Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.196
Filter
1.
J Microbiol Biotechnol ; 34(6): 1322-1327, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38881169

ABSTRACT

The accurate and rapid detection of methicillin-resistant Staphylococcus aureus (MRSA) holds significant clinical importance. This work presents a new method for detecting methicillin-resistant Staphylococcus aureus (S. aureus) in clinical samples. The method uses an aptamer-based colorimetric assay that combines a recognizing probe to identify the target and split DNAzyme to amplify the signal, resulting in a highly sensitive and direct analysis of methicillin-resistance. The identification of the PBP2a protein on the membrane of S. aureus in clinical samples leads to the allosterism of the recognizing probe, and thus provides a template for the proximity ligation of split DNAzyme. The proximity ligation of split DNAzyme forms an intact DNAzyme to identify the loop section in the L probe and generates a nicking site to release the loop sequence ("3" and "4" fragments). The "3" and "4" fragments forms an intact sequence to induce the catalytic hairpin assembly, exposing the G-rich section. The released the G-rich sequence of LR probe induces the formation of G-quadruplex-hemin DNAzyme as a colorimetric signal readout. The absorption intensity demonstrated a strong linear association with the logarithm of the S. aureus concentration across a wide range of 5 orders of magnitude dynamic range under the optimized experimental parameters. The limit of detection was calculated to be 23 CFU/ml and the method showed high selectivity for MRSA.


Subject(s)
Aptamers, Nucleotide , Colorimetry , DNA, Catalytic , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , DNA, Catalytic/metabolism , Colorimetry/methods , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Staphylococcal Infections/microbiology , Staphylococcal Infections/diagnosis , Humans , Biosensing Techniques/methods , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Sensitivity and Specificity , Methicillin Resistance , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/genetics
2.
Anal Methods ; 16(25): 4116-4123, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38855960

ABSTRACT

MicroRNAs (miRNAs) have been involved in many biological processes and are regarded as promising biomarkers. The short sequence, low abundance and highly homologous interference sequences greatly hinder the accurate detection of miRNAs. Here, a cascade branch migration-triggered strand displacement amplification (CBM-TSDA) strategy was developed for the first time for specific and sensitive detection of miRNA-155 (miR-155). In the presence of target miR-155, the CBM was initiated and two Y-shaped probes were eventually produced. Next, the Y-shaped probes were transformed into three-way junction (3WJ) structures and triggered the SDA to produce a large number of G-quadruplex (G4) structures. Finally, the increased fluorescence signal of G4/Thioflavin T (ThT) was used to quantify miR-155. Meanwhile, the colorimetric responses of the G4-hemin DNAzyme could be used as supplementary detection to obtain a dual-mode signal readout. This detection strategy showed high detection sensitivity, and the limit of detection was 0.28 pM in the fluorescence detection mode and 0.34 pM in the colorimetric detection mode. Notably, it showed high detection specificity, being able to discriminate the single-base mutations of the target with a high discrimination factor. The strategy also possessed excellent capacity for miR-155 detection in cell lysates and real human blood samples. The developed strategy provides a promising detection platform for miRNA, which may be applied to early clinical diagnosis.


Subject(s)
Limit of Detection , MicroRNAs , Nucleic Acid Amplification Techniques , MicroRNAs/blood , MicroRNAs/analysis , Humans , Nucleic Acid Amplification Techniques/methods , G-Quadruplexes , Colorimetry/methods , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Biosensing Techniques/methods , Spectrometry, Fluorescence/methods
3.
Molecules ; 29(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38893308

ABSTRACT

8-17 DNAzymes (8-17, 17E, Mg5, and 17EV1) are in vitro-selected catalytic DNA molecules that are capable of cleaving complementary RNAs. The conserved residues in their similar catalytic cores, together with the metal ions, were suggested to contribute to the catalytic reaction. Based on the contribution of the less conserved residues in the bulge loop residues (W12, A15, A15.0) and the internal stem, new catalytic cores of 8-17 DNAzymes were programmed. The internal stem CTC-GAG seems to be more favorable for the DNAzymes than CCG-GGC, while an extra W12.0 led to a significant loss of activity of DNAzymes, which is contrary to the positive effect of A15.0, by which a new active DNAzyme 17EM was derived. It conducts a faster reaction than 17E. It is most active in the presence of Pb2+, with the metal ion preference of Pb2+ >> Zn2+ > Mn2+ > Ca2+ ≈ Mg2+. In the Pb2+ and Zn2+-mediated reactions of 17EM and 17E, the same Na+- and pH dependence were also observed as what was observed for 17E and other 8-17 DNAzymes. Therefore, 17EM is another member of the 8-17 DNAzymes, and it could be applied as a potential biosensor for RNA and metal ions.


Subject(s)
DNA, Catalytic , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Nucleic Acid Conformation , Catalysis , Hydrogen-Ion Concentration , Catalytic Domain , Base Sequence , Metals/chemistry
5.
ACS Appl Mater Interfaces ; 16(24): 30766-30775, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38833714

ABSTRACT

Endowing current artificial chemical reactions (ACRs) with high specificity and intricate activation capabilities is crucial for expanding their applications in accurate bioimaging within living cells. However, most of the reported ACR-based evaluations relied on either single biomarker stimuli or dual activators without obvious biological relevance, still limiting their accuracy and fidelity. Herein, taking the metal-ion-dependent DNAzyme cleavage reaction as a model ACR, two regulators, glutathione (GSH) and telomerase (TE) activated DNAzyme cleavage reactions, were exploited for precise discrimination of cancerous cells from normal cells. DNA probe was self-assembled into the ZIF-90 nanoparticle framework to construct coordination-driven nanoprobes. This approach enhances the stability and specificity of tumor imaging by utilizing biomarkers associated with rapid tumor proliferation and those commonly overexpressed in tumors. In conclusion, the research not only paves the way for new perspectives in cell biology and pathology studies but also lays a solid foundation for the advancement of biomedical imaging and disease diagnostic technologies.


Subject(s)
DNA, Catalytic , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Humans , Nanoparticles/chemistry , Glutathione/metabolism , Glutathione/chemistry , Telomerase/metabolism , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Cell Line, Tumor , Optical Imaging
6.
Anal Chim Acta ; 1312: 342764, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38834269

ABSTRACT

BACKGROUND: Osteopontin (OPN) is closely associated with tumorigenesis, growth, invasion, and immune escape and it serves as a plasma biomarker for hepatocellular carcinoma (HCC). Nevertheless, the accurate and rapid detection of low-abundance OPN still poses significant challenges. Currently, the majority of protein detection methods rely heavily on large precision instruments or involve complex procedures. Therefore, developing a simple, enzyme-free, rapid colorimetric analysis method with high sensitivity is imperative. RESULTS: In this study, we have developed a portable colorimetric biosensor by integrating the triple-helix aptamer probe (THAP) and catalytic hairpin assembly (CHA) strategy, named as T-CHA. After binding to the OPN, the trigger probe can be released from THAP, then initiates the CHA reaction and outputs the signal through the formation of a G-quadruplex/Hemin DNAzyme with horseradish peroxidase-like activity. Consequently, this colorimetric sensor achieves visual free-labeled detection without additional fluorophore modification and allows for accurate quantification by measuring the optical density of the solution at 650 nm. Under optimal conditions, the logarithmic values of various OPN concentrations exhibit satisfactory linearity in the range of 5 pg mL-1 to 5 ng mL-1, with a detection limit of 2.04 pg mL-1. Compared with the widely used ELISA strategy, the proposed T-CHA strategy is rapid (∼105 min), highly sensitive, and cost-effective. SIGNIFICANCE: The T-CHA strategy, leveraging the low background leakage of THAP and the high catalytic efficiency of CHA, has been successfully applied to the detection of OPN in plasma, demonstrating significant promise for the early diagnosis of HCC in point-of-care testing. Given the programmability of DNA and the universality of T-CHA, it can be readily modified for analyzing other useful tumor biomarkers.


Subject(s)
Aptamers, Nucleotide , Colorimetry , Osteopontin , Colorimetry/methods , Aptamers, Nucleotide/chemistry , Humans , Osteopontin/blood , Osteopontin/chemistry , Osteopontin/analysis , Biosensing Techniques/methods , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Limit of Detection , G-Quadruplexes
7.
Anal Chem ; 96(24): 9909-9916, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38830056

ABSTRACT

The development of the Point-of-Care Testing (POCT) platform that combines convenience and cost-effectiveness is crucial for enabling the visual detection of disease biomarkers. In this work, a POCT platform for the sensitive in situ detection of prostate specific antigen (PSA) with dual-signal output was constructed by functionalizing the Eppendorf (EP) tube. This was achieved through the modification of aptamer hairpin probes (AHPs) on the lid of the EP tube and the assembly of a nanoenzyme hydrogel film on its inner wall. The target could trigger the release of Ag+ by AHP and subsequently activate Ag+-dependent DNAzyme (Ag-DNAzyme). This would initiate the cleavage of the DNA-Au/Pt NP hydrogel network, leading to the release of Au/Pt NPs. The released Au/Pt NPs exhibit both peroxidase (POD)-like and catalase (CAT)-like activity to produce a colorimetric response and induce liquid flow under pressure. Therefore, the target can be measured visually and quantitatively through colorimetric analysis and the measurement of total dissolved solids (TDS) using a pressure-triggered liquid flow device integrated into the platform. The designed platform is distinguished by its simplicity, specificity, cost-effectiveness, and remarkable sensitivity. It allows for the visual detection of PSA within concentration ranges of 0.5-100 ng/L (colorimetric) and 3-100 ng/L (TDS reading), boasting detection limits as low as 0.15 ng/L (colorimetric) and 0.57 ng/L (TDS reading). The strategy of target-triggered nanoenzyme release significantly enhances sensitivity and provides a guiding approach for visual biomarker detection.


Subject(s)
Aptamers, Nucleotide , Colorimetry , DNA, Catalytic , Gold , Metal Nanoparticles , Point-of-Care Testing , Prostate-Specific Antigen , Prostate-Specific Antigen/analysis , Humans , Gold/chemistry , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Metal Nanoparticles/chemistry , Aptamers, Nucleotide/chemistry , Platinum/chemistry , Hydrogels/chemistry , Biosensing Techniques , Silver/chemistry , Limit of Detection
8.
Anal Chem ; 96(24): 9866-9875, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38835317

ABSTRACT

Herein, a dual self-protected DNAzyme-based 3D DNA walker (dSPD walker), composed of activated dual self-protected walking particles (ac-dSPWPs) and track particles (TPs), was constructed for ultrasensitive and ultrahigh-speed fluorescence detection and imaging of microRNAs (miRNAs) in living cells. Impressively, compared with the defect that "one" target miRNA only initiates "one" walking arm of the conventional single self-protected DNAzyme walker, the dSPD walker benefits from the secondary amplification and spatial confinement effect and could guide "one" target miRNA to generate "n" secondary targets, thereby initiating "n" nearby walking strands immediately, realizing the initial rate over one-magnitude-order faster than that of the conventional one. Moreover, in the process of relative motion between ac-dSPWPs and TPs, the ac-dSPWPs could cleave multiple substrate strands simultaneously to speed up movement and reduce the derailment rate, as well as combine with successive TPs to facilitate a large amount of continuous signal accumulation, achieving an ultrafast detection of miRNA-221 within 10 min in vitro and high sensitivity with a low detection limit of 0.84 pM. In addition, the DNA nanospheres obtained by the rolling circle amplification reaction can capture the Cy5 fluorescence dispersed in liquids, which achieves the high-contrast imaging of miRNA-221, resulting in further ultrasensitive imaging of miRNA-221 in cancer cells. The proposed strategy has made a bold innovation in the rapid and sensitive detection as well as intracellular imaging of low-abundance biomarkers, offering promising application in early diagnosis and relevant research of cancer and tumors.


Subject(s)
DNA, Catalytic , MicroRNAs , MicroRNAs/analysis , Humans , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Optical Imaging , Limit of Detection , DNA/chemistry , Spectrometry, Fluorescence , Fluorescent Dyes/chemistry , Fluorescence , HeLa Cells
9.
Anal Chem ; 96(24): 10028-10037, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38853671

ABSTRACT

Nucleic acids play a pivotal role in the diagnosis of diseases. However, rapid, cost-efficient, and ultrasensitive identification of nucleic acid targets still represents a significant challenge. Herein, we describe an enzyme-free DNA amplification method capable of achieving accurate and ultrasensitive nucleic acid detection via DNA-templated click ligation chain reaction (DT-CLCR) catalyzed by a heterogeneous nanocatalyst made of Cu2O (hnCu2O). This hnCu2O-DT-CLCR method is built on two cross-amplifying hnCu2O-catalyzed DNA-templated azide-alkyne cycloaddition-driven DNA ligation reactions that boast a fast reaction rate and a high DNA ligation yield in minutes, enabling rapid exponential amplification of specific DNA targets. This newly developed hnCu2O-DT-CLCR-enabled DNA amplification strategy is further integrated with two signal reporting mechanisms to achieve low-cost and easy-to-use biosensors: an electrochemical sensor through the conjugation of a methylene blue redox reporter to a DNA probe used in hnCu2O-DT-CLCR and a colorimetric sensor through the incorporation of the split-to-intact G-quadruplex DNAzyme encoded into hnCu2O-DT-CLCR. Both sensors are able to achieve specific detection of the intended DNA target with a limit of detection at aM ranges, even when challenged in complex biological matrices. The combined hnCu2O-DT-CLCR and sensing strategies offer attractive universal platforms for enzyme-free and yet efficient detection of specific nucleic acid targets.


Subject(s)
Click Chemistry , Copper , DNA , Nucleic Acid Amplification Techniques , Copper/chemistry , DNA/chemistry , Catalysis , Humans , Biosensing Techniques/methods , Limit of Detection , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Azides/chemistry , Colorimetry/methods , Electrochemical Techniques/methods , Cycloaddition Reaction
10.
Talanta ; 276: 126267, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38762976

ABSTRACT

DNA Methyltransferase 1 (DNMT1) serves as a crucial biomarker associated with various diseases and is essential for evaluating DNA methylation levels, diagnosing diseases, and evaluating prognosis. As a result, a convenient, quantitative, and sensitive assay for detecting DNMT1 is in high demand. However, current techniques for DNMT1 detection struggle to balance accuracy, low cost, and high sensitivity, limiting their clinical usefulness. To address this challenge, we have developed a DNMT1 detection method (CAED), which combines aptamer-specific recognition with a highly programmable Entropy-driven catalysis DNA network and is further integrated with the CRISPR-Cas12a system. This innovative approach achieves a detection limit as low as 90.9 fmol/L. To demonstrate the clinical applicability and significance of our CAED method, we successfully measured DNMT1 levels in 10 plasma samples 10 cervical tissue samples. These results underscore the potential of our method as an accurate, affordable, and ultra-sensitive tool for evaluating DNMT1 levels. This innovative method offers a potent means for assessing DNMT1 levels and significantly advances disease diagnosis and health risk prediction. Plus, it establishes an innovative design framework for CRISPR-Cas12a-based biosensors, tailored explicitly for enzyme content quantification.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , CRISPR-Cas Systems , DNA (Cytosine-5-)-Methyltransferase 1 , Entropy , Biosensing Techniques/methods , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Humans , CRISPR-Cas Systems/genetics , Aptamers, Nucleotide/chemistry , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Limit of Detection , Female
11.
Anal Chem ; 96(23): 9666-9675, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38815126

ABSTRACT

Epigenetic modification plays an indispensable role in regulating routine molecular signaling pathways, yet it is rarely used to modulate molecular self-assembly networks. Herein, we constructed a bioorthogonal demethylase-stimulated DNA circuitry (DSC) system for high-fidelity imaging of microRNA (miRNA) in live cells and mice by eliminating undesired off-site signal leakage. The simple and robust DSC system is composed of a primary cell-specific circuitry regulation (CR) module and an ultimate signal-transducing amplifier (SA) module. After the modularly designed DSC system was delivered into target live cells, the DNAzyme of the CR module was site-specifically activated by endogenous demethylase to produce fuel strands for the subsequent miRNA-targeting SA module. Through the on-site and multiply guaranteed molecular recognitions, the lucid yet efficient DSC system realized the reliably amplified in vivo miRNA sensing and enabled the in-depth exploration of the demethylase-involved signal pathway with miRNA in live cells. Our bioorthogonally on-site-activated DSC system represents a universal and versatile biomolecular sensing platform via various demethylase regulations and shows more prospects for more different personalized theragnostics.


Subject(s)
DNA, Catalytic , MicroRNAs , MicroRNAs/analysis , MicroRNAs/metabolism , DNA, Catalytic/metabolism , DNA, Catalytic/chemistry , Animals , Mice , Humans , DNA Methylation , Optical Imaging
12.
Talanta ; 276: 126187, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38733933

ABSTRACT

Gold nanoparticles (Au NPs) have been widely utilized in developing DNAzyme-functionalized nanosensors, most of which were engineered by attaching the thiolated DNAzymes to Au NPs via Au-S bonding. However, the Au NP-DNAzyme nanosensors always suffer from signal distortion when applied in complex environment with abundant thiols, which poses challenge for practical applications. Here, we focus on addressing the root cause of the issue and propose to decorate the Au NPs with a thin layer of platinum, thus facilitating the conjugation of DNAzymes through Pt-S bonding, a thiol-resistant cross-linking. The Pt-S bond stabilized DNAzyme nanosensor effectively minimized false positive signals when detecting l-histidine in infant formulas, as compared to the Au-S stabilized counterpart. This innovative strategy holds promise for high-fidelity biosensing, improving the practical applicability of Au NP-based DNAzyme nanosensor.


Subject(s)
Biosensing Techniques , DNA, Catalytic , Gold , Metal Nanoparticles , Platinum , Sulfhydryl Compounds , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Biosensing Techniques/methods , Platinum/chemistry , Sulfhydryl Compounds/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Histidine/chemistry , Histidine/analysis , Humans
13.
Chem Commun (Camb) ; 60(46): 5976-5979, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38769822

ABSTRACT

Hazardous lead ions (Pb2+) even at a minute level can pose side effects on human health, highlighting the need for tools for trace Pb2+ detection. Herein, we present a DNAzyme-activated CRISPR assay (termed DzCas12T) for sensitive and one-pot detection of lead contamination. Using an extension-bridged strategy eliminates the need for separation to couple the DNAzyme recognition and CRISPR reporting processes. The tandem design endowed the DzCas12T assay with high specificity and sensitivity down to the pM-level. This assay has been used to detect lead contamination in food and water samples, indicating the potential for monitoring lead-associated environmental and food safety.


Subject(s)
CRISPR-Cas Systems , DNA, Catalytic , Lead , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Lead/analysis , CRISPR-Cas Systems/genetics , Biosensing Techniques , Limit of Detection , Food Contamination/analysis , Water Pollutants, Chemical/analysis
14.
Anal Chem ; 96(22): 9244-9253, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38773697

ABSTRACT

Sensitive identification and effective inactivation of the virus are paramount for the early diagnosis and treatment of viral infections to prevent the risk of secondary transmission of viruses in the environment. Herein, we developed a novel two-step fluorescence immunoassay using antibody/streptavidin dual-labeled polystyrene nanobeads and biotin-labeled G-quadruplex/hemin DNAzymes with peroxidase-mimicking activity for sensitive quantitation and efficient inactivation of living Zika virus (ZIKV). The dual-labeled nanobeads can specifically bind ZIKV through E protein targeting and simultaneously accumulate DNAzymes, leading to the catalytic oxidation of Amplex Red indicators and generation of intensified aggregation-induced emission fluorescence signals, with a detection limit down to 66.3 PFU/mL and 100% accuracy. Furthermore, robust reactive oxygen species generated in situ by oxidized Amplex Red upon irradiation can completely kill the virus. This sensitive and efficient detection-inactivation integrated system will expand the viral diagnostic tools and reduce the risk of virus transmission in the environment.


Subject(s)
DNA, Catalytic , Zika Virus , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Immunoassay/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Limit of Detection , G-Quadruplexes , Virus Inactivation/radiation effects , Humans
15.
Anal Chim Acta ; 1311: 342743, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38816160

ABSTRACT

BACKGROUND: MicroRNA (miRNA) emerges as important cancer biomarker, accurate detection of miRNA plays an essential role in clinical sample analysis and disease diagnosis. However, it remains challenging to realize highly sensitive detection of low-abundance miRNA. Traditional detection methods including northern blot and real-time PCR have realized quantitative miRNA detection. However, these detection methods are involved in sophisticated operation and expensive instruments. Therefore, the development of novel sensing platform with high sensitivity and specificity for miRNA detection is urgently demanded for disease diagnosis. RESULTS: In this work, a novel electrochemical biosensor was constructed for miRNA detection based on target-driven cascade amplified assembly of electroactive covalent organic frameworks (COFs) on tetrahedral DNA nanostructure with multiplex recognition domains (m-TDN). COFs were employed as nanocarriers of electroactive prussian blue (PB) molecules by the "freeze-drying-reduction" method without the use of DNA as gatekeeper, which was simple, mild and efficient. The target-triggered catalytic hairpin assembly (CHA) and glutathione reduction could convert low-abundance miRNA into a large amount of Mn2+. Without the addition of exogenous Mn2+, the dynamically-generated Mn2+-powered DNAzyme cleavage process induced abundant PB-COFs probe assembled on the four recognition domains of m-TDN, resulting in significantly signal output. Using miRNA-182-5p as the model target, the proposed electrochemical biosensor achieved ultrasensitive detection of miRNA-182-5p in the range of 10 fM-100 nM with a detection limit of 2.5 fM. SIGNIFICANCE AND NOVELTY: Taking advantages of PB-COFs probe as the enhanced signal labels, the integration of CHA, Mn2+-powered DNAzyme and m-TDN amplification strategy significantly improved the sensitivity and specificity of the biosensor. The designed sensing platform was capable of miRNA detection in complex samples, which provided a new idea for biomarker detection, holding promising potential in clinical diagnosis and disease screening.


Subject(s)
Biosensing Techniques , DNA, Catalytic , DNA , Electrochemical Techniques , Metal-Organic Frameworks , MicroRNAs , Nanostructures , MicroRNAs/analysis , Metal-Organic Frameworks/chemistry , Biosensing Techniques/methods , Nanostructures/chemistry , DNA/chemistry , Humans , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Limit of Detection , Ferrocyanides/chemistry
16.
ACS Nano ; 18(21): 13950-13965, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38751197

ABSTRACT

Manipulating the expression of cellular genes through efficient CRISPR/Cas9 delivery is rapidly evolving into a desirable tumor therapeutics. The exposure of CRISPR/Cas9 to a complex external environment poses challenges for conventional delivery carriers in achieving responsive and accurate release. Here, we report a Trojan horse-like nanocapsule for the on-demand delivery of CRISPR/Cas9 in a microRNA-responsive manner, enabling precise tumor therapy. The nanocapsule comprises a nanoassembled, engineered DNAzyme shell encasing a Cas9/sgRNA complex core. The DNAzyme, functioning as a catalytic unit, undergoes a conformational change in the presence of tumor-associated microRNA, followed by activating a positive feedback-driven autonomous catabolic cycle of the nanocapsule shell. This catabolic cycle is accomplished through chain reactions of DNAzyme "cleavage-hybridization-cleavage", which ensures sensitivity in microRNA recognition and effective release of Cas9/sgRNA. Utilizing this Trojan horse-like nanocapsule, as low as 1.7 pM microRNA-21 can trigger the on-demand release of Cas9/sgRNA, enabling the specific editing of the protumorigenic microRNA coding gene. The resulting upregulation of tumor suppressor genes induces apoptosis in tumor cells, leading to significant inhibition of tumor growth by up to 75.94%. The Trojan horse-like nanocapsule, with superior programmability and biocompatibility, is anticipated to serve as a promising carrier for tailoring responsive gene editing systems, achieving enhanced antitumor specificity and efficacy.


Subject(s)
CRISPR-Cas Systems , DNA, Catalytic , MicroRNAs , Nanocapsules , CRISPR-Cas Systems/genetics , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Humans , Nanocapsules/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Mice , Gene Editing , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/chemistry
17.
Chem Commun (Camb) ; 60(53): 6741-6744, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38809259

ABSTRACT

We reported a colorimetric paper-based device by integrating the modified acid RNA-cleaving DNAzymes (MaRCD-EC1) for highly sensitive (detection limit = 102 CFU mL-1), and rapid (within 30 min) detection of E. coli without amplification. This device exhibited a clinical sensitivity of 100% and a specificity of 100% in identifying E. coli-associated urinary tract infections (UTIs) using the clinical urine samples.


Subject(s)
Colorimetry , DNA, Catalytic , Escherichia coli , Paper , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Escherichia coli/isolation & purification , Urinary Tract Infections/diagnosis , Urinary Tract Infections/microbiology , Urinary Tract Infections/urine , Humans , Limit of Detection , Biosensing Techniques/methods , Escherichia coli Infections/microbiology , Escherichia coli Infections/urine
18.
Nat Commun ; 15(1): 4218, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760331

ABSTRACT

DNAzymes - synthetic enzymes made of DNA - have long attracted attention as RNA-targeting therapeutic agents. Yet, as of now, no DNAzyme-based drug has been approved, partially due to our lacking understanding of their molecular mode of action. In this work we report the solution structure of 8-17 DNAzyme bound to a Zn2+ ion solved through NMR spectroscopy. Surprisingly, it turned out to be very similar to the previously solved Pb2+-bound form (catalytic domain RMSD = 1.28 Å), despite a long-standing literature consensus that Pb2+ recruits a different DNAzyme fold than other metal ion cofactors. Our follow-up NMR investigations in the presence of other ions - Mg2+, Na+, and Pb2+ - suggest that at DNAzyme concentrations used in NMR all these ions induce a similar tertiary fold. Based on these findings, we propose a model for 8-17 DNAzyme interactions with metal ions postulating the existence of only a single catalytically-active structure, yet populated to a different extent depending on the metal ion cofactor. Our results provide structural information on the 8-17 DNAzyme in presence of non-Pb2+ cofactors, including the biologically relevant Mg2+ ion.


Subject(s)
DNA, Catalytic , Lead , Magnesium , Zinc , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Magnesium/metabolism , Magnesium/chemistry , Zinc/metabolism , Zinc/chemistry , Lead/chemistry , Lead/metabolism , Nucleic Acid Conformation , Catalytic Domain , Models, Molecular , Sodium/metabolism , Sodium/chemistry , Metals/metabolism , Metals/chemistry , Magnetic Resonance Spectroscopy , Ions
19.
Nat Commun ; 15(1): 3684, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693181

ABSTRACT

The metal-nucleic acid nanocomposites, first termed metal-nucleic acid frameworks (MNFs) in this work, show extraordinary potential as functional nanomaterials. However, thus far, realized MNFs face limitations including harsh synthesis conditions, instability, and non-targeting. Herein, we discover that longer oligonucleotides can enhance the synthesis efficiency and stability of MNFs by increasing oligonucleotide folding and entanglement probabilities during the reaction. Besides, longer oligonucleotides provide upgraded metal ions binding conditions, facilitating MNFs to load macromolecular protein drugs at room temperature. Furthermore, longer oligonucleotides facilitate functional expansion of nucleotide sequences, enabling disease-targeted MNFs. As a proof-of-concept, we build an interferon regulatory factor-1(IRF-1) loaded Ca2+/(aptamer-deoxyribozyme) MNF to target regulate glucose transporter (GLUT-1) expression in human epidermal growth factor receptor-2 (HER-2) positive gastric cancer cells. This MNF nanodevice disrupts GSH/ROS homeostasis, suppresses DNA repair, and augments ROS-mediated DNA damage therapy, with tumor inhibition rate up to 90%. Our work signifies a significant advancement towards an era of universal MNF application.


Subject(s)
Aptamers, Nucleotide , DNA, Catalytic , Stomach Neoplasms , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Humans , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Cell Line, Tumor , DNA, Catalytic/metabolism , DNA, Catalytic/chemistry , Animals , Receptor, ErbB-2/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Reactive Oxygen Species/metabolism , Mice , DNA Repair , DNA Damage , Glutathione/metabolism , Glutathione/chemistry , Nucleic Acids/metabolism , Nucleic Acids/chemistry
20.
Chem Commun (Camb) ; 60(45): 5848-5851, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38752318

ABSTRACT

A dual-localized DNAzyme walker (dlDW) was constructed by utilizing multiple split DNAzymes with probes, and their substrates are separately localized on streptavidin and AuNPs, serving as walking pedals and tracks, respectively. Based on dlDW, biosensing platform was successfully constructed and showed great potential application in clinical disease diagnosis.


Subject(s)
Biosensing Techniques , DNA, Catalytic , Gold , Streptavidin , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Streptavidin/chemistry , Biosensing Techniques/methods , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Biomarkers/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...