca S, Barbosa Grangeiro T, Gonzaga de França Lopes L, Henrique Silva Sousa E, Holanda Teixeira E, Longhinotti E. Antibacterial and antifungal action of CTAB-containing silica nanoparticles against human pathogens. Int J Pharm 2023; 641:123074. [PMID: 37230370 DOI: 10.1016/j.ijpharm.2023.123074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/16/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
New antibiotic agents are urgently needed worldwide to combat the increasing tolerance and resistance of pathogenic fungi and bacteria to current antimicrobials. Here, we looked at the antibacterial and antifungal effects of minor quantities of cetyltrimethylammonium bromide (CTAB), ca. 93.8 mg g-1, on silica nanoparticles (MPSi-CTAB). Our results show that MPSi-CTAB exhibits antimicrobial activity against Methicillin-resistant Staphylococcus aureus strain (S. aureus ATCC 700698) with MIC and MBC of 0.625 mg mL-1 and 1.25 mg mL-1, respectively. Additionally, for Staphylococcus epidermidis ATCC 35984, MPSi-CTAB reduces MIC and MBC by 99.99% of viable cells on the biofilm. Furthermore, when combined with ampicillin or tetracycline, MPSi-CTAB exhibits reduced MIC values by 32- and 16-folds, respectively. MPSi-CTAB also exhibited in vitro antifungal activity against reference strains of Candida, with MIC values ranging from 0.0625 to 0.5 mg mL-1. This nanomaterial has low cytotoxicity in human fibroblasts, where over 80% of cells remained viable at 0.31 mg mL-1 of MPSi-CTAB. Finally, we developed a gel formulation of MPSi-CTAB, which inhibited in vitro the growth of Staphylococcus and Candida strains. Overall, these results support the efficacy of MPSi-CTAB with potential application in the treatment and/or prevention of infections caused by methicillin-resistant Staphylococcus and/or Candida species.
Collapse
Affiliation(s)
- Pedro Martins da Silva Filho
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará 60440-900 Fortaleza - CE, Brazil; Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza - CE, Brazil.
| | - Pedro Higor Rocha Mariano
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará 60440-900 Fortaleza - CE, Brazil
| | - Alexandre Lopes Andrade
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP 60430-270, Fortaleza - CE, Brazil
| | - Jessica Barros Arrais Cruz Lopes
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP 60430-270, Fortaleza - CE, Brazil
| | - Aryane de Azevedo Pinheiro
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP 60430-270, Fortaleza - CE, Brazil
| | | | - Suelen Carneiro de Medeiros
- Departamento de Biologia, Universidade Federal do Ceará, Campus do Pici s/n, 60440-900, Fortaleza - CE, Brazil
| | - Mayron Alves de Vasconcelos
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP 60430-270, Fortaleza - CE, Brazil; Departamento de Ciências Biológicas, Faculdade de Ciências Exatas e Naturais, Universidade do Estado do Rio Grande do Norte, 59610-090, Mossoró - RN, Brazil; Universidade do Estado de Minas Gerais, Unidade de Divinópolis, 35501-170, Divinópolis - MG, Brazil
| | | | - Thalles Barbosa Grangeiro
- Departamento de Biologia, Universidade Federal do Ceará, Campus do Pici s/n, 60440-900, Fortaleza - CE, Brazil
| | - Luiz Gonzaga de França Lopes
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza - CE, Brazil
| | - Eduardo Henrique Silva Sousa
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza - CE, Brazil.
| | - Edson Holanda Teixeira
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP 60430-270, Fortaleza - CE, Brazil
| | - Elisane Longhinotti
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará 60440-900 Fortaleza - CE, Brazil.
| |
Collapse
|
108
|
Wijaya M, Halleyantoro R, Kalumpiu JF. Biofilm: The invisible culprit in catheter-induced candidemia. AIMS Microbiol 2023; 9:467-485. [PMID: 37649801 PMCID: PMC10462453 DOI: 10.3934/microbiol.2023025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 09/01/2023] Open
Abstract
Candidemia is the most common form of invasive fungal infection associated with several risk factors, and one of them is the use of medical devices, to which microbial biofilms can attach. Candidemia related to the use of peripheral intravascular and central venous catheters (CVC) is referred to as Candida catheter-related bloodstream infection, with more than 90% being related to CVC usage. The infection is associated with a higher morbidity and mortality rate than nosocomial bacterial infections. Candida spp. can protect themselves from the host immune system and antifungal drugs because of the biofilm structure, which is potentiated by the extracellular matrix (ECM). Candida albicans and Candida parapsilosis are the most pathogenic species often found to form biofilms associated with catheter usage. Biofilm formation of C. albicans includes four mechanisms: attachment, morphogenesis, maturation and dispersion. The biofilms formed between C. albicans and non-albicans spp. differ in ECM structure and composition and are associated with the persistence of colonization to infection for various catheter materials and antifungal resistance. Efforts to combat Candida spp. biofilm formation on catheters are still challenging because not all patients, especially those who are critically ill, can be recommended for catheter removal; also to be considered are the characteristics of the biofilm itself, which readily colonizes the permanent medical devices used. The limited choice and increasing systemic antifungal resistance also make treating it more difficult. Hence, alternative strategies have been developed to manage Candida biofilm. Current options for prevention or therapy in combination with systemic antifungal medications include lock therapy, catheter coating, natural peptide products and photodynamic inactivation.
Collapse
Affiliation(s)
- Meiliyana Wijaya
- Department of Parasitology, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Ryan Halleyantoro
- Department of Parasitology, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Jane Florida Kalumpiu
- Department of Parasitology, Faculty of Medicine, Pelita Harapan University, Banten, Indonesia
| |
Collapse
|
109
|
Mohammadi F, Charkhchian M, Mirzadeh M. Phenotypic and genotypic characterization of virulence markers and antifungal susceptibility of oral Candida species from diabetic and non-diabetic hemodialysis patients. BMC Oral Health 2023; 23:261. [PMID: 37143002 PMCID: PMC10157964 DOI: 10.1186/s12903-023-02970-8] [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: 12/03/2022] [Accepted: 04/16/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Patients with chronic kidney disease undergoing hemodialysis are often colonized by Candida species with high possibility of fungal infections. The purposes of this study were to determine the prevalence of Candida species, evaluate antifungal susceptibility profile, biofilm formation, proteinase and phospholipase activities, and the frequency of virulence genes in the Candida species isolated from the oral mucosa of hemodialysis diabetic (DM) and non-diabetic (non-DM) patients. METHODS This study identified several species of Candida isolated from 69 DM and 58 non-DM patients on hemodialysis using phenotypic methods and PCR-RFLP technique. The identification of C. albicans and C. glabrata complex was performed by HWP1 gene and four oligonucleotides (UNI-5.8S, GLA-f, BRA-f, and NIV-f), respectively. Antifungal susceptibility to amphotericin B, fluconazole, itraconazole, voriconazole, and caspofungin was assessed according to CLSI M27-A3/S4. The biomass, metabolic activity of biofilm, proteinase (Prz), phospholipase (Pz), and molecular study for virulence genes were assessed using crystal violet, XTT assay, agar-based hydrolytic enzyme, and PCR technique, respectively. RESULTS Candida prevalence was 44.9% with 47.8% and 41.4% among DM and non-DM patients, respectively (P = .045). The species identified were C. albicans (49.5%), C. glabrata (16.5%), C. tropicalis (12%), C. kefyr (8.8%), C. parapsilosis (6.6%), C. dubliniensis (3.3%), and C. lusitaniae (3.3%). The antifungal susceptibility profile showed that all Candida isolates were sensitive to amphotericin B, itraconazole, voriconazole, and caspofungin whereas fluconazole resistance was observed in 6.3% (MIC ≥ 64 μg/mL) of C. albicans and 6.6% of C. glabrata (MIC ≥ 64 μg/mL). The susceptible- dose-dependent rate was found in 10.5% of C. albicans. The Prz values of C. albicans ranged from 0.37 to 0.66 for the DM and 0.44-0.73 for the non-DM group (P < 0.05). The non-albicans Candida (NAC) species produced higher degree of biomass and metabolic activity compared to C. albicans (P < 0.05). Furthermore, significant (p < 0.05) correlations were detected between the biofilm formation with Prz values and fluconazole MICs. The most detected virulence factors were ALS3 and Sap5. CONCLUSIONS These results showed the importance of prevalence of NAC species in hemodialysis patients. Investigating antifungal susceptibility profile made a better understanding of the role of virulence markers in the pathogenesis of Candida strains.
Collapse
Affiliation(s)
- Faezeh Mohammadi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran.
- Department of Medical Parasitology and Mycology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Maliheh Charkhchian
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Monirsadat Mirzadeh
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
110
|
Bashir A, Ahmad T, Farooq S, Lone WI, Manzoor MM, Nalli Y, Sultan P, Chaubey A, Ali A, Riyaz-Ul-Hassan S. A Secondary Metabolite of Cercospora sp., Associated with Rosa damascena Mill., Inhibits Proliferation, Biofilm Production, Ergosterol Synthesis and Other Virulence Factors in Candida albicans. MICROBIAL ECOLOGY 2023; 85:1276-1287. [PMID: 35366684 DOI: 10.1007/s00248-022-02003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/22/2022] [Indexed: 05/10/2023]
Abstract
Here we describe the antimicrobial potential of secondary metabolites, fulvic acid (F.A.) and anhydrofulvic acid (AFA), produced by RDE147, an endophyte of Rosa damascena Mill. The endophyte was identified as Cercospora piaropi by ITS and β-tubulin-based phylogenetic analyses, while chemoprofiling of the endophyte by column chromatography and spectroscopy yielded two pure compounds, F.A. and AFA. The compounds demonstrated different antimicrobial profiles, with AFA suppressing the growth of C. albicans at 7.3 µg ml-1 IC50. Further studies revealed that AFA strongly restricted the biofilm production and hyphae formation in C. albicans by down-regulating several biofilm and morphogenesis-related genes. The time-kill assays confirmed the fungicidal activity of AFA against C. albicans, killing 83.6% of the pathogen cells in 24 h at the MIC concentration, and the post-antibiotic effect (PAE) experiments established the suppression of C. albicans growth for extended time periods. The compound acted synergistically with amphotericin B and nystatin and reduced ergosterol biosynthesis by the pathogen, confirmed by ergosterol estimation and comparative expression profiling of selected genes and molecular docking of AFA with C. albicans squalene epoxidase. AFA also suppressed the expression of several other virulence genes of the fungal pathogen. The study determines the anti-C. albicans potential of AFA and its impact on the biology of the pathogen. It also indicates that Cercospora species may yield potential bioactive molecules, especially fulvic acid derivatives. However, it is imperative to conduct in vivo studies to explore this molecule's therapeutic potential further.
Collapse
Affiliation(s)
- Abid Bashir
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tanveer Ahmad
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sadaqat Farooq
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Waseem I Lone
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Natural Products Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Malik M Manzoor
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, India
| | - Yedukondalu Nalli
- Natural Products Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Phalisteen Sultan
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, India
| | - Asha Chaubey
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Asif Ali
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Natural Products Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Syed Riyaz-Ul-Hassan
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
111
|
Orive Bañuelos A, Santamaría Carro A, Feijóo Lera R, Sacristán Egüén C, Andollo Victoriano N, Etxebarria Ecenarro J. Candida Colonization on the Surface of Contact Lenses in Long-Term Wearers and Boston Type 1 Keratoprosthesis Patients Presenting as White Mulberry-Shaped Deposits. Eye Contact Lens 2023; 49:188-192. [PMID: 37088944 DOI: 10.1097/icl.0000000000000984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2023] [Indexed: 04/25/2023]
Abstract
PURPOSE To report Candida colonies on the surface of the contact lens in long-term contact lens wearers and patients with Boston type 1 keratoprosthesis (KPro I). METHODS A retrospective study was performed based on analyzing cultures from bandage contact lenses with small whitish mulberry-shaped deposits on their surface. RESULTS Eight samples (from seven patients) were positive for Candida. Seven of the eight were positive for Candida parapsilosis. CONCLUSIONS The whitish deposits on contact lenses are often Candida colonies that colonize the surface but do not cause an active infection in the eye. C. parapsilosis is well-known for colonizing prosthetic devices. We underline the importance of including Candida species in the differential diagnosis of lens deposits, especially in susceptible patients such as keratoprosthesis carriers.
Collapse
Affiliation(s)
- Ana Orive Bañuelos
- Department of Ophthalmology (A.O.B., A.S.C., R.F.L., C.S.E., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain; and Department of Cell Biology and Histology (N.A.V., J.E.E.), School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | | | | | | | | |
Collapse
|
112
|
Sharma A, Solis NV, Huang MY, Lanni F, Filler SG, Mitchell AP. Hgc1 Independence of Biofilm Hyphae in Candida albicans. mBio 2023; 14:e0349822. [PMID: 36779720 PMCID: PMC10128054 DOI: 10.1128/mbio.03498-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/25/2023] [Indexed: 02/14/2023] Open
Abstract
Biofilm and hypha formation are central to virulence of the fungal pathogen Candida albicans. The G1 cyclin gene HGC1 is required for hypha formation under diverse in vitro and in vivo growth conditions. Hgc1 is required for disseminated infection and is a linchpin in the argument that hyphal morphogenesis itself is required for pathogenicity. We report here that HGC1 is dispensable for hypha formation during biofilm formation both in vitro, under strong inducing conditions, and in vivo, in a mouse oropharyngeal candidiasis model. These findings are validated with two or more C. albicans isolates. Systematic screening of overexpressed cyclin genes indicates that CCN1 and CLN3 can compensate partially for Hgc1 function during biofilm growth. This conclusion is also supported by the severity of the hgc1Δ/Δ ccn1Δ/Δ double mutant biofilm defect. Our results suggest that hypha formation in biofilm is accomplished by combined action of multiple cyclins, not solely by Hgc1. IMPORTANCE The HGC1 gene encodes a cyclin that is required for virulence of the fungal pathogen Candida albicans. It is required to produce the elongated hyphal filaments of free-living planktonic cells that are associated with virulence. Here, we show that HGC1 is not required to produce hyphae in the alternative growth form of a biofilm community. We observe Hgc1-independent hyphae in two infection-relevant situations, biofilm growth in vitro and biofilm-like oropharyngeal infection. Our analysis suggests that hypha formation in the biofilm state reflects combined action of multiple cyclins.
Collapse
Affiliation(s)
- Anupam Sharma
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Norma V. Solis
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Manning Y. Huang
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Scott G. Filler
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
113
|
Kulshrestha A, Gupta P. Secreted aspartyl proteases family: a perspective review on the regulation of fungal pathogenesis. Future Microbiol 2023; 18:295-309. [PMID: 37097060 DOI: 10.2217/fmb-2022-0143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Secreted aspartyl proteases (SAPs) are important enzymes for fungal pathogenicity, playing a significant role in infection and survival. This article provides insight into how SAPs facilitate the transformation of yeast cells into hyphae and engage in biofilm formation, invasion and degradation of host cells and proteins. SAPs and their isoenzymes are prevalent during fungal infections, making them a potential target for antifungal and antibiofilm therapies. By targeting SAPs, critical stages of fungal pathogenesis such as adhesion, hyphal development, biofilm formation, host invasion and immune evasion can potentially be disrupted. Developing therapies that target SAPs could provide an effective treatment option for a wide range of fungal infections.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, India
| |
Collapse
|
114
|
Fernández-Manteca MG, Ocampo-Sosa AA, Ruiz de Alegría-Puig C, Pía Roiz M, Rodríguez-Grande J, Madrazo F, Calvo J, Rodríguez-Cobo L, López-Higuera JM, Fariñas MC, Cobo A. Automatic classification of Candida species using Raman spectroscopy and machine learning. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 290:122270. [PMID: 36580749 DOI: 10.1016/j.saa.2022.122270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/29/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
One of the problems that most affect hospitals is infections by pathogenic microorganisms. Rapid identification and adequate, timely treatment can avoid fatal consequences and the development of antibiotic resistance, so it is crucial to use fast, reliable, and not too laborious techniques to obtain quick results. Raman spectroscopy has proven to be a powerful tool for molecular analysis, meeting these requirements better than traditional techniques. In this work, we have used Raman spectroscopy combined with machine learning algorithms to explore the automatic identification of eleven species of the genus Candida, the most common cause of fungal infections worldwide. The Raman spectra were obtained from more than 220 different measurements of dried drops from pure cultures of each Candida species using a Raman Confocal Microscope with a 532 nm laser excitation source. After developing a spectral preprocessing methodology, a study of the quality and variability of the measured spectra at the isolate and species level, and the spectral features contributing to inter-class variations, showed the potential to discriminate between those pathogenic yeasts. Several machine learning and deep learning algorithms were trained using hyperparameter optimization techniques to find the best possible classifier for this spectral data, in terms of accuracy and lowest possible overfitting. We found that a one-dimensional Convolutional Neural Network (1-D CNN) could achieve above 80 % overall accuracy for the eleven classes spectral dataset, with good generalization capabilities.
Collapse
Affiliation(s)
| | - Alain A Ocampo-Sosa
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Carlos Ruiz de Alegría-Puig
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - María Pía Roiz
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Jorge Rodríguez-Grande
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Fidel Madrazo
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Jorge Calvo
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Rodríguez-Cobo
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Photonics Engineering Group, Universidad de Cantabria, Santander, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - José Miguel López-Higuera
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Photonics Engineering Group, Universidad de Cantabria, Santander, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María Carmen Fariñas
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Enfermedades Infecciosas, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Departamento de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Spain
| | - Adolfo Cobo
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain; Photonics Engineering Group, Universidad de Cantabria, Santander, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
115
|
Prasad P, Tippana M. Morphogenic plasticity: the pathogenic attribute of Candida albicans. Curr Genet 2023; 69:77-89. [PMID: 36947241 DOI: 10.1007/s00294-023-01263-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 03/23/2023]
Abstract
Candida albicans is a commensal organism of the human gastrointestinal tract and a prevalent opportunistic pathogen. It exhibits different morphogenic forms to survive in different host niches with distinct environmental conditions (pH, temperature, oxidative stress, nutrients, serum, chemicals, radiation, etc.) and genetic factors (transcription factors and genes). The different morphogenic forms of C. albicans are yeast, hyphal, pseudohyphal, white, opaque, and transient gray cells, planktonic and biofilm forms of cells. These forms differ in the parameters like cellular phenotype, colony morphology, adhesion to solid surfaces, gene expression profile, and the virulent traits. Each form is functionally distinct and responds discretely to the host immune system and antifungal drugs. Hence, morphogenic plasticity is the key to virulence. In this review, we address the characteristics, the pathogenic potential of the different morphogenic forms and the conditions required for morphogenic transitions.
Collapse
Affiliation(s)
- Priya Prasad
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, India.
| | - Meena Tippana
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, India
| |
Collapse
|
116
|
Characterization of Virulence Factors in Candida Species Causing Candidemia in a Tertiary Care Hospital in Bangkok, Thailand. J Fungi (Basel) 2023; 9:jof9030353. [PMID: 36983521 PMCID: PMC10059995 DOI: 10.3390/jof9030353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/26/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
Candidemia is often associated with high mortality, and Candida albicans, Candida tropicalis, Candida glabrata, and Candida parapsilosis are common causes of this disease. The pathogenicity characteristics of specific Candida spp. that cause candidemia in Thailand are poorly understood. This study aimed to characterize the virulence factors of Candida spp. Thirty-eight isolates of different Candida species from blood cultures were evaluated for their virulence properties, including exoenzyme and biofilm production, cell surface hydrophobicity, tissue invasion, epithelial cell damage, morphogenesis, and phagocytosis resistance; the identity and frequency of mutations in ERG11 contributing to azole-resistance were also determined. C. albicans had the highest epithelial cell invasion rate and phospholipase activity, with true hyphae formation, whereas C. tropicalis produced the most biofilm, hydrophobicity, protease activity, and host cell damage and true hyphae formation. ERG11 mutations Y132F and S154F were observed in all azole-resistant C. tropicalis. C. glabrata had the most hemolytic activity while cell invasion was low with no morphologic transition. C. glabrata was more easily phagocytosed than other species. C. parapsilosis generated pseudohyphae but not hyphae and did not exhibit any trends in exoenzyme production. This knowledge will be crucial for understanding the pathogenicity of Candida spp. and will help to explore antivirulence-based treatment.
Collapse
|
117
|
La Bella AA, Andersen MJ, Gervais NC, Molina JJ, Molesan A, Stuckey PV, Wensing L, Nobile CJ, Shapiro RS, Santiago-Tirado FH, Flores-Mireles AL. The catheterized bladder environment promotes Efg1- and Als1-dependent Candida albicans infection. SCIENCE ADVANCES 2023; 9:eade7689. [PMID: 36867691 PMCID: PMC9984171 DOI: 10.1126/sciadv.ade7689] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Catheter-associated urinary tract infections (CAUTIs) account for 40% of hospital-acquired infections (HAIs). As 20 to 50% of hospitalized patients receive catheters, CAUTIs are one of the most common HAIs, resulting in increased morbidity, mortality, and health care costs. Candida albicans is the second most common CAUTI uropathogen, yet relative to its bacterial counterparts, little is known about how fungal CAUTIs are established. Here, we show that the catheterized bladder environment induces Efg1- and fibrinogen (Fg)-dependent biofilm formation that results in CAUTI. In addition, we identify the adhesin Als1 as the critical fungal factor for C. albicans Fg-urine biofilm formation. Furthermore, we show that in the catheterized bladder, a dynamic and open system, both filamentation and attachment are required, but each by themselves are not sufficient for infection. Our study unveils the mechanisms required for fungal CAUTI establishment, which may aid in the development of future therapies to prevent these infections.
Collapse
Affiliation(s)
- Alyssa Ann La Bella
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | | | - Nicholas C. Gervais
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | - Alex Molesan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Peter V. Stuckey
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Lauren Wensing
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, USA
- Health Sciences Research Institute, University of California, Merced, Merced, CA, USA
| | - Rebecca S. Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | | |
Collapse
|
118
|
Singha R, Aggarwal R, Sanyal K. Negative regulation of biofilm development by the CUG-Ser1 clade-specific histone H3 variant is dependent on the canonical histone chaperone CAF-1 complex in Candida albicans. Mol Microbiol 2023; 119:574-585. [PMID: 36855815 DOI: 10.1111/mmi.15050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/02/2023]
Abstract
The CUG-Ser1 clade-specific histone H3 variant (H3VCTG ) has been reported to be a negative regulator of planktonic to biofilm growth transition in Candida albicans. The preferential binding of H3VCTG at the biofilm gene promoters makes chromatin repressive for the biofilm mode of growth. The two evolutionarily conserved chaperone complexes involved in incorporating histone H3 are CAF-1 and HIRA. In this study, we sought to identify the chaperone complex(es) involved in loading H3VCTG . We demonstrate that C. albicans cells lacking either Cac1 or Cac2 subunit of the CAF-1 chaperone complex, exhibit a hyper-filamentation phenotype on solid surfaces and form more robust biofilms than wild-type cells, thereby mimicking the phenotype of the H3VCTG null mutant. None of the subunits of the HIRA chaperone complex shows any significant difference in biofilm growth as compared to the wild type. The occupancy of H3VCTG is found to be significantly reduced at the promoters of biofilm genes in the absence of CAF-1 subunits. Hence, we provide evidence that CAF-1, a chaperone known to load canonical histone H3 in mammalian cells, is involved in chaperoning of variant histone H3VCTG at the biofilm gene promoters in C. albicans. Our findings also illustrate the acquisition of an unconventional role of the CAF-1 chaperone complex in morphogenesis in C. albicans.
Collapse
Affiliation(s)
- Rima Singha
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Rashi Aggarwal
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Kaustuv Sanyal
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
119
|
Bose S, Singh DV, Adhya TK, Acharya N. Escherichia coli, but Not Staphylococcus aureus, Functions as a Chelating Agent That Exhibits Antifungal Activity against the Pathogenic Yeast Candida albicans. J Fungi (Basel) 2023; 9:jof9030286. [PMID: 36983454 PMCID: PMC10057578 DOI: 10.3390/jof9030286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 02/24/2023] Open
Abstract
Humans are colonized by diverse populations of microbes. Infections by Candida albicans, an opportunistic fungal pathogen, are a result of imbalances in the gut microbial ecosystem and are due to the suppressed immunity of the host. Here, we explored the potential effects of the polymicrobial interactions of C. albicans with Staphylococcus aureus, a Gram-positive bacterium, and Escherichia coli, a Gram-negative bacterium, in dual and triple in vitro culture systems on their respective growth, morphology, and biofilms. We found that S. aureus promoted the fungal growth and hyphal transition of C. albicans through cell-to-cell contacts; contrarily, both the cell and cell-free culture filtrate of E. coli inhibited fungal growth. A yet to be identified secretory metabolite of E. coli functionally mimicked EDTA and EGTA to exhibit antifungal activity. These findings suggested that E. coli, but not S. aureus, functions as a chelating agent and that E. coli plays a dominant role in regulating excessive growth and, potentially, the commensalism of C. albicans. Using animal models of systemic candidiasis, we found that the E. coli cell-free filtrate suppressed the virulence of C. albicans. In general, this study unraveled a significant antimicrobial activity and a potential role in the nutritional immunity of E. coli, and further determining the underlying processes behind the E. coli–C. albicans interaction could provide critical information in understanding the pathogenicity of C. albicans.
Collapse
Affiliation(s)
- Swagata Bose
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar 751023, India
- KIIT School of Biotechnology, Bhubaneswar 751021, India
| | - Durg Vijai Singh
- Department of Biotechnology, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya 824236, India
| | | | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar 751023, India
- Correspondence: ; Tel.: +91-674-230-4278; Fax: +91-674-230-0728
| |
Collapse
|
120
|
Amorim CF, Iglesias BA, Pinheiro TR, Lacerda LE, Sokolonski AR, Pedreira BO, Moreira KS, Burgo TAL, Meyer R, Azevedo V, Portela RW. Photodynamic inactivation of different Candida species and inhibition of biofilm formation induced by water-soluble porphyrins. Photodiagnosis Photodyn Ther 2023; 42:103343. [PMID: 36806829 DOI: 10.1016/j.pdpdt.2023.103343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND Candida spp. is the main fungal genus related to infections in humans, and its treatment has become a challenge due to the production of biofilm and its resistance/multi-resistance profile to conventional antifungals. Antimicrobial photodynamic therapy stands out as a treatment characterized by a broad spectrum of antimicrobial action, being able to induce oxidative stress in pathogens, and porphyrins are photosensitizers with high selectivity to pathogens. Thus, this work aimed to analyze the photoinactivation of different species of Candida by two cationic (4-H2TMeP+ and 3-H2TMeP+) and one anionic (4-H2TPSP‒) porphyrins. MATERIALS AND METHODS Microdilution assays were performed to determine the MIC100, with subsequent determination of MFC100. Determination of oxidative species was done through the use of scavengers, while biofilm morphological features were investigated using the atomic force microscopy. RESULTS Cationic porphyrins were significantly efficient in inactivating Candida albicans and non-albicans species with 100% growth inhibition and fungicidal activity (MFC100/MIC100 ≤ 4.0). The cationic porphyrins were also able to interfere in Candida spp biofilm formation. The photo-oxidative mechanism activated by 3-H2TMeP+ in Candida spp. is concurrent with the production of singlet oxygen and oxygen radical species. In the AFM analysis, 3-H2TMeP+ was able to reduce yeast adhesion to the surface. CONCLUSIONS Cationic porphyrins can photo-inactivate different species of Candida in both planktonic and biofilm-associated forms, and reduce the adhesion of these fungi to the surface.
Collapse
Affiliation(s)
- Carolina Ferreira Amorim
- Laboratory of Immunology and Molecular Biology, Health Sciences Institute, Universidade Federal da Bahia, Salvador, Bahia State 40110-100, Brazil
| | - Bernardo Almeida Iglesias
- Bioinorganic and Porphyrinoid Materials Laboratory, Department of Chemistry, Universidade Federal de Santa Maria, Rio Grande do Sul State, Santa Maria 97105-900, Brazil.
| | - Ticiane Rosa Pinheiro
- Bioinorganic and Porphyrinoid Materials Laboratory, Department of Chemistry, Universidade Federal de Santa Maria, Rio Grande do Sul State, Santa Maria 97105-900, Brazil
| | - Luiz Eduardo Lacerda
- Laboratory of Immunology and Molecular Biology, Health Sciences Institute, Universidade Federal da Bahia, Salvador, Bahia State 40110-100, Brazil
| | - Ana Rita Sokolonski
- Laboratory of Oral Biochemistry, Health Sciences Institute, Universidade Federal da Bahia, Salvador, Bahia State 40110-100, Brazil
| | - Beatriz Oliveira Pedreira
- Laboratory of Immunology and Molecular Biology, Health Sciences Institute, Universidade Federal da Bahia, Salvador, Bahia State 40110-100, Brazil
| | - Kelly Schneider Moreira
- Coulomb Electrostatic and Mechanochemistry Laboratory, Universidade Federal de Santa Maria, Rio Grande do Sul State, Santa Maria 97105-900, Brazil
| | - Thiago Augusto Lima Burgo
- Coulomb Electrostatic and Mechanochemistry Laboratory, Universidade Federal de Santa Maria, Rio Grande do Sul State, Santa Maria 97105-900, Brazil
| | - Roberto Meyer
- Laboratory of Immunology and Molecular Biology, Health Sciences Institute, Universidade Federal da Bahia, Salvador, Bahia State 40110-100, Brazil
| | - Vasco Azevedo
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais State 31270-901, Brazil
| | - Ricardo Wagner Portela
- Laboratory of Immunology and Molecular Biology, Health Sciences Institute, Universidade Federal da Bahia, Salvador, Bahia State 40110-100, Brazil.
| |
Collapse
|
121
|
Kim SH, Mun SJ, Kang JS, Moon C, Kim HT, Lee HY. Multifaceted Evaluation of Antibiotic Therapy as a Factor Associated with Candidemia in Non-Neutropenic Patients. J Fungi (Basel) 2023; 9:jof9020270. [PMID: 36836385 PMCID: PMC9960229 DOI: 10.3390/jof9020270] [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: 12/14/2022] [Revised: 02/04/2023] [Accepted: 02/12/2023] [Indexed: 02/22/2023] Open
Abstract
We aimed to evaluate various aspects of antibiotic therapy as factors associated with candidemia in non-neutropenic patients. A retrospective, matched, case-control study was conducted in two teaching hospitals. Patients with candidemia (cases) were compared to patients without candidemia (controls), matched by age, intensive care unit admission, duration of hospitalization, and type of surgery. Logistic regression analyses were performed to identify factors associated with candidemia. A total of 246 patients were included in the study. Of 123 candidemia patients, 36% had catheter-related bloodstream infections (CRBSIs). Independent factors in the whole population included immunosuppression (adjusted odds ratio [aOR] = 2.195; p = 0.036), total parenteral nutrition (aOR = 3.642; p < 0.001), and anti-methicillin-resistant S. aureus (MRSA) therapy for ≥11 days (aOR = 5.151; p = 0.004). The antibiotic factor in the non-CRBSI population was anti-pseudomonal beta-lactam treatment duration of ≥3 days (aOR = 5.260; p = 0.008). The antibiotic factors in the CRBSI population included anti-MRSA therapy for ≥11 days (aOR = 10.031; p = 0.019). Antimicrobial stewardship that reduces exposure to these antibacterial spectra could help prevent the development of candidemia.
Collapse
Affiliation(s)
- Si-Ho Kim
- Division of Infectious Diseases, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Seok Jun Mun
- Division of Infectious Diseases, Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea
- Correspondence: ; Tel.: +82-51-890-6986; Fax: +82-51-890-6341
| | - Jin Suk Kang
- Division of Infectious Diseases, Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea
| | - Chisook Moon
- Division of Infectious Diseases, Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea
| | - Hyoung-Tae Kim
- Department of Laboratory Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Ho Young Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, Busan 47392, Republic of Korea
| |
Collapse
|
122
|
Tang N, Yuan S, Luo Y, Wang AJ, Sun K, Liu NN, Tao K. Nanoparticle-Based Photodynamic Inhibition of Candida albicans Biofilms with Interfering Quorum Sensing. ACS OMEGA 2023; 8:4357-4368. [PMID: 36743058 PMCID: PMC9893753 DOI: 10.1021/acsomega.2c07740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 06/18/2023]
Abstract
Biofilm formation is a critical event in the pathogenesis and virulence of fungal infections caused by Candida albicans, giving rise to about a 1000-fold increase in the resistance to antifungal agents. Although photodynamic treatment (PDT) has been excellently implicated in bacterial infections, studies on its potential against fungal infection through the clearance of fungal biofilm formation remain at its infancy stage. Here, we have designed photodynamic nanoparticles with different sizes, modifications, and the ability of generating reactive oxygen species (ROS) to examine their effects on inhibiting biofilm formation and destructing mature biofilms of C. albicans. We found that the nanoparticles modified with oligo-chitosan exhibited a better binding efficiency for planktonic cells, leading to stronger inhibitory efficacy of the filamentation and the early-stage biofilm formation. However, for mature biofilms, the nanoparticles with the smallest size (∼15 nm) showed the fastest penetration speed and a pronounced destructing effect albeit conferring the lowest ROS-producing capability. The inhibitory effect of photodynamic nanoparticles was dependent on the disruption of fungal quorum sensing (QS) by the upregulation of QS molecules, farnesol and tyrosol, mediated through the upregulation of ARO 8 and DPP 3 expression. Our findings provide a powerful strategy of nanoparticulate PDT to combat fungal infections through the inhibition of both hyphal and biofilm formation by disrupting QS.
Collapse
Affiliation(s)
- Na Tang
- State
Key Lab of Metal Matrix Composites, School of Materials Science and
Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Shenghao Yuan
- State
Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell
Omics, School of Public Health, Shanghai
Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuxuan Luo
- State
Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell
Omics, School of Public Health, Shanghai
Jiao Tong University School of Medicine, Shanghai 200025, China
| | - An-Jun Wang
- State
Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell
Omics, School of Public Health, Shanghai
Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kang Sun
- State
Key Lab of Metal Matrix Composites, School of Materials Science and
Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Ning-Ning Liu
- State
Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell
Omics, School of Public Health, Shanghai
Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ke Tao
- State
Key Lab of Metal Matrix Composites, School of Materials Science and
Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| |
Collapse
|
123
|
Cravener MV, Do E, May G, Zarnowski R, Andes DR, McManus CJ, Mitchell AP. Reinforcement amid genetic diversity in the Candida albicans biofilm regulatory network. PLoS Pathog 2023; 19:e1011109. [PMID: 36696432 PMCID: PMC9901766 DOI: 10.1371/journal.ppat.1011109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/06/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Biofilms of the fungal pathogen Candida albicans include abundant long filaments called hyphae. These cells express hypha-associated genes, which specify diverse virulence functions including surface adhesins that ensure biofilm integrity. Biofilm formation, virulence, and hypha-associated gene expression all depend upon the transcription factor Efg1. This transcription factor has been characterized extensively in the C. albicans type strain SC5314 and derivatives, but only recently has its function been explored in other clinical isolates. Here we define a principal set of Efg1-responsive genes whose expression is significantly altered by an efg1Δ/Δ mutation across 17 clinical isolates. This principal gene set includes 68 direct Efg1 targets, whose 5' regions are bound by Efg1 in five clinical isolates, and 42 indirect Efg1 targets, whose 5' regions are not detectably bound by Efg1. Three direct Efg1 target genes encode transcription factors-BRG1, UME6, and WOR3 -whose increased expression in an efg1Δ/Δ mutant restores expression of multiple indirect and direct principal targets, as well as biofilm formation ability. Although BRG1 and UME6 are well known positive regulators of hypha-associated genes and biofilm formation, WOR3 is best known as an antagonist of Efg1 in the sexual mating pathway. We confirm the positive role of WOR3 in biofilm formation with the finding that a wor3Δ/Δ mutation impairs biofilm formation in vitro and in an in vivo biofilm model. Positive control of Efg1 direct target genes by other Efg1 direct target genes-BRG1, UME6, and WOR3 -may buffer principal Efg1-responsive gene expression against the impact of genetic variation in the C. albicans species.
Collapse
Affiliation(s)
- Max V. Cravener
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Eunsoo Do
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Gemma May
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Robert Zarnowski
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - David R. Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
124
|
Guevara-Lora I, Bras G, Juszczak M, Karkowska-Kuleta J, Gorecki A, Manrique-Moreno M, Dymek J, Pyza E, Kozik A, Rapala-Kozik M. Cecropin D-derived synthetic peptides in the fight against Candida albicans cell filamentation and biofilm formation. Front Microbiol 2023; 13:1045984. [PMID: 36713201 PMCID: PMC9880178 DOI: 10.3389/fmicb.2022.1045984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
The recent progressive increase in the incidence of invasive fungal infections, especially in immunocompromised patients, makes the search for new therapies crucial in the face of the growing drug resistance of prevalent nosocomial yeast strains. The latest research focuses on the active compounds of natural origin, inhibiting fungal growth, and preventing the formation of fungal biofilms. Antimicrobial peptides are currently the subject of numerous studies concerning effective antifungal therapy. In the present study, the antifungal properties of two synthetic peptides (ΔM3, ΔM4) derived from an insect antimicrobial peptide - cecropin D - were investigated. The fungicidal activity of both compounds was demonstrated against the yeast forms of Candida albicans, Candida tropicalis, and Candida parapsilosis, reaching a MFC99.9 in the micromolar range, while Candida glabrata showed greater resistance to these peptides. The scanning electron microscopy revealed a destabilization of the yeast cell walls upon treatment with both peptides; however, their effectiveness was strongly modified by the presence of salt or plasma in the yeast environment. The transition of C. albicans cells from yeast to filamentous form, as well as the formation of biofilms, was effectively reduced by ΔM4. Mature biofilm viability was inhibited by a higher concentration of this peptide and was accompanied by increased ROS production, activation of the GPX3 and SOD5 genes, and finally, increased membrane permeability. Furthermore, both peptides showed a synergistic effect with caspofungin in inhibiting the metabolic activity of C. albicans cells, and an additive effect was also observed for the mixtures of peptides with amphotericin B. The results indicate the possible potential of the tested peptides in the prevention and treatment of candidiasis.
Collapse
Affiliation(s)
- Ibeth Guevara-Lora
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Grazyna Bras
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Andrzej Gorecki
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Marcela Manrique-Moreno
- Chemistry Institute, Faculty of Exact and Natural Sciences, University of Antioquia, Medellin, Colombia
| | - Jakub Dymek
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland,*Correspondence: Maria Rapala-Kozik,
| |
Collapse
|
125
|
Zainudin RA, Sabri S, Salleh AB, Abu A, Khairuddin RFR, Oslan SN. In silico identification of prospective virulence factors associated with candidiasis in Meyerozyma guilliermondii strain SO from genome dataset. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2023. [DOI: 10.1186/s43042-023-00384-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Abstract
Background
Meyerozyma guilliermondii is a prospective yeast that has extensively contributed to the biotechnology sector. In 2015, M. guilliermondii strain SO which was isolated from spoiled orange has successfully been developed as an inducer-free expression system and attained a significant impact in producing industrially important recombinant proteins. The species possesses high similarity to Candida albicans which may cause candidiasis. The industrial-benefiting M. guilliermondii strain SO has been underexplored for its virulence status. Thus, this study aimed to document the potential virulence factors through the comprehensive in silico analysis of M. guilliermondii strain SO genome. This analysis demonstrated the molecular characterization which could distinguish the pathogenicity status of M. guilliermondii.
Results
The genome data were generated from Illumina HiSeq 4000 sequencing platform and assembled into 51 scaffolds successfully accumulating a genome size of 10.63 Mbp. These enclosed 5,335 CDS genes and 5,349 protein sequences with 43.72% GC content. About 99.29% of them were annotated to public databases. Komagataella phaffii, Saccharomyces cerevisiae and the reference strain of M. guilliermondii (ATCC 6260) were used as the controls. They were compared with our in-house strain SO to identify the consensus domain or subdomain which could putatively be considered as virulence factors. Candida albicans was used as the pathogenic model. Hence, hidden Markov model against strain SO proteome had identified secreted aspartic proteases (SAP), phospholipase C (PLC) and phospholipase D (PLD) with an E-value of 2.4e−107, 9.5e−200 and 0.0e+00, respectively, in resemblance of C. albicans. The topology of the phylogenetic analysis indicated that these virulence factors in M. guilliermondii strain SO and C. albicans branched from the same node and clustered together as a clade, signifying their molecular relatedness and congeneric among these species, subsequently proposing the virulence status of M. guilliermondii.
Conclusion
The SAP, PLC and PLD genes’ features that were significant in expressing determinants of pathogenicity were successfully identified in M. guilliermondii strain SO genome dataset, thus concluding the virulency of this species. On account of this finding, the strategy of gene knockout through CRISPR-Cas9 or homologous recombination strategies is needed to engineer the feasible novel expression host system. Over and above, the genetically modified strain of M. guilliermondii allegedly may eradicate the risk of candidiasis infection.
Collapse
|
126
|
Branco J, Miranda IM, Rodrigues AG. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. J Fungi (Basel) 2023; 9:jof9010080. [PMID: 36675901 PMCID: PMC9862255 DOI: 10.3390/jof9010080] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Candida parapsilosis is the second most common Candida species isolated in Asia, Southern Europe, and Latin America and is often involved in invasive infections that seriously impact human health. This pathogen is part of the psilosis complex, which also includes Candida orthopsilosis and Candida metapsilosis. C. parapsilosis infections are particularly prevalent among neonates with low birth weights, individuals who are immunocompromised, and patients who require prolonged use of a central venous catheter or other indwelling devices, whose surfaces C. parapsilosis exhibits an enhanced capacity to adhere to and form biofilms. Despite this well-acknowledged prevalence, the biology of C. parapsilosis has not been as extensively explored as that of Candida albicans. In this paper, we describe the molecular mechanistic pathways of virulence in C. parapsilosis and show how they differ from those of C. albicans. We also describe the mode of action of antifungal drugs used for the treatment of Candida infections, namely, polyenes, echinocandins, and azoles, as well as the resistance mechanisms developed by C. parapsilosis to overcome them. Finally, we stress the importance of the ongoing search for species-specific features that may aid the development of effective control strategies and thus reduce the burden on patients and healthcare costs.
Collapse
Affiliation(s)
- Joana Branco
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Correspondence: ; Tel./Fax: +351-225513662
| | - Isabel M. Miranda
- Cardiovascular Research & Development Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Acácio G. Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
127
|
Triastuti A, Vansteelandt M, Barakat F, Amasifuen C, Jargeat P, Haddad M. Untargeted metabolomics to evaluate antifungal mechanism: a study of Cophinforma mamane and Candida albicans interaction. NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:1. [PMID: 36595109 PMCID: PMC9810774 DOI: 10.1007/s13659-022-00365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Microbial interactions between filamentous fungi and yeast are still not fully understood. To evaluate a potential antifungal activity of a filamentous fungus while highlighting metabolomic changes, co-cultures between an endophytic strain of Cophinforma mamane (CM) and Candida albicans (CA) were performed. The liquid cultures were incubated under static conditions and metabolite alterations during the course were investigated by ultra-performance liquid chromatography-tandem mass spectrophotometry (UPLC-MS/MS). Results were analyzed using MS-DIAL, MS-FINDER, METLIN, Xcalibur, SciFinder, and MetaboAnalyst metabolomics platforms. The metabolites associated with catabolic processes, including the metabolism of branched-chain amino acids, carnitine, and phospholipids were upregulated both in the mono and co-cultures, indicating fungal adaptability to environmental stress. Several metabolites, including C20 sphinganine 1-phosphate, myo-inositol, farnesol, gamma-undecalactone, folinic acid, palmitoleic acid, and MG (12:/0:0/0:0) were not produced by CA during co-culture with CM, demonstrating the antifungal mechanism of CM. Our results highlight the crucial roles of metabolomics studies to provide essential information regarding the antifungal mechanism of C. mamane against C. albicans, especially when the lost/undetected metabolites are involved in fungal survival and pathogenicity.
Collapse
Affiliation(s)
- Asih Triastuti
- UMR 152 Pharma Dev, IRD, UPS, Université de Toulouse, 31400, Toulouse, France.
- Department of Pharmacy, Universitas Islam Indonesia, Yogyakarta, 55584, Indonesia.
| | | | - Fatima Barakat
- UMR 152 Pharma Dev, IRD, UPS, Université de Toulouse, 31400, Toulouse, France
| | - Carlos Amasifuen
- Dirección de Recursos Genéticos y Biotecnología, Instituto Nacional de Innovación Agraria, Avenida La Molina 1981, La Molina, Lima, 15024, Peru
| | - Patricia Jargeat
- Laboratoire Evolution et Diversité Biologique UMR 5174, CNRS, IRD, UPS, Université de Toulouse, 31062, Toulouse, France
| | - Mohamed Haddad
- UMR 152 Pharma Dev, IRD, UPS, Université de Toulouse, 31400, Toulouse, France.
| |
Collapse
|
128
|
Powell LC, Adams JYM, Quoraishi S, Py C, Oger A, Gazze SA, Francis LW, von Ruhland C, Owens D, Rye PD, Hill KE, Pritchard MF, Thomas DW. Alginate oligosaccharides enhance the antifungal activity of nystatin against candidal biofilms. Front Cell Infect Microbiol 2023; 13:1122340. [PMID: 36798083 PMCID: PMC9927220 DOI: 10.3389/fcimb.2023.1122340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Background The increasing prevalence of invasive fungal infections in immuno-compromised patients is a considerable cause of morbidity and mortality. With the rapid emergence of antifungal resistance and an inadequate pipeline of new therapies, novel treatment strategies are now urgently required. Methods The antifungal activity of the alginate oligosaccharide OligoG in conjunction with nystatin was tested against a range of Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis and C. dubliniensis), in both planktonic and biofilm assays, to determine its potential clinical utility to enhance the treatment of candidal infections. The effect of OligoG (0-6%) ± nystatin on Candida spp. was examined in minimum inhibitory concentration (MIC) and growth curve assays. Antifungal effects of OligoG and nystatin treatment on biofilm formation and disruption were characterized using confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM) and ATP cellular viability assays. Effects on the cell membrane were determined using permeability assays and transmission electron microscopy (TEM). Results MIC and growth curve assays demonstrated the synergistic effects of OligoG (0-6%) with nystatin, resulting in an up to 32-fold reduction in MIC, and a significant reduction in the growth of C. parapsilosis and C. auris (minimum significant difference = 0.2 and 0.12 respectively). CLSM and SEM imaging demonstrated that the combination treatment of OligoG (4%) with nystatin (1 µg/ml) resulted in significant inhibition of candidal biofilm formation on glass and clinical grade silicone surfaces (p < 0.001), with increased cell death (p < 0.0001). The ATP biofilm disruption assay demonstrated a significant reduction in cell viability with OligoG (4%) alone and the combined OligoG/nystatin (MIC value) treatment (p < 0.04) for all Candida strains tested. TEM studies revealed the combined OligoG/nystatin treatment induced structural reorganization of the Candida cell membrane, with increased permeability when compared to the untreated control (p < 0.001). Conclusions Antimicrobial synergy between OligoG and nystatin against Candida spp. highlights the potential utility of this combination therapy in the prevention and topical treatment of candidal biofilm infections, to overcome the inherent tolerance of biofilm structures to antifungal agents.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- Microbiology and Infectious Disease group, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Lydia C. Powell,
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Sadik Quoraishi
- Otolaryngology Department, New Cross Hospital, Wolverhampton, United Kingdom
| | - Charlène Py
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Anaϊs Oger
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Salvatore A. Gazze
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Christopher von Ruhland
- Central Biotechnology Services, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David Owens
- Head and Neck Directorate, University Hospital of Wales, Cardiff, United Kingdom
| | | | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| |
Collapse
|
129
|
Gershater E, Liu Y, Xue B, Shin MK, Koo H, Zheng Z, Li C. Characterizing the microbiota of cleft lip and palate patients: a comprehensive review. Front Cell Infect Microbiol 2023; 13:1159455. [PMID: 37143743 PMCID: PMC10152472 DOI: 10.3389/fcimb.2023.1159455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Orofacial cleft disorders, including cleft lip and/or palate (CL/P), are one of the most frequently-occurring congenital disorders worldwide. The health issues of patients with CL/P encompass far more than just their anatomic anomaly, as patients with CL/P are prone to having a high incidence of infectious diseases. While it has been previously established that the oral microbiome of patients with CL/P differs from that of unaffected patients, the exact nature of this variance, including the relevant bacterial species, has not been fully elucidated; likewise, examination of anatomic locations besides the cleft site has been neglected. Here, we intended to provide a comprehensive review to highlight the significant microbiota differences between CL/P patients and healthy subjects in various anatomic locations, including the teeth inside and adjacent to the cleft, oral cavity, nasal cavity, pharynx, and ear, as well as bodily fluids, secretions, and excretions. A number of bacterial and fungal species that have been proven to be pathogenic were found to be prevalently and/or specifically detected in CL/P patients, which can benefit the development of CL/P-specific microbiota management strategies.
Collapse
Affiliation(s)
| | - Yuan Liu
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Binglan Xue
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Min Kyung Shin
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hyun Koo
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhong Zheng
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Zhong Zheng, ; Chenshuang Li,
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Zhong Zheng, ; Chenshuang Li,
| |
Collapse
|
130
|
Chudzik-Rząd B, Zalewski D, Kasela M, Sawicki R, Szymańska J, Bogucka-Kocka A, Malm A. The Landscape of Gene Expression during Hyperfilamentous Biofilm Development in Oral Candida albicans Isolated from a Lung Cancer Patient. Int J Mol Sci 2022; 24:ijms24010368. [PMID: 36613809 PMCID: PMC9820384 DOI: 10.3390/ijms24010368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The filamentation ability of Candida albicans represents one of the main virulence factors allowing for host tissue penetration and biofilm formation. The aim of this paper was to study the genetic background of the hyperfilamentous biofilm development in vitro in C. albicans isolated from the oral cavity of a lung cancer patient. Analyzed C. albicans isolates (CA1, CA2, CA3) were chosen based on their different structures of mature biofilm. The CA3 isolate possessing hyperfilamentation properties and forming high biofilm was compared with CA1 and CA2 isolates exhibiting low or average biofilm-forming ability, respectively. The detailed biofilm organization was studied with the use of confocal scanning laser microscopy. The whole transcriptome analysis was conducted during three stages of biofilm development (24 h, 48 h, 72 h). In contrast to CA1 and/or CA2 isolate, the CA3 isolate was characterized by a significant upregulation of genes encoding for cell wall proteins (HWP1, PGA13, PGA44, ALS3) and candidalysin (ECE1), as well as being involved in iron metabolism (FRE1, ALS3), sulfur metabolism (HAL21), the degradation of aromatic compounds (HQD2), and membrane transport (DIP5, PHO89, TNA1). In contrast, some genes (SCW11, FGR41, RBE1) in the CA3 were downregulated. We also observed the overexpression of a few genes over time-mainly FRE1, ATX1, CSA2 involved in iron metabolism. This is the first insight into the potential function of multiple genes in the hyperfilamentous biofilm formation in C. albicans, primarily isolated from host tissue, which may have an important clinical impact on cancer patients. Moreover, the presented data can lay the foundation for further research on novel pathogen-specific targets for antifungal drugs.
Collapse
Affiliation(s)
- Beata Chudzik-Rząd
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
| | - Daniel Zalewski
- Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Martyna Kasela
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
- Correspondence: (M.K.); (A.M.); Tel.: +48-81448-7100 (M.K. & A.M.)
| | - Rafał Sawicki
- Department of Biochemistry and Biotechnology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
| | - Jolanta Szymańska
- Department of Comprehensive Paediatric and Adult Dentistry, Medical University of Lublin, 6 Chodźki St., 20-093 Lublin, Poland
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland
- Correspondence: (M.K.); (A.M.); Tel.: +48-81448-7100 (M.K. & A.M.)
| |
Collapse
|
131
|
Mochochoko BM, Pohl CH, O’Neill HG. Candida albicans-enteric viral interactions-The prostaglandin E 2 connection and host immune responses. iScience 2022; 26:105870. [PMID: 36647379 PMCID: PMC9839968 DOI: 10.1016/j.isci.2022.105870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The human microbiome comprises trillions of microorganisms residing within different mucosal cavities and across the body surface. The gut microbiota modulates host susceptibility to viral infections in several ways, and microbial interkingdom interactions increase viral infectivity within the gut. Candida albicans, a frequently encountered fungal species in the gut, produces highly structured biofilms and eicosanoids such as prostaglandin E2 (PGE2), which aid in viral protection and replication. These biofilms encompass viruses and provide a shield from antiviral drugs or the immune system. PGE2 is a key modulator of active inflammation with the potential to regulate interferon signaling upon microbial invasion or viral infections. In this review, we raise the perspective of gut interkingdom interactions involving C. albicans and enteric viruses, with a special focus on biofilms, PGE2, and viral replication. Ultimately, we discuss the possible implications of C. albicans-enteric virus associations on host immune responses, particularly the interferon signaling pathway.
Collapse
Affiliation(s)
- Bonang M. Mochochoko
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, 9301, South Africa
| | - Carolina H. Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, 9301, South Africa,Corresponding author
| | - Hester G. O’Neill
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, 9301, South Africa,Corresponding author
| |
Collapse
|
132
|
Loaiza-Oliva M, Arias-Durango L, Martínez-Pabón MC. The Cytotoxic and Inhibitory Effects of Plant Derivatives on Candida albicans Biofilms: A Scoping Review. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010130. [PMID: 36615324 PMCID: PMC9822484 DOI: 10.3390/molecules28010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Candida albicans infections are related to biofilm formation. The increase in antifungal resistance and their adverse effects have led to the search for therapeutic options as plant derivatives. This scoping review aims to identify the current status of in vitro research on the cytotoxicity and inhibitory effects of plant derivatives on C. albicans biofilms. In this study, PRISMA items were followed. After recognition of the inclusion criteria, full texts were read and disagreements were resolved with a third party. A risk of bias assessment was performed, and information was summarized using Microsoft Office Excel. Thirty-nine papers fulfilling the selection criteria were included. The risk of bias analysis identified most of the studies as low risk. Studies evaluated plant derivatives such as extracts, essential oils, terpenes, alkaloids, flavonoids and polyphenols. Some studies evaluated the inhibition of C. albicans biofilm formation, inhibition on preformed biofilms or both. The derivatives at concentrations greater than or equal to those that have an inhibitory effect on C. albicans biofilms, without showing cytotoxicity, include magnoflorin, ellagic acid, myricetin and eucarobustol from Eucalyptus robusta and, as the works in which these derivatives were studied are of good quality, it is desirable to carry out study in other experimental phases, with methodologies that generate comparable information.
Collapse
|
133
|
Jothi R, Sangavi R, Raja V, Kumar P, Pandian SK, Gowrishankar S. Alteration of Cell Membrane Permeability by Cetyltrimethylammonium Chloride Induces Cell Death in Clinically Important Candida Species. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:ijerph20010027. [PMID: 36612353 PMCID: PMC9819714 DOI: 10.3390/ijerph20010027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 05/25/2023]
Abstract
The increased incidence of healthcare-related Candida infection has necessitated the use of effective disinfectants/antiseptics in healthcare settings as a preventive measure to decontaminate the hospital environment and stop the persistent colonization of the offending pathogens. Quanternary ammonium surfactants (QASs), with their promising antimicrobial efficacy, are considered as intriguing and appealing candidates for disinfectants. From this perspective, the present study investigated the antifungal efficacy and action mechanism of the QAS cetyltrimethylammonium chloride (CTAC) against three clinically important Candida species: C. albicans, C. tropicalis, and C. glabrata. CTAC exhibited phenomenal antifungal activity against all tested Candida spp., with minimum inhibitory concentrations (MIC) and minimum fungicidal concentrations (MFC) between 2 and 8 µg/mL. The time−kill kinetics of CTAC (at 2XMIC) demonstrated that an exposure time of 2 h was required to kill 99.9% of the inoculums in all tested strains. An important observation was that CTAC treatment did not influence intracellular reactive oxygen species (ROS), signifying that its phenomenal anticandidal efficacy was not mediated via oxidative stress. In addition, sorbitol supplementation increased CTAC’s MIC values against all tested Candida strains by three times (8−32 μg/mL), indicating that CTAC’s possible antifungal activity involves fungus cell membrane destruction. Interestingly, the increased fluorescence intensity of CTAC-treated cells in both propidium iodide (PI) and DAPI staining assays indicated the impairment of cell plasma membrane and nuclear membrane integrity by CTAC, respectively. Additionally, CTAC at MIC and 2XMIC was sufficient (>80%) to disrupt the mature biofilms of all tested spp., and it inhibited the yeast-to-hyphae transition at sub-MIC in C. albicans. Finally, the non-hemolytic activity of CTAC (upto 32 µg/mL) in human blood cells and HBECs signified its non-toxic nature at the investigated concentrations. Furthermore, thymol and citral, two phytocompounds, together with CTAC, showed synergistic fungicidal effectiveness against C. albicans planktonic cells. Altogether, the data of the present study appreciably broaden our understanding of the antifungal action mechanism of CTAC and support its future translation as a potential disinfectant against Candida-associated healthcare infections.
Collapse
Affiliation(s)
- Ravi Jothi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Ravichellam Sangavi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Veerapandian Raja
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Ponnuchamy Kumar
- Department of Animal Health and Management, Science Campus, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | | | - Shanmugaraj Gowrishankar
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| |
Collapse
|
134
|
In Silico and In Vitro Analysis of Sulforaphane Anti- Candida Activity. Antibiotics (Basel) 2022; 11:antibiotics11121842. [PMID: 36551499 PMCID: PMC9774275 DOI: 10.3390/antibiotics11121842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Oropharyngeal candidiasis/candidosis is a common and recurrent opportunistic fungal infection. Fluconazole (FLZ), one of the most used and effective antifungal agents, has been associated with a rise of resistant Candida species in immunocompromised patients undergoing prophylactic therapy. Sulforaphane (SFN), a compound from cruciferous vegetables, is an antimicrobial with yet controversial activities and mechanisms on fungi. Herein, the in silico and antifungal activities of SFN against C. albicans were investigated. In silico analyzes for the prediction of the biological activities and oral bioavailability of SFN, its possible toxicity and pharmacokinetic parameters, as well as the estimates of its gastrointestinal absorption, permeability to the blood-brain barrier and skin, and similarities to drugs, were performed by using different software. SFN in vitro anti-Candida activities alone and in combination with fluconazole (FLZ) were determined by the broth microdilution method and the checkerboard, biofilm and hyphae formation tests. Amongst the identified probable biological activities of SFN, nine indicated an antimicrobial potential. SFN was predicted to be highly absorbable by the gastrointestinal tract, to present good oral availability, and not to be irritant and/or hepatotoxic. SFN presented antifungal activity against C. albicans and prevented both biofilm and hyphae formation by this microorganism. SFN was additive/synergistic to FLZ. Overall, the data highlights the anti-Candida activity of SFN and its potential to be used as an adjuvant therapy to FLZ in clinical settings.
Collapse
|
135
|
Characterization of Defensin-like Protein 1 for Its Anti-Biofilm and Anti-Virulence Properties for the Development of Novel Antifungal Drug against Candida auris. J Fungi (Basel) 2022; 8:jof8121298. [PMID: 36547631 PMCID: PMC9786216 DOI: 10.3390/jof8121298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Candida auris has emerged as a pan-resistant pathogenic yeast among immunocompromised patients worldwide. As this pathogen is involved in biofilm-associated infections with serious medical manifestations due to the collective expression of pathogenic attributes and factors associated with drug resistance, successful treatment becomes a major concern. In the present study, we investigated the candidicidal activity of a plant defensin peptide named defensin-like protein 1 (D-lp1) against twenty-five clinical strains of C. auris. Furthermore, following the standard protocols, the D-lp1 was analyzed for its anti-biofilm and anti-virulence properties. The impact of these peptides on membrane integrity was also evaluated. For cytotoxicity determination, a hemolytic assay was conducted using horse blood. The minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) values ranged from 0.047-0.78 mg/mL and 0.095-1.56 mg/mL, respectively. D-lp1 at sub-inhibitory concentrations potentially abrogated both biofilm formation and 24-h mature biofilms. Similarly, the peptide severely impacted virulence attributes in the clinical strain of C. auris. For the insight mechanism, D-lp1 displayed a strong impact on the cell membrane integrity of the test pathogen. It is important to note that D-lp1 at sub-inhibitory concentrations displayed minimal hemolytic activity against horse blood cells. Therefore, it is highly useful to correlate the anti-Candida property of D-lp1 along with anti-biofilm and anti-virulent properties against C. auris, with the aim of discovering an alternative strategy for combating serious biofilm-associated infections caused by C. auris.
Collapse
|
136
|
Harikrishnan P, Arayambath B, Jayaraman VK, Ekambaram K, Ahmed EA, Senthilkumar P, Ibrahim HIM, Sundaresan A, Thirugnanasambantham K. Thidiazuron, a phenyl-urea cytokinin, inhibits ergosterol synthesis and attenuates biofilm formation of Candida albicans. World J Microbiol Biotechnol 2022; 38:224. [DOI: 10.1007/s11274-022-03410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
|
137
|
Quintieri L, Mani S, Lentini G, Maisetta G. Editorial: Advances in the discovery of natural molecules and their analogues against microbial infection-related biofilms. Front Microbiol 2022; 13:1092209. [PMID: 36504771 PMCID: PMC9733423 DOI: 10.3389/fmicb.2022.1092209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Laura Quintieri
- Institute of Sciences of Food Production, National Research Council (CNR), Bari, Italy,*Correspondence: Laura Quintieri
| | - Sridhar Mani
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Giovanni Lentini
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy,Giuseppantonio Maisetta
| |
Collapse
|
138
|
The Bovhyaluronidase Azoximer (Longidaza ®) Disrupts Candida albicans and Candida albicans-Bacterial Mixed Biofilms and Increases the Efficacy of Antifungals. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121710. [PMID: 36556912 PMCID: PMC9782602 DOI: 10.3390/medicina58121710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Background and Objectives: Candida albicans causes various diseases ranging from superficial mycoses to life-threatening systemic infections often associated with biofilm formation, including mixed fungal−bacterial consortia. The biofilm matrix protects cells, making Candida extremely resistant to treatment. Here, we show that the bovhyaluronidase azoximer (Longidaza®) in vitro destroys the biofilm formed by either C. albicans alone or mixed with bacteria, this way decreasing the concentrations of antimicrobials required for the pathogen’s eradication. Materials and Methods: Bovhyaluronidase azoximer, Longidaza® was obtained from NPO Petrovax Pharm Ltd., Moscow, Russia as lyophilized powder. The antifungal activity was assessed by microdilution assay and CFUs counting. Antibiofilm activity was evaluated via biofilms staining and scanning electron microscopy. Results: Thus, treatment with Longidaza® reduced the biofilm biomass of nine C. albicans clinical isolates by 30−60%, while mixed biofilms of C. albicans with various bacteria were destroyed by 30−40%. Furthermore, the concentration of fluconazole required to achieve a similar reduction of the residual respiratory activity of detached cell clumps of four C. albicans isolates has been reduced four-fold when combined with Longidaza®. While in the biofilm, two of four isolates became significantly more susceptible to fluconazole in combination with Longidaza®. Conclusion: Taken together, our data indicate that Longidaza® is capable of suppression of tissues and artificial surfaces biofouling by C. albicans biofilms, as well as facilitating drug penetration into the cell clumps, this way decreasing the effective MIC of antifungals.
Collapse
|
139
|
Similarities and Differences among Species Closely Related to Candida albicans: C. tropicalis, C. dubliniensis, and C. auris. Cell Microbiol 2022. [DOI: 10.1155/2022/2599136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Although Candida species are widespread commensals of the microflora of healthy individuals, they are also among the most important human fungal pathogens that under certain conditions can cause diseases (candidiases) of varying severity ranging from mild superficial infections of the mucous membranes to life-threatening systemic infections. So far, the vast majority of research aimed at understanding the molecular basis of pathogenesis has been focused on the most common species—Candida albicans. Meanwhile, other closely related species belonging to the CTG clade, namely, Candida tropicalis and Candida dubliniensis, are becoming more important in clinical practice, as well as a relatively newly identified species, Candida auris. Despite the close relationship of these microorganisms, it seems that in the course of evolution, they have developed distinct biochemical, metabolic, and physiological adaptations, which they use to fit to commensal niches and achieve full virulence. Therefore, in this review, we describe the current knowledge on C. tropicalis, C. dubliniensis, and C. auris virulence factors, the formation of a mixed species biofilm and mutual communication, the environmental stress response and related changes in fungal cell metabolism, and the effect of pathogens on host defense response and susceptibility to antifungal agents used, highlighting differences with respect to C. albicans. Special attention is paid to common diagnostic problems resulting from similarities between these species and the emergence of drug resistance mechanisms. Understanding the different strategies to achieve virulence, used by important opportunistic pathogens of the genus Candida, is essential for proper diagnosis and treatment.
Collapse
|
140
|
In Vitro Antifungal Activity of LL-37 Analogue Peptides against Candida spp. J Fungi (Basel) 2022; 8:jof8111173. [PMID: 36354940 PMCID: PMC9694450 DOI: 10.3390/jof8111173] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Fungal infections have increased in recent decades with considerable morbidity and mortality, mainly in immunosuppressed or admitted-to-the-ICU patients. The fungal resistance to conventional antifungal treatments has become a public health problem, especially with Candida that presents resistance to several antifungals. Therefore, generating new alternatives of antifungal therapy is fundamental. One of these possibilities is the use of antimicrobial peptides, such as LL-37, which acts on the disruption of the microorganism membrane and promotes immunomodulatory effects in the host. In this study, we evaluated the in vitro antifungal activity of the LL-37 analogue peptides (AC-1, LL37-1, AC-2, and D) against different Candida spp. and clinical isolates obtained from patients with vulvovaginal candidiasis. Our results suggest that the peptides with the best ranges of MICs were LL37-1 and AC-2 (0.07 µM) against the strains studied. This inhibitory effect was confirmed by analyzing the yeast growth curves that evidenced a significant decrease in the fungal growth after exposure to LL-37 peptides. By the XTT technique we observed a significant reduction in the biofilm formation process when compared to yeasts untreated with the analogue peptides. In conclusion, we suggest that LL-37 analogue peptides may play an important antimicrobial role against Candida spp.
Collapse
|
141
|
Haslene‐Hox H, Nærdal GK, Mørch Y, Hageskal G, Tøndervik A, Turøy AV, Johnsen H, Klinkenberg G, Sletta H. High-throughput assay for effect screening of amphotericin B and bioactive components on filamentous Candida albicans. J Appl Microbiol 2022; 133:3113-3125. [PMID: 35947058 PMCID: PMC9804330 DOI: 10.1111/jam.15770] [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: 04/28/2022] [Revised: 07/01/2022] [Accepted: 08/06/2022] [Indexed: 01/05/2023]
Abstract
AIMS The aim of this study was to develop a high-throughput robotic microtiter plate-based screening assay for Candida albicans, optimizing growth conditions to replicate the filamentous biofilm growth found in vivo, and subsequently, to demonstrate the assay by evaluating the effect of nutritional drinks alone and in combination with the antifungal amphotericin B (AmB). METHODS AND RESULTS Candida albicans cultured in a defined growth medium showed filamentous growth in microcolonies, mimicking the morphology of oral mucosal disease (oral candidiasis). Addition of nutrient drinks containing fruit juices, fish oil and whey protein to the medium resulted in changed morphology and promoted growth as free yeast cells and with weak biofilm structures. Minimum inhibitory concentration of AmB on the biofilms was 0.25 μg ml-1 , and this was eightfold reduced (0.0038 μg ml-1 ) in the presence of the nutritional drinks. CONCLUSIONS The established assay demonstrated applicability for screening of antifungal and anti-biofilm effects of bioactive substances on C. albicans biofilm with clinically relevant morphology. SIGNIFICANCE AND IMPACT OF THE STUDY Candida albicans is the causative agent of the majority of fungal infections globally. The filamentous morphology of C. albicans and the ability to form biofilm are traits known to increase virulence and resistance towards antifungals. This study describes the development of a plate-based in vitro screening method mimicking the filamentous morphology of C. albicans found in vivo. The assay established can thus facilitate efficient antifungal drug discovery and development.
Collapse
Affiliation(s)
- Hanne Haslene‐Hox
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Guro Kruge Nærdal
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Yrr Mørch
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Gunhild Hageskal
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Anne Tøndervik
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | | | - Heidi Johnsen
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Geir Klinkenberg
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Håvard Sletta
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| |
Collapse
|
142
|
Menthol Inhibits Candida albicans Growth by Affecting the Membrane Integrity Followed by Apoptosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1297888. [PMID: 36337581 PMCID: PMC9635957 DOI: 10.1155/2022/1297888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/29/2022] [Accepted: 10/15/2022] [Indexed: 11/09/2022]
Abstract
Inclusion of Candida albicans in the list of pathogens with potential drug resistance threat in recent years has compelled scientists to explore novel and potent antifungal agents. In this study, we have evaluated anti-Candida potential of menthol against different growth forms and synergistic potential with fluconazole. Menthol inhibited planktonic growth of all the isolates completely at ≤3.58 mM and killed 99.9% inoculum at MIC, indicating that menthol is fungicidal. Menthol inhibited hyphal form growth completely at 0.62 mM. It has inhibited developing a biofilm by 79% at 3.58 mM, exhibiting excellent activity against recalcitrant biofilms. FIC index values of 0.182 and 0.093 indicate excellent synergistic activity between fluconazole and menthol against planktonic and biofilm growth, respectively. Menthol enhanced rate of OxPhos among 22% cells; arrested 71% cells at G2-M phase of cell cycle and induced apoptosis in 15% cells. Thus, menthol exhibits excellent anti-Candida activity against differentially susceptible isolates as well as various growth and morphological forms of C. albicans. Menthol affects membrane integrity thereby inducing oxidative stress followed by cell cycle arrest and apoptosis. Considering the excellent anti-Candida potential and as it is Generally Recognized as Safe by the Food and Drug Administration, it may find use in antifungal chemotherapy, alone or in combination.
Collapse
|
143
|
Kim YG, Lee JH, Park S, Khadke SK, Shim JJ, Lee J. Hydroquinones Including Tetrachlorohydroquinone Inhibit Candida albicans Biofilm Formation by Repressing Hyphae-Related Genes. Microbiol Spectr 2022; 10:e0253622. [PMID: 36190417 PMCID: PMC9602536 DOI: 10.1128/spectrum.02536-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/14/2022] [Indexed: 01/04/2023] Open
Abstract
Candida albicans is an opportunistic pathogenic fungus responsible for candidiasis. The pathogen readily forms antifungal agent-resistant biofilms on implanted medical devices or human tissue. Morphologic transition from yeast to filamentous cells and subsequent biofilm formation is a key virulence factor and a prerequisite for biofilm development by C. albicans. We investigated the antibiofilm and antifungal activities of 18 hydroquinones against fluconazole-resistant C. albicans. Tetrachlorohydroquinone (TCHQ) at subinhibitory concentrations (2 to 10 μg/mL) significantly inhibited C. albicans biofilm formation with an MIC of 50 μg/mL, whereas the backbone hydroquinone did not (MIC > 400 μg/mL), and it markedly inhibited cell aggregation and hyphal formation. Transcriptomic analyses showed that TCHQ downregulated the expressions of several hyphae-forming and biofilm-related genes (ALS3, ECE1, HWP1, RBT5, and UME6) but upregulated hyphae- and biofilm-inhibitory genes (IFD6 and YWP1). Furthermore, it prevented C. albicans biofilm development on porcine skin and at concentrations of 20 to 50 μg/mL was nontoxic to the nematode Caenorhabditis elegans and did not adversely affect Brassica rapa seed germination and growth. This study indicates that hydroquinones, particularly TCHQ, diminish the virulence, biofilm formation, and animal tissue adhesion of C. albicans, which suggests hydroquinones should be considered potential candidate antifungal agents against drug-resistant C. albicans strains. IMPORTANCE Persistence in chronic infections by Candida albicans is due to its ability of biofilm formation that endures conventional antifungals and host immune systems. Hence, the inhibition of biofilm formation and virulence characteristics is another mean of addressing infections. This study is a distinctive one since 18 hydroquinone analogues were screened and TCHQ efficiently inhibited the biofilm formation by C. albicans with significantly changed expressional profile of hyphae-forming and biofilm-related genes. The antibiofilm efficacy was confirmed using a porcine skin model and chemical toxicity was investigated using plant seed germination and nematode models. Our findings reveal that TCHQ can efficiently control the C. albicans biofilms and virulence characteristics.
Collapse
Affiliation(s)
- Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Sunyoung Park
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Sagar Kiran Khadke
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jae-Jin Shim
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
144
|
Khalil FO, Taj MB, Ghonaim EM, Abed El-Sattar S, Elkhadry SW, El-Refai H, Ali OM, Elgawad ASA, Alshater H. Hydrothermal assisted biogenic synthesis of silver nanoparticles: A potential study on virulent candida isolates from COVID-19 patients. PLoS One 2022; 17:e0269864. [PMID: 36201485 PMCID: PMC9536612 DOI: 10.1371/journal.pone.0269864] [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: 01/05/2022] [Accepted: 05/27/2022] [Indexed: 11/07/2022] Open
Abstract
Till now the exact mechanism and effect of biogenic silver nanoparticles on fungus is an indefinable question. To focus on this issue, the first time we prepared hydrothermal assisted thyme coated silver nanoparticles (T/AgNPs) and their toxic effect on Candida isolates were determined. The role of thyme (Thymus Vulgaris) in the reduction of silver ions and stabilization of T/AgNPs was estimated by Fourier transforms infrared spectroscopy, structure and size of present silver nanoparticles were detected via atomic force microscopy as well as high-resolution transmission electron microscopy. The biological activity of T/AgNPs was observed against Candida isolates from COVID-19 Patients. Testing of virulence of Candida species using Multiplex PCR. T/AgNPs proved highly effective against Candida albicans, Candida kruzei, Candida glabrata and MIC values ranging from 156.25 to 1,250 μg/mL and MFC values ranging from 312.5 to 5,000 μg/mL. The structural and morphological modifications due to T/AgNPs on Candida albicans were detected by TEM. It was highly observed that when Candida albicans cells were subjected to 50 and 100 μg/mL T/AgNPs, a remarkable change in the cell wall and cell membrane was observed.
Collapse
Affiliation(s)
- Fatma O. Khalil
- Clinical and Molecular Microbiology and Immunology Department, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Muhammad B. Taj
- Division of Inorganic Chemistry, Institute of Chemistry, The Islamia University Bahawalpur, Bahawalpur, Pakistan
- * E-mail: (MBT); (OMA)
| | - Enas M. Ghonaim
- Clinical and Molecular Microbiology and Immunology Department, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Shimaa Abed El-Sattar
- Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Sally W. Elkhadry
- Epidemiology and Preventive Medicine Department, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Hala El-Refai
- Clinical and Molecular Microbiology and Immunology Department, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Omar M. Ali
- Department of Chemistry, Turabah University College, Turabah Branch, Taif University, Taif Saudi Arabia
- * E-mail: (MBT); (OMA)
| | - Ahmed Salah A. Elgawad
- Department of Clinical Pathology, National Liver Institute, Menoufia University Hospital, Menoufia University, Shebin El-Kom, Egypt
| | - Heba Alshater
- Department of Forensic Medicine and Clinical Toxicology, Menoufia University Hospital, Menoufia University, Shebin El-Kom, Egypt
| |
Collapse
|
145
|
Raman Metabolomics of Candida auris Clades: Profiling and Barcode Identification. Int J Mol Sci 2022; 23:ijms231911736. [PMID: 36233043 PMCID: PMC9569935 DOI: 10.3390/ijms231911736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
This study targets on-site/real-time taxonomic identification and metabolic profiling of seven different Candida auris clades/subclades by means of Raman spectroscopy and imaging. Representative Raman spectra from different Candida auris samples were systematically deconvoluted by means of a customized machine-learning algorithm linked to a Raman database in order to decode structural differences at the molecular scale. Raman analyses of metabolites revealed clear differences in cell walls and membrane structure among clades/subclades. Such differences are key in maintaining the integrity and physical strength of the cell walls in the dynamic response to external stress and drugs. It was found that Candida cells use the glucan structure of the extracellular matrix, the degree of α-chitin crystallinity, and the concentration of hydrogen bonds between its antiparallel chains to tailor cell walls’ flexibility. Besides being an effective ploy in survivorship by providing stiff shields in the α–1,3–glucan polymorph, the α–1,3–glycosidic linkages are also water-insoluble, thus forming a rigid and hydrophobic scaffold surrounded by a matrix of pliable and hydrated β–glucans. Raman analysis revealed a variety of strategies by different clades to balance stiffness, hydrophobicity, and impermeability in their cell walls. The selected strategies lead to differences in resistance toward specific environmental stresses of cationic/osmotic, oxidative, and nitrosative origins. A statistical validation based on principal component analysis was found only partially capable of distinguishing among Raman spectra of clades and subclades. Raman barcoding based on an algorithm converting spectrally deconvoluted Raman sub-bands into barcodes allowed for circumventing any speciation deficiency. Empowered by barcoding bioinformatics, Raman analyses, which are fast and require no sample preparation, allow on-site speciation and real-time selection of appropriate treatments.
Collapse
|
146
|
Sousa IS, Mello TP, Pereira EP, Granato MQ, Alviano CS, Santos ALS, Kneipp LF. Biofilm Formation by Chromoblastomycosis Fungi Fonsecaea pedrosoi and Phialophora verrucosa: Involvement with Antifungal Resistance. J Fungi (Basel) 2022; 8:jof8090963. [PMID: 36135688 PMCID: PMC9504689 DOI: 10.3390/jof8090963] [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: 07/01/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Patients with chromoblastomycosis (CBM) suffer chronic tissue lesions that are hard to treat. Considering that biofilm is the main growth lifestyle of several pathogens and it is involved with both virulence and resistance to antimicrobial drugs, we have investigated the ability of CBM fungi to produce this complex, organized and multicellular structure. Fonsecaea pedrosoi and Phialophora verrucosa conidial cells were able to adhere on a polystyrene abiotic substrate, differentiate into hyphae and produce a robust viable biomass containing extracellular matrix. Confocal laser scanning microscopy (CLSM) and scanning electron microscopy (SEM) showed the tridimensional architecture of the mature biofilms, revealing a dense network of interconnected hyphae, inner channels and amorphous extracellular polymeric material. Interestingly, the co-culture of each fungus with THP-1 macrophage cells, used as a biotic substrate, induced the formation of a mycelial trap covering and damaging the macrophages. In addition, the biofilm-forming cells of F. pedrosoi and P. verrucosa were more resistant to the conventional antifungal drugs than the planktonic-growing conidial cells. The efflux pump activities of P. verrucosa and F. pedrosoi biofilms were significantly higher than those measured in conidia. Taken together, the data pointed out the biofilm formation by CBM fungi and brought up a discussion of the relevance of studies about their antifungal resistance mechanisms.
Collapse
Affiliation(s)
- Ingrid S. Sousa
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| | - Thaís P. Mello
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil
| | - Elaine P. Pereira
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| | - Marcela Q. Granato
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| | - Celuta S. Alviano
- Laboratório de Estrutura de Microrganismos, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 20020-000, Brazil
| | - Lucimar F. Kneipp
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 20020-000, Brazil
- Correspondence:
| |
Collapse
|
147
|
Mullis MN, Ghione C, Lough-Stevens M, Goldstein I, Matsui T, Levy SF, Dean MD, Ehrenreich IM. Complex genetics cause and constrain fungal persistence in different parts of the mammalian body. Genetics 2022; 222:6698696. [PMID: 36103708 PMCID: PMC9630980 DOI: 10.1093/genetics/iyac138] [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: 05/26/2022] [Accepted: 08/26/2022] [Indexed: 12/05/2022] Open
Abstract
Determining how genetic polymorphisms enable certain fungi to persist in mammalian hosts can improve understanding of opportunistic fungal pathogenesis, a source of substantial human morbidity and mortality. We examined the genetic basis of fungal persistence in mice using a cross between a clinical isolate and the lab reference strain of the budding yeast Saccharomyces cerevisiae. Employing chromosomally encoded DNA barcodes, we tracked the relative abundances of 822 genotyped, haploid segregants in multiple organs over time and performed linkage mapping of their persistence in hosts. Detected loci showed a mix of general and antagonistically pleiotropic effects across organs. General loci showed similar effects across all organs, while antagonistically pleiotropic loci showed contrasting effects in the brain vs the kidneys, liver, and spleen. Persistence in an organ required both generally beneficial alleles and organ-appropriate pleiotropic alleles. This genetic architecture resulted in many segregants persisting in the brain or in nonbrain organs, but few segregants persisting in all organs. These results show complex combinations of genetic polymorphisms collectively cause and constrain fungal persistence in different parts of the mammalian body.
Collapse
Affiliation(s)
- Martin N Mullis
- University of Southern California Molecular and Computational Biology Section, Department of Biological Sciences, , Los Angeles, CA 90089, USA
| | - Caleb Ghione
- University of Southern California Molecular and Computational Biology Section, Department of Biological Sciences, , Los Angeles, CA 90089, USA
| | - Michael Lough-Stevens
- University of Southern California Molecular and Computational Biology Section, Department of Biological Sciences, , Los Angeles, CA 90089, USA
| | - Ilan Goldstein
- University of Southern California Molecular and Computational Biology Section, Department of Biological Sciences, , Los Angeles, CA 90089, USA
| | - Takeshi Matsui
- Stanford University Joint Initiative for Metrology in Biology, , CA 94305, USA
- SLAC National Accelerator Laboratory , Menlo Park, CA, 94025, USA
- Stanford University Department of Genetics, , Stanford, CA 94305, USA
| | - Sasha F Levy
- Stanford University Joint Initiative for Metrology in Biology, , CA 94305, USA
- SLAC National Accelerator Laboratory , Menlo Park, CA, 94025, USA
- Stanford University Department of Genetics, , Stanford, CA 94305, USA
| | - Matthew D Dean
- University of Southern California Molecular and Computational Biology Section, Department of Biological Sciences, , Los Angeles, CA 90089, USA
| | - Ian M Ehrenreich
- University of Southern California Molecular and Computational Biology Section, Department of Biological Sciences, , Los Angeles, CA 90089, USA
| |
Collapse
|
148
|
Mohd Badri PEA, Rismayuddin NAR, Kenali NM, Darnis DS, Arzmi MH. Characterization of Cervus timorensis Velvet Antler and its Effect on Biofilm Formation of Candida Species. Med Mycol 2022; 60:6696968. [PMID: 36099875 DOI: 10.1093/mmy/myac073] [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: 05/18/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/12/2022] Open
Abstract
Oral biofilms comprise extracellular polysaccharides and polymicrobial microorganisms. The objectives of the study were to characterize the deer velvet antler (DVA) compounds and their effect on Candida species biofilm formation with the hypothesis that DVA inhibits the biofilm of Candida spp. Liquid Chromatography-Quadrupole Time of Flight-Mass Spectrometry (LC-QTOF-MS) was conducted to characterize the DVA compounds. To study the effect of DVA on biofilm, Candida albicans ATCC MYA-4901 (ALT5), AIDS isolate (ALC2), oral cancer isolate (ALC3), C. dubliniensis ATCC MYA-2975, C. glabrata ATCC 90030, C. krusei 14 243, C. lusitaniae ATCC 34449, C. parapsilosis ATCC 22019, and C. tropicalis ATCC 13803 were inoculated with DVA in separate wells of a 96-well plate containing RPMI-1640 followed by 72 h incubation. A total of 45 compounds were detected in the DVA extract. C. lusitaniae exhibited a higher percentage of biofilm biomass reduction when treated with DVA extract (66.10% ± 5.33), followed by ALC3 (44.12% ± 6.24). However, C. glabrata, C. krusei, and C. parapsilosis showed no reduction in biofilm biomass after being treated with DVA extract. Most Candida strains also exhibited decreased total cell count when treated with DVA extract, except for ALC3 and C. krusei. ALT5 had the lowest total cell count (0.17 × 105 cells/mL) when cultured with DVA extract. In conclusion, DVA extract inhibits Candida spp. biofilm formation except for C. glabrata, C. krusei, and C. parapsilosis.
Collapse
Affiliation(s)
- Puteri Elysa Alia Mohd Badri
- Cluster of Cancer Research Initiative IIUM (COCRII), International Islamic University Malaysia, Kuantan, Pahang, Malaysia.,Department of Biotechnology, Kulliyyah of Science, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Nurul Alia Risma Rismayuddin
- Cluster of Cancer Research Initiative IIUM (COCRII), International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Norzaiti Mohd Kenali
- Cluster of Cancer Research Initiative IIUM (COCRII), International Islamic University Malaysia, Kuantan, Pahang, Malaysia.,Department of Paediatric Dentistry and Dental Public Health, Kulliyyah of Dentistry, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Deny Susanti Darnis
- Department of Chemistry, Kulliyyah of Science, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Mohd Hafiz Arzmi
- Cluster of Cancer Research Initiative IIUM (COCRII), International Islamic University Malaysia, Kuantan, Pahang, Malaysia.,Department of Fundamental Dental and Medical Sciences, Kulliyyah of Dentistry, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| |
Collapse
|
149
|
Biofilm Alterations on the Stepwise Acquisition of Fluconazole-resistant Candida Albicans Isolates. INTERNATIONAL JOURNAL OF DERMATOLOGY AND VENEREOLOGY 2022; 5:132-139. [PMID: 36199677 PMCID: PMC9521591 DOI: 10.1097/jd9.0000000000000223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 12/24/2021] [Accepted: 01/11/2022] [Indexed: 12/05/2022]
Abstract
By assessing and comparing the phenotypic changes on the stepwise acquisition of fluconazole resistant Candida albicans isolates, we could find and describe the relationship between drug resistance and biofilm formation ability in a series of clonal strains.
Collapse
|
150
|
Rivera-Yañez CR, Ruiz-Hurtado PA, Reyes-Reali J, Mendoza-Ramos MI, Vargas-Díaz ME, Hernández-Sánchez KM, Pozo-Molina G, Méndez-Catalá CF, García-Romo GS, Pedroza-González A, Méndez-Cruz AR, Nieto-Yañez O, Rivera-Yañez N. Antifungal Activity of Mexican Propolis on Clinical Isolates of Candida Species. Molecules 2022; 27:molecules27175651. [PMID: 36080417 PMCID: PMC9457601 DOI: 10.3390/molecules27175651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Infections caused by micro-organisms of the genus Candida are becoming a growing health problem worldwide. These fungi are opportunistic commensals that can produce infections—clinically known as candidiasis—in immunocompromised individuals. The indiscriminate use of different anti-fungal treatments has triggered the resistance of Candida species to currently used therapies. In this sense, propolis has been shown to have potent antimicrobial properties and thus can be used as an approach for the inhibition of Candida species. Therefore, this work aims to evaluate the anti-Candida effects of a propolis extract obtained from the north of Mexico on clinical isolates of Candida species. Candida species were specifically identified from oral lesions, and both the qualitative and quantitative anti-Candida effects of the Mexican propolis were evaluated, as well as its inhibitory effect on C. albicans isolate’s germ tube growth and chemical composition. Three Candida species were identified, and our results indicated that the inhibition halos of the propolis ranged from 7.6 to 21.43 mm, while that of the MFC and FC50 ranged from 0.312 to 1.25 and 0.014 to 0.244 mg/mL, respectively. Moreover, the propolis was found to inhibit germ tube formation (IC50 ranging from 0.030 to 1.291 mg/mL). Chemical composition analysis indicated the presence of flavonoids, including pinocembrin, baicalein, pinobanksin chalcone, rhamnetin, and biochanin A, in the Mexican propolis extract. In summary, our work shows that Mexican propolis presents significant anti-Candida effects related to its chemical composition, and also inhibits germ tube growth. Other Candida species virulence factors should be investigated in future research in order to determine the mechanisms associated with antifungal effects against them.
Collapse
Affiliation(s)
- Claudia Rebeca Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Porfirio Alonso Ruiz-Hurtado
- Laboratorio de Toxicología de Productos Naturales, Departamento de Farmacia, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Av. Wilfrido Massieu, Gustavo A. Madero 07738, Mexico
| | - Julia Reyes-Reali
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - María Isabel Mendoza-Ramos
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - María Elena Vargas-Díaz
- Laboratorio de Química de Productos Naturales, Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala, Ciudad de México 11340, Mexico
| | - Karla Mariela Hernández-Sánchez
- Laboratorio de Química de Productos Naturales, Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala, Ciudad de México 11340, Mexico
| | - Glustein Pozo-Molina
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Claudia Fabiola Méndez-Catalá
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Gina Stella García-Romo
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Alexander Pedroza-González
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Adolfo René Méndez-Cruz
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Oscar Nieto-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (O.N.-Y.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| | - Nelly Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (O.N.-Y.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| |
Collapse
|