Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Technol Adv Mater ; 24(1): 2246867, 2023.
Article in English | MEDLINE | ID: mdl-37680974

ABSTRACT

Effective treatment of bacterial infections requires methods that accurately and quickly identify which antibiotic should be prescribed. This review describes recent research on the development of optotracing methodologies for bacterial and biofilm detection and diagnostics. Optotracers are small, chemically well-defined, anionic fluorescent tracer molecules that detect peptide- and carbohydrate-based biopolymers. This class of organic molecules (luminescent conjugated oligothiophenes) show unique electronic, electrochemical and optical properties originating from the conjugated structure of the compounds. The photophysical properties are further improved as donor-acceptor-donor (D-A-D)-type motifs are incorporated in the conjugated backbone. Optotracers bind their biopolymeric target molecules via electrostatic interactions. Binding alters the optical properties of these tracer molecules, shown as altered absorption and emission spectra, as well as ON-like switch of fluorescence. As the optotracer provides a defined spectral signature for each binding partner, a fingerprint is generated that can be used for identification of the target biopolymer. Alongside their use for in situ experimentation, optotracers have demonstrated excellent use in studies of a number of clinically relevant microbial pathogens. These methods will find widespread use across a variety of communities engaged in reducing the effect of antibiotic resistance. This includes basic researchers studying molecular resistance mechanisms, academia and pharma developing new antimicrobials targeting biofilm infections and tests to diagnose biofilm infections, as well as those developing antibiotic susceptibility tests for biofilm infections (biofilm-AST). By iterating between the microbial world and that of plants, development of the optotracing technology has become a prime example of successful cross-feeding across the boundaries of disciplines. As optotracers offers a capacity to redefine the way we work with polysaccharides in the microbial world as well as with plant biomass, the technology is providing novel outputs desperately needed for global impact of the threat of antimicrobial resistance as well as our strive for a circular bioeconomy.

2.
Front Cell Infect Microbiol ; 12: 981454, 2022.
Article in English | MEDLINE | ID: mdl-36118028

ABSTRACT

Candida albicans is the most common fungal pathogen in humans, implicated in hospital-acquired infections, secondary infections in human immunodeficiency virus (HIV) patients, and is a significant contributor to the global antimicrobial resistance (AMR) burden. Early detection of this pathogen is needed to guide preventative strategies and the selection and development of therapeutic treatments. Fungal biofilms are a unique heterogeneous mix of cell types, extracellular carbohydrates and amyloid aggregates. Perhaps due to the dominance of carbohydrates in fungi, to date, few specific methods are available for the detection of fungal biofilms. Here we present a new optotracing-based method for the detection and analysis of yeast and biofilms based on C. albicans SC5314 as a model. Using commercial extracts of cell wall carbohydrates, we showed the capability of the optotracer EbbaBiolight 680 for detecting chitin and ß-glucans. The sensitivity of this tracer to these carbohydrates in their native environment within fungal cells enabled the visualization of both yeast and hyphal forms of the microbe. Analysis of optotracer fluorescence by confocal laser scanning microscopy revealed extensive staining of fungi cell walls as well as the presence of intracellular amyloid aggregates within a subpopulation of cells within the biofilm. Further analysis of the photophysical properties of bound tracers by spectroscopy and spectral imaging revealed polymorphisms between amyloid aggregates within yeast and hyphal cells and enabled their differentiation. With exceptional spatial and temporal resolution, this assay adds a new technique that facilitates future understanding of fungal biofilms and their formation, and enables direct, unbiased diagnostics of these medically relevant biofilms, as well as the development of antifungal strategies.


Subject(s)
Candida albicans , beta-Glucans , Antifungal Agents , Biofilms , Carbohydrates , Chitin , Humans , Plant Extracts
SELECTION OF CITATIONS
SEARCH DETAIL
...