Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.916
Filter
1.
Anal Chem ; 96(23): 9570-9575, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38822787

ABSTRACT

MicroRNA (miRNA) and apurinic/apyrimidinic endonuclease 1 (APE1) have been reported to be closely associated with cancers, making them potential crucial biomarkers and therapeutic targets. However, focusing on the detection of a single target is not conducive to the diagnosis and prognosis assessment of diseases. In this study, an AND logic-gate-based dual-locking hairpin-mediated catalytic hairpin assembly (DL-CHA) was developed for sensitive and specific detection of microRNA and APE1. By addition of a lock to each of the hairpins, with APE1 and microRNA serving as keys, fluorescence signals could only be detected in the presence of simultaneous stimulation by APE1 and miRNA-224. This indicated that the biosensor could operate as an AND logic gate. DL-CHA exhibited advantages such as a low background, rapid response, and high logic capability. Therefore, the biosensor serves as a novel approach to cancer diagnosis with significant potential applications.


Subject(s)
Biosensing Techniques , DNA-(Apurinic or Apyrimidinic Site) Lyase , MicroRNAs , MicroRNAs/analysis , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Humans , Biosensing Techniques/methods , Logic , Limit of Detection
2.
Anal Chim Acta ; 1315: 342822, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38879216

ABSTRACT

In this study, a novel electrochemiluminescence (ECL) biosensor was developed to detect microRNA-21 (miRNA-21) with high sensitivity by leveraging the combined mechanisms of resonance energy transfer (RET) and surface plasmon coupling (SPC). Initially, the glassy carbon electrode (GCE) were coated with Cu-Zn-In-S quantum dots (CZIS QDs), known for their defect-related emission suitable for ECL sensing. Subsequently, a hairpin DNA H3 with gold nanoparticles (Au NPs) attached at the end was modified over the surface of the quantum dots. The Au NPs could effectively quench the ECL signals of CZIS QDs via RET. Further, a significant amount of report DNA was generated through the action of a 3D DNA walker. When the report DNA opened H3-Au NPs, the hairpin structure experienced a conformational change to a linear shape, increasing the gap between the CZIS QDs and the Au NPs. Consequently, the localized surface plasmon resonance ECL (LSPR-ECL) effect replaced ECL resonance energy transfer (ECL-RET). Moreover, the report DNA was released following the addition of H4-Au NPs, resulting in the formation of Au dimers and a surface plasma-coupled ECL (SPC-ECL) effect that enhanced the ECL intensity to 6.97-fold. The integration of new ECL-RET and SPC-ECL biosensor accurately quantified miRNA-21 concentrations from 10-8 M to 10-16 M with a limit of detection (LOD) of 0.08 fM, as well as successfully applied to validate human serum samples.


Subject(s)
Biosensing Techniques , DNA , Electrochemical Techniques , Luminescent Measurements , MicroRNAs , Quantum Dots , Surface Plasmon Resonance , MicroRNAs/analysis , MicroRNAs/blood , Humans , Electrochemical Techniques/methods , Biosensing Techniques/methods , DNA/chemistry , Quantum Dots/chemistry , Surface Plasmon Resonance/methods , Luminescent Measurements/methods , Gold/chemistry , Limit of Detection , Energy Transfer , Metal Nanoparticles/chemistry
3.
Mikrochim Acta ; 191(7): 395, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877347

ABSTRACT

With their regulated Boolean logic operations in vitro and in vivo, DNA logic circuits have shown great promise for target recognition and disease diagnosis. However, significant obstacles must be overcome to improve their operational efficiency and broaden their range of applications. In this study, we propose an Exo III-powered closed-loop DNA circuit (ECDC) architecture that integrates four highly efficient AND logic gates. The ECDC utilizes Exo III as the sole enzyme-activated actuator, simplifying the circuit design and ensuring optimal performance. Moreover, the use of Exo III enables a self-feedback (autocatalytic) mechanism in the dynamic switching between AND logic gates within this circulating logic circuit. After validating the signal flow and examining the impact of each AND logic gate on the regulation of the circuit, we demonstrate the intelligent determination of miR-21 using the carefully designed ECDC architecture in vitro. The proposed ECDC exhibits a linear detection range for miR-21 from 0 to 300 nM, with a limit of detection (LOD) of approximately 0.01 nM, surpassing most reported methods. It also shows excellent selectivity for miR-21 detection and holds potential for identifying and imaging live cancer cells. This study presents a practical and efficient strategy for monitoring various nucleic acid-based biomarkers in vitro and in vivo through specific sequence modifications, offering significant potential for early cancer diagnosis, bioanalysis, and prognostic clinical applications.


Subject(s)
Biosensing Techniques , Exodeoxyribonucleases , Limit of Detection , MicroRNAs , Humans , MicroRNAs/analysis , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , DNA/chemistry
4.
Bioelectrochemistry ; 159: 108753, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38833812

ABSTRACT

MiR-1246 in breast cancer-derived exosomes was a promising biomarker for early diagnosis of breast cancer(BC). However, the low abundance, high homology and complex background interference make the accurate quantitative detection of miR-1246 facing great challenges. In this study, we developed an electrochemical biosensor based on the subtly combined of CRISPR/Cas12a, double-stranded specific nuclease(DSN) and magnetic nanoparticles(MNPs) for the detection of miR-1246 in BC-derived exosomes. Ascribed to the good synergistic effect of DSN, Cas12a and MNPs, the developed electrochemical biosensor exhibited excellent performance with the linear range from 500 aM to 5 pM, and the detection limit as low down to about 50 aM. The target-specific triggered enzyme-digest activity of DSN and Cas12a system, as well as the powerful separation ability of MNPs ensure the high specificity of developed electrochemical biosensor which can distinguish single base mismatches. In addition, the developed electrochemical biosensor has been successfully applied to detect miR-1246 in blood-derived exosomes and realize distinguishing the BC patients from the healthy individuals. It is expected that the well-designed biosensing platform will open up new avenues for clinical liquid biopsy and early screening of breast cancer, as well as provide deeper insights into clinical oncology treatment.


Subject(s)
Biosensing Techniques , Breast Neoplasms , CRISPR-Cas Systems , Electrochemical Techniques , Exosomes , MicroRNAs , Exosomes/chemistry , Exosomes/metabolism , Humans , Biosensing Techniques/methods , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , MicroRNAs/analysis , MicroRNAs/genetics , Female , Electrochemical Techniques/methods , Limit of Detection , Magnetite Nanoparticles/chemistry , Bacterial Proteins , Endodeoxyribonucleases , CRISPR-Associated Proteins
5.
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
6.
Anal Chem ; 96(24): 10084-10091, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38836421

ABSTRACT

Due to the potential off-tumor signal leakage and limited biomarker content, there is an urgent need for stimulus-responsive and amplification-based tumor molecular imaging strategies. Therefore, two tetrahedral framework DNA (tFNA-Hs), tFNA-H1AP, and tFNA-H2, were rationally engineered to form a polymeric tFNA network, termed an intelligent DNA network, in an AND-gated manner. The intelligent DNA network was designed for tumor-specific molecular imaging by leveraging the elevated expression of apurinic/apyrimidinic endonuclease 1 (APE1) in tumor cytoplasm instead of normal cells and the high expression of miRNA-21 in tumor cytoplasm. The activation of tFNA-H1AP can be achieved through specific recognition and cleavage by APE1, targeting the apurinic/apyrimidinic site (AP site) modified within the stem region of hairpin 1 (H1AP). Subsequently, miRNA-21 facilitates the hybridization of activated H1AP on tFNA-H1AP with hairpin 2 (H2) on tFNA-H2, triggering a catalytic hairpin assembly (CHA) reaction that opens the H1AP at the vertices of tFNA-H1AP to bind with H2 at the vertices of tFNA-H2 and generate fluorescence signals. Upon completion of hybridization, miRNA-21 is released, initiating the subsequent cycle of the CHA reaction. The AND-gated intelligent DNA network can achieve specific tumor molecular imaging in vivo and also enables risk stratification of neuroblastoma patients.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA , MicroRNAs , Humans , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , MicroRNAs/metabolism , MicroRNAs/analysis , DNA/chemistry , DNA/metabolism , Molecular Imaging/methods , Animals , Optical Imaging
7.
Anal Chem ; 96(24): 9961-9968, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38838250

ABSTRACT

In this study, a novel europium dual-ligand metal-organic gel (Eu-D-MOGs) with high-efficient anodic annihilation electrochemiluminescence (ECL) was synthesized as an ECL emitter to construct a biosensor for ultrasensitive detection of microRNA-221 (miR-221). Impressively, compared to the ECL signal of europium single-ligand metal-organic gels (Eu-S-MOGs), the ECL signal of Eu-D-MOGs was significantly improved since the two organic ligands could jointly replace the H2O and coordinate with Eu3+, which could remarkably reduce the nonradiative vibrational energy transfer caused by the coordination between H2O and Eu3+ with a high coordination demand. In addition, Eu-D-MOGs could be electrochemically oxidized to Eu-D-MOGs•+ at 1.45 V and reduced to Eu-D-MOGs•- at 0.65 V to achieve effective annihilation of ECL, which overcame the side reaction brought by the remaining emitters at negative potential. This benefited from the annihilation ECL performance of the central ion Eu3+ caused by its redox in the electrochemical process. Furthermore, the annihilation ECL signal of Eu3+ could be improved by sensitizing Eu3+ via the antenna effect. In addition, combined with the improved rolling circle amplification-assisted strand displacement amplification strategy (RCA-SDA), a sensitive biosensor was constructed for the sensitive detection of miR-221 with a low detection limit of 5.12 aM and could be successfully applied for the detection of miR-221 in the lysate of cancer cells. This strategy offered a unique approach to synthesizing metal-organic gels as ECL emitters without a coreactant for the construction of ECL biosensing platforms in biomarker detection and disease diagnosis.


Subject(s)
Electrochemical Techniques , Electrodes , Europium , Gels , Luminescent Measurements , MicroRNAs , Europium/chemistry , MicroRNAs/analysis , Electrochemical Techniques/methods , Ligands , Gels/chemistry , Biosensing Techniques/methods , Limit of Detection , Humans
8.
Anal Chim Acta ; 1314: 342792, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38876514

ABSTRACT

Thyroid cancer is the most prevalent endocrine malignancy. The development of sensitive and reliable methods to detect the thyroid cancer is the currently urgent requirement. Herein, we developed an electrochemiluminescence (ECL) biosensor based on MBene derivative quantum dots (MoB QDs) and Ag NP-on-mirror (NPoM) nanocavity structure. On the one hand, MBene QDs as a novel luminescent material in the ECL process was reported for the first time, which can react with H2O2 as co-reactant. On the other hand, the NPoM nanostructure was successfully constructed with the Ag mirror and Ag NPs to provide highly localized hot spots. The NPoM structure had high degree of light field confinement and electromagnetic field enhancement, which can amplify the ECL signal as the signal modulator. Therefore, the synergistic effect of the nanocavity and localized surface plasmon resonance (LSPR) mode in the NPoM facilitated the enhancement of the ECL signal of MoB QDs over 21.7 times. Subsequently, the proposed ECL biosensing system was employed to analyze the expression level of miRNA-222-3p in the thyroid cancer exosome. The results indicated the relative association between miRNA-222-3p and BRAFV600E mutation. The MoB QDs/NPoM biosensor displayed the ideal potential in assessing thyroid cancer progression for advancing clinical diagnosis applications.


Subject(s)
Exosomes , MicroRNAs , Quantum Dots , Thyroid Neoplasms , MicroRNAs/analysis , Quantum Dots/chemistry , Humans , Exosomes/chemistry , Thyroid Neoplasms/diagnosis , Luminescent Measurements , Electrochemical Techniques , Biosensing Techniques , Silver/chemistry , Metal Nanoparticles/chemistry , Limit of Detection
9.
Mikrochim Acta ; 191(7): 376, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849560

ABSTRACT

CRISPR/Cas system has been widely applied in the assay of disease-related nucleic acids. However, it is still challenging to use CRISPR/Cas system to detect multiple nucleic acids at the same time. Herein, we combined the preponderance of DNA logic circuit, label-free, and CRISPR/Cas technology to construct a label-free "AND" logical gate for multiple microRNAs detection with high specificity and sensitivity. With the simultaneous input of miRNA-155 and miRNA-141, the logic gate starts, and the activation chain of Cas12a is destroyed; thus, the activity is inhibited and the fluorescence of the signal probe ssDNA-AgNCs is turned on. The detection limit of this method for simultaneous quantitative detection of double target is 84 fmol/L (S/N = 3). In this "AND" logic gate, it is only necessary for the design of a simple DNA hairpin probe, which is inexpensive and easy, and since this method involves only one signal output, the data processing is very simple. What is more important, in this strategy two types of microRNAs can be monitored simultaneously by only using CRISPR/Cas12a and a type of crRNA, which offers a new design concept for the exploitation of single CRISPR/Cas system for multiple nucleic acid assays.


Subject(s)
CRISPR-Cas Systems , MicroRNAs , MicroRNAs/analysis , MicroRNAs/genetics , CRISPR-Cas Systems/genetics , Humans , Limit of Detection , CRISPR-Associated Proteins/genetics , Endodeoxyribonucleases/genetics , Metal Nanoparticles/chemistry , Biosensing Techniques/methods , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , Bacterial Proteins/genetics , DNA/genetics , DNA/chemistry
10.
Anal Chem ; 96(22): 9097-9103, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38768044

ABSTRACT

Herein, a fluorescence light-up 3D DNA walker (FLDW) was powered and accelerated by endogenous adenosine-5'-triphosphate (ATP) molecules to construct a biosensor for sensitive and rapid label-free detection and imaging of microRNA-221 (miRNA-221) in malignant tumor cells. Impressively, ATP as the driving force and accelerator for FLDW could significantly accelerate the operation rate of FLDW, reduce the likelihood of errors in signaling, and improve the sensitivity of detection and imaging. When FLDW was initiated by output DNA H1-op transformed by target miRNA-221, G-rich sequences in the S strand, anchored to AuNP, were exposed to form G-quadruplexes (G4s), and thioflavin T (ThT) embedded in the G4s emitted intense fluorescence to realize sensitive and rapid detection of target miRNA-221. Meanwhile, the specific binding of ThT to G4 with a weak background fluorescence response was utilized to enhance the signal-to-noise ratio of the label-free assay straightforwardly and cost-effectively. The proposed FLDW system could realize sensitive detection of the target miRNA-221 in the range of 1 pM to 10 nM with a detection limit of 0.19 pM by employing catalytic hairpin assembly (CHA) to improve the conversion of the target. Furthermore, by harnessing the abundant ATP present in the tumor microenvironment, FLDW achieved rapid and accurate imaging of miRNA-221 in cancer cells. This strategy provides an innovative and high-speed label-free approach for the detection and imaging of biomarkers in cancer cells and is expected to be a powerful tool for bioanalysis, diagnosis, and prognosis of human diseases.


Subject(s)
Adenosine Triphosphate , Biosensing Techniques , DNA , MicroRNAs , MicroRNAs/analysis , MicroRNAs/metabolism , Humans , Adenosine Triphosphate/analysis , Adenosine Triphosphate/metabolism , DNA/chemistry , Biosensing Techniques/methods , Optical Imaging , G-Quadruplexes , Fluorescence , Fluorescent Dyes/chemistry , Limit of Detection , Gold/chemistry
11.
Anal Chem ; 96(22): 9078-9087, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38770734

ABSTRACT

As an important disease biomarker, the development of sensitive detection strategies for miRNA, especially intracellular miRNA imaging strategies, is helpful for early diagnosis of diseases, pathological research, and drug development. Hybridization chain reaction (HCR) is widely used for miRNA imaging analysis because of its high specificity and lack of biological enzymes. However, the classic HCR reaction exhibits linear amplification with low efficiency, limiting its use for the rapid analysis of trace miRNA in living cells. To address this problem, we proposed a toehold-mediated exponential HCR (TEHCR) to achieve highly sensitive and efficient imaging of miRNA in living cells using ß-FeOOH nanoparticles as transfection vectors. The detection limit of TEHCR was as low as 92.7 fM, which was 8.8 × 103 times lower compared to traditional HCR, and it can effectively distinguish single-base mismatch with high specificity. The TEHCR can also effectively distinguish the different expression levels of miRNA in cancer cells and normal cells. Furthermore, TEHCR can be used to construct OR logic gates for dual miRNA analysis without the need for additional probes, demonstrating high flexibility. This method is expected to play an important role in clinical miRNA-related disease diagnosis and drug development as well as to promote the development of logic gates.


Subject(s)
MicroRNAs , Nucleic Acid Hybridization , MicroRNAs/analysis , MicroRNAs/metabolism , Humans , Limit of Detection , Nucleic Acid Amplification Techniques/methods , Ferric Compounds/chemistry
12.
Anal Chem ; 96(22): 8914-8921, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38776971

ABSTRACT

MicroRNAs (miRNAs) are a class of endogenous noncoding small RNAs that play important roles in various biological processes and diseases. Direct determination of miRNAs is a cost-efficient and accurate method for analysis. Herein, we established a novel method for the analysis of miRNAs based on a narrow constant-inner-diameter mass spectrometry emitter. We utilized the gravity-assisted sleeving etching method to prepare a constant-inner-diameter mass spectrometry emitter with a capillary inner diameter of 5.5 µm, coupled it with a high-voltage power supply and a high-resolution mass spectrometer, and used it for miRNA direct detection. The method showed high sensitivity and reproducibility for the analysis of four miRNAs, with a limit of detection of 100 nmol/L (170 amol) for the Hsa-miR-1290 analysis. Compared with commercial ion sources, our method achieved higher sensitivity for miRNA detection. In addition, we analyzed the total miRNAs in the A549 cells. The result indicated that both spiked and endogenous miRNAs could be quantified with high accuracy. As a result, this method offers a promising platform for highly sensitive and accurate miRNA analysis. Furthermore, this approach can be extended to the analysis of other small oligonucleotides and holds the potential for studying clinical samples and facilitating disease diagnosis.


Subject(s)
Mass Spectrometry , MicroRNAs , MicroRNAs/analysis , Humans , A549 Cells , Limit of Detection
13.
Anal Chem ; 96(22): 8973-8980, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38780221

ABSTRACT

Epithelial-mesenchymal transformation (EMT) is one of the important mechanisms of malignancy in endometrial cancer, and detection of EMT targets is a key challenge to explore the mechanism of endometrial carcinoma (EC) malignancy and discover novel therapeutic targets. This study attempts to use surface-enhanced Raman spectroscopy (SERS), a highly sensitive, ultrafast, and highly specific analytical technology, to rapidly detect microRNA-200a-3p and ZEB1 in endometrial cancer cell lines. The silver nanoparticles were decorated with iodine and calcium ions, can capture the SERS fingerprints of microRNA-200a-3p and ZEB1 protein, and effectively avoid the interference of impurity signals. At the same time, the method has high sensitivity for the detection of the above EMT targets, and the lowest detection limits for microRNA-200a-3p and ZEB1 are 4.5 pmol/mL and 10 ng/mL, respectively. At the lowest detection concentration, the method still has high stability. In addition, principal component analysis can not only identify microRNA-200a-3p and ZEB1 protein from a variety of EMT-associated microRNA and proteins but also identify them in the total RNA and total protein of endometrial cancer cell lines and normal endometrial epithelial cell lines. This study modified silver nanoparticles with iodine and calcium ions and for the first time captured the fingerprints of EMT-related targets microRNA-200a-3p and ZEB1 at the same time without label, and the method has high sensitivity and stability. This SERS-based method has immense potential for elucidating the molecular mechanisms of EMT-related EC, as well as identifying biomarkers for malignant degree and prognosis prediction.


Subject(s)
Endometrial Neoplasms , Epithelial-Mesenchymal Transition , Metal Nanoparticles , MicroRNAs , Silver , Spectrum Analysis, Raman , Zinc Finger E-box-Binding Homeobox 1 , Spectrum Analysis, Raman/methods , Humans , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/pathology , Female , MicroRNAs/analysis , MicroRNAs/metabolism , Silver/chemistry , Metal Nanoparticles/chemistry , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cell Line, Tumor , Prognosis , Surface Properties
14.
Article in English | MEDLINE | ID: mdl-38783564

ABSTRACT

MicroRNA (miRNA), crucial non-coding RNAs, have emerged as key biomarkers in molecular diagnostics, prognosis, and personalized medicine due to their significant role in gene expression regulation. Salivary miRNA, in particular, stands out for its non-invasive collection method and ease of accessibility, offering promising avenues for the development of point-of-care diagnostics for a spectrum of diseases, including cancer, neurodegenerative disorders, and infectious diseases. Such development promises rapid and precise diagnosis, enabling timely treatment. Despite significant advancements in salivary miRNA-based testing, challenges persist in the quantification, multiplexing, sensitivity, and specificity, particularly for miRNA at low concentrations in complex biological mixtures. This work delves into these challenges, focusing on the development and application of salivary miRNA tests for point-of-care use. We explore the biogenesis of salivary miRNA and analyze their quantitative expression and their disease relevance in cancer, infection, and neurodegenerative disorders. We also examined recent progress in miRNA extraction, amplification, and multiplexed detection methods. This study offers a comprehensive view of the development of salivary miRNA-based point-of-care testing (POCT). Its successful advancement could revolutionize the early detection, monitoring, and management of various conditions, enhancing healthcare outcomes. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > Diagnostic Nanodevices.


Subject(s)
MicroRNAs , Point-of-Care Testing , Saliva , Humans , MicroRNAs/analysis , MicroRNAs/metabolism , Saliva/chemistry , Saliva/metabolism , Point-of-Care Systems , Neoplasms/diagnosis , Neoplasms/metabolism , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/metabolism
15.
Mikrochim Acta ; 191(6): 351, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806809

ABSTRACT

A target-triggered strand displacement-assisted target recycling based on carbon dots-based fluorescent probe and mesoporous silica nanoparticles@polydopamine (MSNs@PDA) was established to detect miRNA. The surface of MSNs rich in mesopores was coated with a layer of PDA, which can adsorb and quench the fluorescence of single-stranded Fuel DNA with fluorescent carbon dots (CDs) modified at the end through fluorescence resonance energy transfer (FRET). After adding double-stranded DNA-gold nanoparticles (dsDNA-AuNPs) and target let-7a, it will trigger two toehold-mediated strand displacement reactions (TSDR), leading to the recovery of fluorescence and the recycling of target let-7a (excitation wavelength: 380 nm; emission wavelength: 458 nm). The recovery value of fluorescence is proportional to the logarithm of the target microRNA let-7a concentration, thus realizing the sensitivity amplification detection of disease markers. The MSNs@PDA@Fuel DNA-CDs/dsDNA-AuNPs nanoplatform based on the strategy of "on-off-on" and TSDR cyclic amplification may hold great potential as an effective and safe nanoprobe for accurate fluorescence imaging of diseases related to miRNA with low abundances.


Subject(s)
Carbon , Fluorescent Dyes , Gold , Indoles , MicroRNAs , Polymers , Quantum Dots , Silicon Dioxide , MicroRNAs/analysis , Fluorescent Dyes/chemistry , Carbon/chemistry , Humans , Quantum Dots/chemistry , Polymers/chemistry , Gold/chemistry , Silicon Dioxide/chemistry , Indoles/chemistry , Fluorescence Resonance Energy Transfer/methods , Metal Nanoparticles/chemistry , Optical Imaging/methods , Limit of Detection , Porosity , DNA/chemistry
16.
Biosens Bioelectron ; 259: 116402, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38788342

ABSTRACT

In this work, a high-performance conjugated microporous polymer (CMP) decorated with BiOBr (Tr(PhXOD)3-CMP/BiOBr) is synthesized to application in construction of ultrasensitive photoelectrochemical (PEC) biosensor for sensing miRNA-122, by firstly coupling with efficient clip toehold-mediated allosteric bicycle strand displacement (ABSD). Notably, the Tr(PhXOD)3-CMP/BiOBr not only owns self-enhanced D-A-D structure that extremely shortens migration distance of photo-generated electron, but also forms Z-type heterostructure for accelerating electron-hole separation, thereby significantly enhancing the photocurrent with 10-fold higher than commonly used methods. Meanwhile, the clip toehold-mediated ABSD based on ternary linkage structure transformation avoids the attrition of invading strand, endowing the conservation of high concentration for undergoing rapid reaction with high-efficiency DNA amplification, which dramatically improves reaction time and superior target conversion. The experimental results indicate that proposed PEC biosensor had a high sensitivity to miRNA-122 with a detection limit of 0.49 fM, which provides a newly organic/inorganic photosensitive nanomaterials and efficient DNA strand displacement in bioanalytical and early clinical disease diagnosis.


Subject(s)
Biosensing Techniques , Limit of Detection , MicroRNAs , Polymers , Biosensing Techniques/methods , Polymers/chemistry , MicroRNAs/analysis , Electrochemical Techniques/methods , Humans , DNA/chemistry
17.
Biosens Bioelectron ; 259: 116409, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38795495

ABSTRACT

DNA-based molecular amplifiers offer significant promise for molecular-level disease diagnosis and treatment, yet tailoring their activation for precise timing and localization remains a challenge. Herein, we've pioneered a dual activation strategy harnessing external light and internal ATP to create a highly controlled DNA logic amplifier (FDLA) for accurate miRNA monitoring in cancer cells. The FDLA was constructed by tethered the two functionalized catalytic hairpin assembly (CHA) hairpin modules (ATP aptamer sealed hairpin aH1 and photocleavable (PC-linker) sites modified hairpin pH2) to DNA tetrahedron (DTN). The FDLA system incorporates ATP aptamers and PC-linkers as logic control units, allowing them to respond to both exogenous UV light and endogenous ATP present within cancer cells. This response triggers the release of CHA hairpin modules, enabling amplified FRET miRNA imaging through an AND-AND gate. The DTN structure could improve the stability of FDLA and accelerate the kinetics of the strand displacement reaction. It is noteworthy that the UV and ATP co-gated DNA circuit can control the DNA bio-computing at specific time and location, offering spatial and temporal capabilities that can be harnessed for miRNA imaging. Furthermore, the miRNA-sensing FDLA amplifier demonstrates reliable imaging of intracellular miRNA with minimal background noise and false-positive signals. This highlights the feasibility of utilizing both exogenous and endogenous regulatory strategies to achieve spatial and temporal control of DNA molecular circuits within living cancer cells. Such advancements hold immense potential for unraveling the correlation between miRNA and associated diseases.


Subject(s)
Adenosine Triphosphate , Aptamers, Nucleotide , Biosensing Techniques , DNA , MicroRNAs , MicroRNAs/analysis , Humans , Biosensing Techniques/methods , Adenosine Triphosphate/analysis , Aptamers, Nucleotide/chemistry , DNA/chemistry , DNA/genetics , Fluorescence Resonance Energy Transfer/methods , Ultraviolet Rays
18.
Biosens Bioelectron ; 259: 116412, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38795498

ABSTRACT

While there is significant potential for DNA machine-built enzyme-free fluorescence biosensors in the imaging analysis of live biological samples, they persist certain shortcomings. These encompass a deficiency of signal enrichment within a singular interface, uncontrolled premature activation during bio-delivery, and a slow reaction rate due to free nucleic acid collisions. In this contribution, we are committed to resolving the above challenges. Firstly, a single-interface-integrated domino-like driving amplification is constructed. In this conception, a specific target acts as the domino promotor (namely the energy source), initiating a cascading chain reaction that grafts onto a singular interface. Next, an 808 nm near-infrared (NIR) light-excited up-converting luminescence-induced light-activatable biosensing technique is introduced. By locking the target-specific identification segment with a photo-cleavage connector, the up-converted ultraviolet emission can activate target binding in a completely controlled manner. Moreover, a fast reaction rate is achieved by confining nucleic acid collisions within the surface of a DNA wire nano-scaffold, leading to a substantial enhancement in local contact concentration (30.8-fold increase, alongside a 15 times elevation in rate). When a non-coding microRNA (miRNA-221) is positioned as the model low-abundance target for proof-of-concept validation, our intelligent DNA machine demonstrates ultra-high sensitivity (with a limit of detection down to 62.65 fM) and good specificity for this hepatic malignant tumor-associated biomarker in solution detection. Going further, it is worth highlighting that the biosensing system can be employed to carry out high-performance imaging analysis in live bio-samples (ranging from the cellular level to the nude mouse body), thereby propelling the field of DNA machines in disease diagnosis.


Subject(s)
Biosensing Techniques , DNA , Infrared Rays , MicroRNAs , Biosensing Techniques/methods , Humans , DNA/chemistry , DNA/genetics , MicroRNAs/analysis , MicroRNAs/genetics , Animals , Mice , Nucleic Acid Amplification Techniques/methods , Optical Imaging/methods , Nanostructures/chemistry
19.
Biosens Bioelectron ; 259: 116416, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38797033

ABSTRACT

The low abundance, heterogeneous expression, and temporal changes of miRNA in different cellular locations pose significant challenges for both the detection sensitivity of miRNA liquid biopsy and intracellular imaging. In this work, we report an intelligently assembled biosensor based on catalytic hairpin assembly (CHA) and aggregation-induced emission (AIE), named as catalytic hairpin aggregation-induced emission (CHAIE), for the ultrasensitive detection and intracellular imaging of miRNA-155. To achieve such goal, tetraphenylethylene-N3 (TPE-N3) is used as AIE luminogen (AIEgen), while graphene oxide is introduced to quench the fluorescence. When the target miRNA is present, CHA reaction is triggered, causing the AIEgen to self-assemble with the hairpin DNA. This will restrict the intramolecular rotation of the AIEgen and produce a strong AIE fluorescence. Interestingly, CHAIE does not require any enzyme or expensive thermal cycling equipment, and therefore provides a rapid detection. Under optimal conditions, the proposed biosensor can determine miRNA in the concentration range from 2 pM to 200 nM within 30 min, with the detection limit of 0.42 pM. The proposed CHAIE biosensor in this work offers a low background signal and high sensitivity, making it applicable for highly precise spatiotemporal imaging of target miRNA in living cells.


Subject(s)
Biosensing Techniques , Graphite , MicroRNAs , Nanocomposites , Graphite/chemistry , MicroRNAs/analysis , Biosensing Techniques/methods , Humans , Nanocomposites/chemistry , Fluorescent Dyes/chemistry , Limit of Detection , Stilbenes/chemistry , Catalysis , Optical Imaging/methods , Spectrometry, Fluorescence/methods , Fluorescence
20.
ACS Appl Mater Interfaces ; 16(22): 28896-28904, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38770712

ABSTRACT

Herein, we present a novel ultrasensitive graphene field-effect transistor (GFET) biosensor based on lithium niobate (LiNbO3) ferroelectric substrate for the application of breast cancer marker detection. The electrical properties of graphene are varied under the electrostatic field, which is generated through the spontaneous polarization of the ferroelectric substrate. It is demonstrated that the properties of interface between graphene and solution are also altered due to the interaction between the electrostatic field and ions. Compared with the graphene field-effect biosensor based on the conventional Si/SiO2 gate structure, our biosensor achieves a higher sensitivity to 64.7 mV/decade and shows a limit of detection down to 1.7 fM (equivalent to 12 fg·mL-1) on the detection of microRNA21 (a breast cancer marker). This innovative design combining GFETs with ferroelectric substrates holds great promise for developing an ultrahigh-sensitivity biosensing platform based on graphene that enables rapid and early disease diagnosis.


Subject(s)
Biomarkers, Tumor , Biosensing Techniques , Breast Neoplasms , Graphite , MicroRNAs , Niobium , Oxides , Graphite/chemistry , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Humans , Niobium/chemistry , Breast Neoplasms/diagnosis , Oxides/chemistry , MicroRNAs/analysis , Biomarkers, Tumor/analysis , Female , Limit of Detection , Transistors, Electronic
SELECTION OF CITATIONS
SEARCH DETAIL
...