Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
J Am Chem Soc ; 143(14): 5413-5424, 2021 04 14.
Article in English | MEDLINE | ID: covidwho-1387160

ABSTRACT

Methods for tracking RNA inside living cells without perturbing their natural interactions and functions are critical within biology and, in particular, to facilitate studies of therapeutic RNA delivery. We present a stealth labeling approach that can efficiently, and with high fidelity, generate RNA transcripts, through enzymatic incorporation of the triphosphate of tCO, a fluorescent tricyclic cytosine analogue. We demonstrate this by incorporation of tCO in up to 100% of the natural cytosine positions of a 1.2 kb mRNA encoding for the histone H2B fused to GFP (H2B:GFP). Spectroscopic characterization of this mRNA shows that the incorporation rate of tCO is similar to cytosine, which allows for efficient labeling and controlled tuning of labeling ratios for different applications. Using live cell confocal microscopy and flow cytometry, we show that the tCO-labeled mRNA is efficiently translated into H2B:GFP inside human cells. Hence, we not only develop the use of fluorescent base analogue labeling of nucleic acids in live-cell microscopy but also, importantly, show that the resulting transcript is translated into the correct protein. Moreover, the spectral properties of our transcripts and their translation product allow for their straightforward, simultaneous visualization in live cells. Finally, we find that chemically transfected tCO-labeled RNA, unlike a state-of-the-art fluorescently labeled RNA, gives rise to expression of a similar amount of protein as its natural counterpart, hence representing a methodology for studying natural, unperturbed processing of mRNA used in RNA therapeutics and in vaccines, like the ones developed against SARS-CoV-2.


Subject(s)
Fluorescence , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Molecular Imaging , RNA, Messenger/analysis , RNA, Messenger/metabolism , Cell Line, Tumor , Cytosine/analogs & derivatives , Cytosine/analysis , Cytosine/chemical synthesis , Cytosine/chemistry , Fluorescent Dyes/chemical synthesis , Green Fluorescent Proteins/metabolism , Histones/metabolism , Humans , Molecular Structure , RNA, Messenger/chemistry , RNA, Messenger/therapeutic use , Spectrometry, Fluorescence , COVID-19 Drug Treatment
2.
Chem Commun (Camb) ; 57(51): 6229-6232, 2021 Jun 24.
Article in English | MEDLINE | ID: covidwho-1246405

ABSTRACT

Tracking the viral progression of SARS-CoV-2 in COVID-19 infected body tissues is an emerging need of the current pandemic. Imaging at near infrared second biological window (NIR-II) offers striking benefits over the other technologies to explore deep-tissue information. Here we design, synthesise and characterise a molecular probe that selectively targets the N-gene of SARS-CoV-2. Highly specific antisense oligonucleotides (ASOs) were conjugated to lead sulfide quantum dots using a UV-triggered thiol-ene click chemistry for the recognition of viral RNA. Our ex vivo imaging studies demonstrated that the probe exhibits aggregation induced NIR-II emission only in presence of SARS-CoV-2 RNA which can be attributed to the efficient hybridisation of the ASOs with their target RNA strands.


Subject(s)
COVID-19/diagnosis , COVID-19/virology , Fluorescent Dyes/chemistry , Oligonucleotides, Antisense/chemistry , Quantum Dots/chemistry , SARS-CoV-2/isolation & purification , Spectroscopy, Near-Infrared/methods , Animals , COVID-19/diagnostic imaging , COVID-19/metabolism , Click Chemistry/methods , Fluorescent Dyes/chemical synthesis , Humans , Lung/diagnostic imaging , Lung/metabolism , Lung/virology , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Models, Animal , SARS-CoV-2/genetics , SARS-CoV-2/metabolism
3.
Anal Chem ; 92(19): 13396-13404, 2020 10 06.
Article in English | MEDLINE | ID: covidwho-933642

ABSTRACT

Rapid, accurate, reliable, and risk-free tracking of pathogenic microorganisms at the single-cell level is critical to achieve efficient source control and prevent outbreaks of microbial infectious diseases. For the first time, we report a promising approach for integrating the concepts of a remarkably large Stokes shift and dual-recognition into a single matrix to develop a pathogenic microorganism stimuli-responsive ratiometric fluorescent nanoprobe with speed, cost efficiency, stability, ultrahigh specificity, and sensitivity. As a proof-of-concept, we selected the Gram-positive bacterium Staphylococcus aureus (S. aureus) as the target analyte model, which easily bound to its recognition aptamer and the broad-spectrum glycopeptide antibiotic vancomycin (Van). To improve the specificity and short sample-to-answer time, we employed classic noncovalent π-π stacking interactions as a driving force to trigger the binding of Van and aptamer dual-functionalized near-infrared (NIR) fluorescent Apt-Van-QDs to the surface of an unreported blue fluorescent π-rich electronic carbon nanoparticles (CNPs), achieving S. aureus stimuli-responsive ratiometric nanoprobe Apt-Van-QDs@CNPs. In the assembly of Apt-Van-QDs@CNPs, the blue CNPs (energy donor) and NIR Apt-Van-QDs (energy acceptor) became close to allow the fluorescence resonance energy transfer (FRET) process, leading to a remarkable blue fluorescence quenching for the CNPs at ∼465 nm and a clear NIR fluorescence enhancement for Apt-Van-QDs at ∼725 nm. In the presence of S. aureus, the FRET process from CNPs to Apt-Van-QDs was disrupted, causing the nanoprobe Apt-Van-QDs@CNPs to display a ratiometric fluorescent response to S. aureus, which exhibited a large Stokes shift of ∼260 nm and rapid sample-to-answer detection time (∼30.0 min). As expected, the nanoprobe Apt-Van-QDs@CNPs showed an ultrahigh specificity for ratiometric fluorescence detection of S. aureus with a good detection limit of 1.0 CFU/mL, allowing the assay at single-cell level. Moreover, we also carried out the precise analysis of S. aureus in actual samples with acceptable results. We believe that this work offers new insight into the rational design of efficient ratiometric nanoprobes for rapid on-site accurate screening of pathogenic microorganisms at the single-cell level in the early stages, especially during the worldwide spread of COVID-19 today.


Subject(s)
Bacteria/chemistry , Bacterial Infections/diagnosis , Bacterial Infections/microbiology , Biosensing Techniques/methods , Fluorescent Dyes/chemical synthesis , Nanotechnology/methods , Anti-Bacterial Agents/pharmacology , Aptamers, Nucleotide , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/microbiology , Fluorescence , Fluorescence Resonance Energy Transfer , Food Microbiology/methods , Humans , Nanoparticles , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/microbiology , Sensitivity and Specificity , Spectroscopy, Near-Infrared , Staphylococcal Infections/diagnosis , Staphylococcal Infections/microbiology , Staphylococcus aureus/chemistry , Vancomycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL