Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 225
Filtrar
1.
Biomed Eng Online ; 23(1): 73, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39061069

RESUMO

BACKGROUND: Minimally invasive glaucoma surgery (MIGS) has experienced a surge in popularity in recent years. Glaucoma micro-stents serve as the foundation for these minimally invasive procedures. Nevertheless, the utilization of these stents still presents certain short-term and long-term complications. This study aims to elucidate the creation of a novel drainage stent implant featuring a diverging channel, produced through microfluidic template processing technology. Additionally, an analysis of the mechanical properties, biocompatibility, and feasibility of implantation is conducted. RESULTS: The stress concentration value of the proposed stent is significantly lower, approximately two to three times smaller, compared to the currently available commercial XEN gel stent. This indicates a stronger resistance to bending in theory. Theoretical calculations further reveal that the initial drainage efficiency of the gradient diverging drainage stent is approximately 5.76 times higher than that of XEN stents. Notably, in vivo experiments conducted at the third month demonstrate a favorable biocompatibility profile without any observed cytotoxicity. Additionally, the drainage stent exhibits excellent material stability in an in vitro simulation environment. CONCLUSIONS: In summary, the diverging drainage stent presents a novel approach to the cost-effective and efficient preparation process of minimally invasive glaucoma surgery (MIGS) devices, offering additional filtering treatment options for glaucoma.


Assuntos
Glaucoma , Stents , Glaucoma/cirurgia , Animais , Microfluídica/instrumentação , Teste de Materiais , Humanos , Procedimentos Cirúrgicos Minimamente Invasivos/instrumentação , Fenômenos Mecânicos , Desenho de Equipamento , Coelhos
2.
Anal Chem ; 96(27): 10962-10968, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38925633

RESUMO

Overexpression of receptor tyrosine kinases (RTKs) or binding to ligands can lead to the formation of specific unliganded and liganded RTK dimers, and these two RTK dimers are potential targets for preventing tumor metastasis. Traditional RTK dimer inhibitor analysis was mostly based on end point assays, which required cumbersome cell handling and behavior monitoring. There are still challenges in developing intuitive process-based analytical methods to study RTK dimer inhibitors, especially those used to visually distinguish between unliganded and liganded RTK dimer inhibitors. Herein, taking the mesenchymal-epithelial transition factor (MET) receptor, an intuitive method for evaluating MET inhibitors has been developed based on atomic force microscopy (AFM) lifetime analysis. The time interval between the start of the force and the bond break point was regarded as the bond lifetime, which could reflect the stability of the MET dimer. The results showed that there was a significant difference in the lifetime (τ) of unliganded MET dimers (τ1 = 207.87 ± 4.69 ms) and liganded MET dimers (τ2 = 330.58 ± 15.60 ms) induced by the hepatocyte growth factor, and aptamer SL1 could decrease τ1 and τ2, suggesting that SL1 could inhibit both unliganded and liganded MET dimers. However, heparin only decreased τ2, suggesting that it could inhibit only the liganded MET dimer. AFM-based lifetime analysis methods could monitor RTK dimer status rather than provide overall average results, allowing for intuitive process-based analysis and evaluation of RTK dimers and related inhibitors at the single-molecule level. This study provides a novel complementary strategy for simple and intuitive RTK inhibitor research.


Assuntos
Microscopia de Força Atômica , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-met , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Humanos , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/metabolismo , Multimerização Proteica/efeitos dos fármacos , Ligantes , Fator de Crescimento de Hepatócito/metabolismo , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/metabolismo
3.
Proc Natl Acad Sci U S A ; 121(15): e2321116121, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38557176

RESUMO

Multidrug resistance (MDR) is a major factor in the failure of many forms of tumor chemotherapy. Development of a specific ligand for MDR-reversal would enhance the intracellular accumulation of therapeutic agents and effectively improve the tumor treatments. Here, an aptamer was screened against a doxorubicin (DOX)-resistant human hepatocellular carcinoma cell line (HepG2/DOX) via cell-based systematic evolution of ligands by exponential enrichment. A 50 nt truncated sequence termed d3 was obtained with high affinity and specificity for HepG2/DOX cells. Multidrug resistance protein 1 (MDR1) is determined to be a possible recognition target of the selected aptamer. Aptamer d3 binding was revealed to block the MDR of the tumor cells and increase the accumulation of intracellular anticancer drugs, including DOX, vincristine, and paclitaxel, which led to a boost to the cell killing of the anticancer drugs and lowering their survival of the tumor cells. The aptamer d3-mediated MDR-reversal for effective chemotherapy was further verified in an in vivo animal model, and combination of aptamer d3 with DOX significantly improved the suppression of tumor growth by treating a xenograft HepG2/DOX tumor in vivo. This work demonstrates the feasibility of a therapeutic DNA aptamer as a tumor MDR-reversal agent, and combination of the selected aptamer with chemotherapeutic drugs shows great potential for liver cancer treatments.


Assuntos
Antineoplásicos , Resistencia a Medicamentos Antineoplásicos , Animais , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Resistência a Múltiplos Medicamentos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Quimioterapia Combinada , Linhagem Celular Tumoral
4.
Mikrochim Acta ; 190(8): 322, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37491600

RESUMO

A simple and wash-free POCT platform based on microcapillary was developed, using breast cancer cell-derived exosomes as a model. This method adopted the "one suction and one extrusion" mode. The hybridized complex of epithelial cell adhesion molecule (EpCAM) aptamer and complementary DNA-horseradish peroxidase conjugate (CDNA-HRP) was pre-modified on the microcapillary's inner surface. "One suction" meant inhaling the sample into the functionalized microcapillary. The exosomes could specifically bind with the EpCAM aptamer on the microcapillary's inner wall, and then the CDNA-HRP complex was released. "One extrusion" referred to squeezing the shedding CDNA-HRP into the 3,3',5,5'-tetramethylbenzidine (TMB)/H2O2 solution, and then the enzyme-catalyzed reaction would occur to make the solution yellow using sulfuric acid as the terminator. Therefore, exosome detection could be realized. The limit of detection was 2.69 × 104 particles mL-1 and the signal value had excellent linearity in the concentration range from 2.75 × 104 to 2.75 × 108 particles⋅mL-1 exosomes. In addition, the wash-free POCT platform also displayed a favorable reproducibility (RSD = 2.9%) in exosome detection. This method could effectively differentiate breast cancer patients from healthy donors. This work provided an easy-to-operate method for detecting cancer-derived exosomes without complex cleaning steps, which is expected to be applied to breast cancer screening.


Assuntos
Neoplasias da Mama , Exossomos , Humanos , Feminino , Neoplasias da Mama/diagnóstico , DNA Complementar/metabolismo , Exossomos/metabolismo , Peróxido de Hidrogênio/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Reprodutibilidade dos Testes , Sucção , Peroxidase do Rábano Silvestre/metabolismo
5.
ACS Appl Mater Interfaces ; 15(22): 27299-27306, 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37235561

RESUMO

Carcinoembryonic antigen (CEA) is a recognized biomarker for lung cancer and can be used for early detection. However, the clinical value of CEA is not fully realized due to the rigorous requirement for high-sensitivity and wide-range detection methods. Field-effect transistor (FET) biosensors, as one of the potentially powerful platforms, may detect CEA with a significantly higher sensitivity than conventional clinical testing equipment, while their sensitivity and detection range for CEA are far below the requirement for early detection. Here, we construct a floating gate FET biosensor to detect CEA based on a semiconducting carbon nanotube (CNT) film combined with an undulating yttrium oxide (Y2O3) dielectric layer as the biosensing interface. Utilizing an undulating biosensing interface, the proposed device showed a wider detection range and optimized sensitivity and detection limit, which benefited from an increase of probe-binding sites on the sensing interface and an increase of electric double-layer capacitance, respectively. The outcomes of analytical studies confirm that the undulating Y2O3 provided the desired biosensing surface for probe immobilization and performance optimization of a CNT-FET biosensor toward CEA including a wide detection range from 1 fg/mL to 1 ng/mL, good linearity, and high sensitivity of 72 ag/mL. More crucially, the sensing platform can function normally in the complicated environment of fetal bovine serum, indicating its great promise for early lung cancer screening.


Assuntos
Técnicas Biossensoriais , Neoplasias Pulmonares , Nanotubos de Carbono , Humanos , Biomarcadores Tumorais , Antígeno Carcinoembrionário , Transistores Eletrônicos , Nanotubos de Carbono/química , Detecção Precoce de Câncer , Neoplasias Pulmonares/diagnóstico , Pulmão , Técnicas Biossensoriais/métodos
6.
Anal Chim Acta ; 1254: 341130, 2023 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-37005015

RESUMO

Exosomes, as a non-invasive biomarker, perform an important role in breast cancer screening and prognosis monitoring. However, establishing a simple, sensitive, and reliable exosome analysis technique remains challenging. Herein, a one-step multiplex analysis electrochemical aptasensor based on a multi-probe recognition strategy was constructed to analyze breast cancer exosomes. HER2-positive breast cancer cell (SK-BR-3) exosomes were selected as the model targets and three aptamers including CD63, HER2 and EpCAM aptamers were used as the capture units. Methylene blue (MB) functionalized HER2 aptamer and ferrocene (Fc) functionalized EpCAM aptamer, which were modified on gold nanoparticles (Au NPs), i.e. MB-HER2-Au NPs and Fc-EpCAM-Au NPs, were used as signal units. When the mixture of target exosomes, MB-HER2-Au NPs and Fc-EpCAM-Au NPs were added on the CD63 aptamer modified gold electrode, two Au NPs modified by MB and Fc could be specifically captured on the electrode by the recognition of three aptamers with target exosomes. Then one-step multiplex analysis of exosomes was achieved by detecting two independent electrochemical signals. This strategy can not only distinguish breast cancer exosomes from other exosomes (including normal exosomes and other tumor exosomes) but also HER2-positive breast cancer exosomes and HER2-negative breast cancer exosomes. Besides, it had high sensitivity and can detect SK-BR-3 exosomes with a concentration as low as 3.4 × 103 particles mL-1. Crucially, this method can be applicable to the examination of exosomes in complicated samples, which is anticipated to afford assistance for the screening and prognosis of breast cancer.


Assuntos
Aptâmeros de Nucleotídeos , Técnicas Biossensoriais , Neoplasias da Mama , Exossomos , Nanopartículas Metálicas , Humanos , Feminino , Neoplasias da Mama/diagnóstico , Ouro , Molécula de Adesão da Célula Epitelial , Técnicas Eletroquímicas/métodos , Técnicas Biossensoriais/métodos
7.
Anal Chim Acta ; 1252: 341043, 2023 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-36935149

RESUMO

Monitoring the dimerization state of the mesenchymal-epithelial transition factor (Met) was essential for in-depth understanding of the tumor signal transduction network. At present, the dimerization activation pathway of Met protein was mainly studied at the macro level, while the research at the single molecule level was far from comprehensive. Herein, the dimerization activation of Met protein's extracellular domain induced by ligand hepatocyte growth factor (HGF) was dynamically studied by single-molecule force spectroscopy. Met protein was immobilized on a biomimetic lipid membrane for ensuring its physiological environment, and then the Met dimers were recognized by bivalent probe which was formed by two Met-binding aptamers. Then the dimeric state of Met protein could be distinguished from monomeric state of Met protein through some parameters, (such as unimodal ratio, bimodal ratio and separation work). The unimodal indicates the occurrence of single molecule binding event, and the bimodal represents the occurrence of double binding event (also represents the presence of Met dimer). Before HGF treatment, most of the Met protein on the lipid membrane was still in the form of monomer, so the unimodal ratio in the force curve was larger (78.8 ± 5.2%), and the bimodal ratio was smaller (17.0 ± 4.1%). After HGF treatment, the unimodal ratio decreased to 54.0 ± 7.4%, and the bimodal ratio increased to 43.2 ± 7.3%. It was due to the formation of dimers after the binding of Met protein on the fluidity lipid membrane with HGF. In addition, the average separation work increased to about 2 times after HGF treatment. Given that studies of Met protein dimerization inhibitors have contributed to the development of more potent and safe inhibitors to significantly inhibit tumor metastasis, the effects of different medicines (including anticoagulant medicines, different antibiotics and anti-cancer medicines) on the dimerization activation of Met protein were then explored by the platform described above. The results showed that anticoagulant medicines heparin and its analogs can significantly inhibit HGF-mediated Met protein activation, while different antibiotics and anticancer medicines had no significant effect on the dimerization of Met protein. This work provided a platform for studying protein dimerization as well as for screening Met protein dimerization inhibitors at the single-molecule level.


Assuntos
Anticoagulantes , Proteínas Proto-Oncogênicas c-met , Multimerização Proteica , Proteínas Proto-Oncogênicas c-met/química , Proteínas Proto-Oncogênicas c-met/metabolismo , Análise Espectral , Lipídeos
8.
Nano Lett ; 23(5): 1801-1809, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36826373

RESUMO

The reprogramming of cell signaling and behavior through the artificial control of cell surface receptor oligomerization shows great promise in biomedical research and cell-based therapy. However, it remains challenging to achieve combinatorial recognition in a complicated environment and logical regulation of receptors for desirable cellular behavior. Herein, we develop a logic-gated DNA nanodevice with responsiveness to multiple environmental inputs for logically controlled assembly of heterogeneous receptors to modulate signaling. The "AND" gate nanodevice uses an i-motif and an ATP-binding aptamer as environmental cue-responsive units, which can successfully implement a logic operation to manipulate receptors on the cell surface. In the presence of both protons and ATP, the DNA nanodevice is activated to selectively assemble MET and CD71, which modulate the HGF/MET signaling, resulting in cytoskeletal reorganization to inhibit cancer cell motility in a tumor-like microenvironment. Our strategy would be highly promising for precision therapeutics, including controlled drug release and cancer treatment.


Assuntos
DNA , Neoplasias , Humanos , DNA/genética , Oligonucleotídeos , Transdução de Sinais , Neoplasias/tratamento farmacológico , Trifosfato de Adenosina , Microambiente Tumoral
9.
Anal Chem ; 95(9): 4529-4535, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36814089

RESUMO

Sensitive detection of miRNA targets in complex biological samples possesses great value in biopsy analysis and disease diagnosis but is still challenging because of low abundance and nonspecific interferences. In this work, self-primer DNA polymerization-propelled stochastic walkers (SWs) were proposed to detect miRNA-24 by combining magnetic microbeads (MMBs) and flow cytometry. The MMBs not only provide a three-dimensional interface for DNA walkers but also facilitate the enrichment and isolation of RNA targets from complex biological samples such as serum. The SWs can be initiated to walk through the entire surface of MMBs and transduce RNA walking into amplified fluorescence signals, with the detection limit of miRNA-24 at 0.95 pM. Moreover, this strategy integrating with flow cytometry was demonstrated to have good specificity with other homologous miRNAs. This platform offers promising applications in RNA biosensing and biomedical diagnostics.


Assuntos
Técnicas Biossensoriais , MicroRNAs , MicroRNAs/análise , Microesferas , Polimerização , Limite de Detecção , DNA/análise , Fenômenos Magnéticos
10.
Small ; 19(12): e2205903, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36638250

RESUMO

DNA cascaded circuits have great potential in detecting low abundance molecules in complex biological environment due to their powerful signal amplification capability and nonenzymatic feature. However, the problem of the cascaded circuits is that the design is relatively complex and the kinetics is slow. Herein, a new design paradigm called catalyst-accelerated circular cascaded circuits is proposed, where the catalyst inlet is implanted and the reaction speed can be adjusted by the catalyst concentration. This new design is very simple and only requires three hairpin probes. Meanwhile, the results of a series of studies demonstrate that the reaction speed can be accelerated and the sensitivity can be also improved. Moreover, endogenous mRNA can also be used as a catalyst to drive the circuits to amplify the detection of target miRNA in live cells and in mice. These catalyst-accelerated circular cascaded circuits can substantially expand the toolbox for intracellular low abundance molecular detection.


Assuntos
Técnicas Biossensoriais , MicroRNAs , Animais , Camundongos , DNA Circular , DNA , MicroRNAs/genética , RNA Mensageiro , Cinética , Técnicas Biossensoriais/métodos , Técnicas de Amplificação de Ácido Nucleico/métodos
11.
Analyst ; 147(22): 5203-5209, 2022 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-36255234

RESUMO

Mesenchymal stem cells (MSCs) mainly found in the bone marrow of adult mammals demonstrate unique capacities of differentiating into multiple cell lineages and undifferentiated MSCs are considered an ideal source of seed cells for cell therapy and tissue engineering. However, MSCs are heterogeneous and not abundant in bone marrow, and there are few specific markers for these cells currently. Therefore, new methods to isolate and characterize MSCs are urgently required. To address the problem, we successfully developed a high-specificity aptamer, called Apt-W2, to specifically recognize mouse bone marrow mesenchymal stem cells (mBMSCs). We synthesized Apt-W2 modified magnetic beads (Apt-W2-MBs) and used them as bait to fish out the MSCs from mouse bone marrow accurately by magnetic-activated cell sorting (MACS). Next, the sorted cells could break free from the Apt-W2-MBs by the competition of C-W2 (complementary strands of Apt-W2). As a result, the sorted cells were intact, and maintained the stem cell phenotype and good proliferative ability. Simultaneously, the sorted cells showed high pluripotency to differentiate into osteoblasts, chondrocytes, and adipocytes. More importantly, the Apt-W2-MB cocktail showed a fine capture performance for MSCs (∼88.33%). This new methodological approach can greatly facilitate MSC isolation efficiently and intactly, thereby enhancing the rate of in vitro differentiation of MSC-derived cells for the emerging field of tissue engineering and regenerative medicine. This new instrumental application of aptamers is an important innovation that achieved both high efficiency and nondestructive cell sorting, opening the door to novel cell sorting approaches.


Assuntos
Aptâmeros de Nucleotídeos , Células-Tronco Mesenquimais , Camundongos , Animais , Medula Óssea , Diferenciação Celular , Células da Medula Óssea , Células Cultivadas , Proliferação de Células , Mamíferos
12.
Nano Lett ; 22(20): 8216-8223, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36194690

RESUMO

Visualizing intracellular microRNA (miRNA) is of great importance for revealing its roles in the development of disease. However, cell membrane barrier, complex intracellular environment and low abundance of target miRNA are three main challenges for efficient imaging of intracellular miRNA. Here, we report a size-controllable and self-assembled DNA nanosphere with ATP-fueled dissociation property for amplified miRNA imaging in live cells and mice. The DNA nanosphere was self-assembled from Y-shaped DNA (Y-DNA) monomers through predesigned base pair hybridization, and the size could be easily controlled by varying the concentration of Y-DNA. Once the nanosphere was internalized into cells, the intracellular specific target miRNA would trigger the cyclic dissociation of the DNA nanosphere driven by ATP, resulting in amplified FRET signal. The programmable DNA nanosphere has been proven to work well for detecting the expression of miRNA in cancer cells and in mice, which demonstrates its fairish cell penetration, stability and sensitivity.


Assuntos
Técnicas Biossensoriais , MicroRNAs , Nanosferas , Camundongos , Animais , DNA/genética , Hibridização de Ácido Nucleico , Trifosfato de Adenosina
13.
Biomaterials ; 288: 121738, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36008186

RESUMO

Despite DNA nanotechnology has spawned a broad variety and taken a giant leap toward cancer theranostic applications over the last decade, the homogeneous DNA nanostructures often suffer from fatal degradation due to their limited stability and specificity. Herein, for the first time, we report a stable DNA tetrahedra-gold nanoclusters (DT/AuNCs) nanohybrid with a self-assembly/programmed disassembly manner for stimuli-responsive tumor imaging and gene-chemo therapy. By utilizing the multifunctional peptides with positive and legumain-specific domains as bioligands, AuNCs were synthesized as signal generators and gate guard attached on the dual-responsive DT, forming the DT/AuNCs with sequential response to legumain-TK1 mRNA & glutathione. The tumorous biomarker of legumain initiated the signal generation relying on the nanosurface energy transfer effect of AuNCs and denudation of DT-Dox (preliminary disassembly). Successively, the dual-responsive DT-Dox administrated a sequential fragmentation along with Dox release in response to the up-regulated glutathione and TK1 mRNA (secondary disassembly), thereby leading to combined gene silencing and chemo-therapy. The results revealed that the DT/AuNCs nanohybrids significantly improved the stability and enhanced the therapeutic efficiency compared to naked DT. Endowing with remarkable stability against biological milieu and site specificity for drug release, our work exhibits a new prospect of fabricating DNA-based nanohybrids for precise tumor theranostics.


Assuntos
Nanopartículas Metálicas , Neoplasias , DNA/química , Glutationa , Ouro/química , Nanopartículas Metálicas/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , RNA Mensageiro/química
14.
Chem Commun (Camb) ; 58(73): 10221-10224, 2022 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-36000965

RESUMO

MicroRNAs (miRNAs) can be used as biomarkers for the diagnosis and therapy of cancers. However, their low abundance and the complex environment in biological samples hinder miRNA detection. A dual amplification strategy based on the bio-barcode technique (BCA) and auto-cycling primer extension (APE) is proposed to detect miRNA targets in complex biological samples. The strategy shows a good sensitivity for miRNA-19a with a detection limit of 50 fM, and can effectively distinguish other similar miRNAs. It provides a new idea to combine nanoparticle-based amplification with nucleic acid-based amplification together for the sensitive detection of nucleic acid targets.


Assuntos
Técnicas Biossensoriais , MicroRNAs , Limite de Detecção , MicroRNAs/genética , Técnicas de Amplificação de Ácido Nucleico/métodos
15.
Theranostics ; 12(7): 3474-3487, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35547767

RESUMO

RATIONALE: Static assembled multivalent DNA nanotheranostics system have encountered some bottleneck problems in cancer imaging and therapy, such as poor penetration and high immunogenicity. Herein, we proposed an acidic tumor microenvironment triggered assembly of activatable multivalent nanodevice, called "three-arm aptamer nanoclaw" (TA-aptNC), assembled from three pH-responsive aptamer-decorated DNA monomers (pH-aptDMs) to facilitate their functions of imaging and therapy. METHODS: The activated TA-aptNC was constructed by acidic microenvironment triggered in situ assembly of three pH-aptDMs. Designer pH-aptDM was established based on the combination of a pH-responsive i-motif switch and an assembly module with a cell membrane anchoring aptamer ligand. Acidic microenvironment-triggered the assembly of the TA-aptNC was characterized by electrophoresis and atomic force microscopic (AFM). The binding affinity and stability of the TA-aptNC, comparing the monovalent pH-aptDM, were studied via the flow cytometry and nuclease resistance assays. Acidic microenvironment-activated contrast-enhanced tumor imaging and significantly antitumor efficiency were evaluated in vitro and in vivo. Results: At physiological pH environment, the pH-aptDMs with excellent tissue permeability exited as inactivated and monodispersed small monomer. When encountering acidic microenvironment at the tumor site, pH-responsive i-motif switch liberated from the pH-aptDMs, and the three unconstrained DNA modules (DM1, DM2 and DM3) subsequently assembled in situ into the TA-aptNC. Compared with monovalent pH-aptDMs, the spontaneously formed activatable TA-aptNC afforded 2-fold enhanced binding ability via the multivalent effect, which further facilitated the selective tumor cell uptake capability, thus enabling a contrast-enhanced tumor imaging and significantly antitumor efficiency in vivo without systemic toxicity. Conclusions: The proposed strategy offers valuable insight into excavating an endogenous stimuli-triggered assembly of multivalent nanodevice for accurate diagnosis and efficient tumor therapy.


Assuntos
Aptâmeros de Nucleotídeos , Microambiente Tumoral , Aptâmeros de Nucleotídeos/química , Linhagem Celular Tumoral , DNA , Diagnóstico por Imagem , Concentração de Íons de Hidrogênio
16.
Anal Chem ; 94(15): 5937-5945, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35380798

RESUMO

The desire for a cancer theranostic system with simultaneously accurate diagnosis and efficient therapy is undeniably interminable. Heretofore, theranostic systems with simple components were designed for cancer theranostics but with confined accuracy of diagnosis and side effects of administered drugs. Here, we report an activatable theranostic system for simultaneously imaging dual cancer-related RNAs, mRNA Bcl-2 and piRNA-36026, and combined gene-chemotherapy through the target-induced intracellular disassembly of DNA tetrahedron. Briefly, five customized oligonucleotides are used to assemble the functionalized DNA tetrahedron. The relevant functional nucleic acids, including the antisequence of mRNA Bcl-2, the antisequence of piRNA-36026, and aptamer AS1411, are designed in the customized oligonucleotides with the signal reporters Cy3 and Cy5. Doxorubicin (DOX) is loaded in the functionalized DNA tetrahedron by inlaying between cytosine and guanine to form the activatable cancer theranostic system. The activatable cancer theranostic system is able to recognize MCF-7 cells by aptamer AS1411 and then enter the cells. In the presence of targets, the antisequences in the activatable cancer theranostic system hybridize with intracellular mRNA Bcl-2 and piRNA-36026, leading to the fluorescence signal recovery of Cy3 and Cy5 and the downregulation of two targets in the cytoplasm as well as the consequent apoptosis of MCF-7 cells in the form of gene therapy. Interestingly, as the antisequences are designed in the assembly strands, the hybridization between targets and the antisequences results in the disassembly of the activatable cancer theranostic system and the release of DOX as well as sequential chemotherapy. Advantageously, the activatable cancer theranostic system can achieve imaging of dual cancer-related RNAs with an imaging time window as long as 15 h and exhibit an obvious therapeutic effect in vivo. Therefore, this work is in furtherance of exploration for activatable cancer theranostic systems with high accuracy and efficiency and sheds new light on the development of precision medicine.


Assuntos
Neoplasias , Nanomedicina Teranóstica , DNA , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro , RNA Interferente Pequeno , Nanomedicina Teranóstica/métodos
17.
Angew Chem Int Ed Engl ; 61(19): e202116932, 2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35199894

RESUMO

Sensitive and accurate analysis of low-concentration of tumor-derived exosomes (Exos) in biofluids is essential for noninvasive cancer diagnosis but is still challenging due to the lack of high-sensitive methods with low-cost and easy-operation. Herein, exploiting target Exos as a three-dimensional (3D) track for the first time, we developed a self-serviced-track DNA walker (STDW) for wash-free detection of tumor Exos using exosomal glycoprotein, which was enabled by split aptamer-recognition-initiated autonomous running powered by a catalytic hairpin assembly (CHA). Benefiting from high selectivity and sensitivity of the STDW assay, direct detection of tumor Exos in cell culture medium and serum could also be realized. Furthermore, this method exhibited high accuracy in clinical sample analysis, offering the potential for early cancer diagnosis and postoperative response prediction.


Assuntos
Técnicas Biossensoriais , Exossomos , Neoplasias , DNA/genética , Glicoproteínas , Humanos , Neoplasias/diagnóstico
18.
Chem Commun (Camb) ; 58(9): 1414-1417, 2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-34994763

RESUMO

A novel FRET-based dendritic hybridization chain reaction (D-HCR) for TK1 mRNA imaging in living cells was developed. Compared with traditional complex D-HCR methods, it includes the advantages of having a simple design, an accurate signal and is suitable for use with living cells.


Assuntos
Corantes Fluorescentes/química , Nanoestruturas/química , RNA Mensageiro/química , Transferência Ressonante de Energia de Fluorescência , Células Hep G2 , Humanos , Limite de Detecção , Células MCF-7 , Hibridização de Ácido Nucleico , Imagem Óptica
19.
Anal Chem ; 94(5): 2502-2509, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35089704

RESUMO

DNA logic gates, as a class of smart molecular devices with excellent biocompatibility and convenient information processing mode, have been widely used for identification of cancer cells based on logic analysis of cancer biomarkers. However, most of the developed DNA logic gates for identification of cancer cells are mainly driven by homogeneous biomarkers such as membrane proteins or RNAs, which may suffer from insufficient accuracy. Herein, we reported a membrane protein and extracellular acid heterogeneity-driven amplified DNA logic gate (HDLG) for accurate and sensitive identification of cancer cells by combining the superior signal amplification characteristics of the hybridization chain reaction (HCR) and the precise computation ability of the logic operation. In this strategy, a DNA aptamer was employed for membrane protein recognition, and a split i-motif was used for the response of the extracellular acid. Only when the two heterogeneous biomarkers existed simultaneously, the DNA logic gate could be driven to perform the "AND" logic operation and induce the formation of an intact trigger to initiate a HCR process on the cell surface, generating an amplified "ON" fluorescence signal. Benefiting from the design of heterogeneity-driven and signal amplification, this DNA logic gate could not only autonomously perform high-resolution fluorescence imaging on the surface of target cancer cells, but also perform sensitive analysis of target cancer cells with a cell number of 70 detected in 200 µL of buffer and desirable accuracy in differentiating target cancer cells from complicated cell mixtures. We anticipate that this novel HDLG is expected to be applied in precise disease diagnosis.


Assuntos
Aptâmeros de Nucleotídeos , Computadores Moleculares , DNA , Proteínas de Membrana , Neoplasias , Aptâmeros de Nucleotídeos/metabolismo , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , DNA/genética , DNA/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neoplasias/diagnóstico , Neoplasias/genética
20.
Nucleic Acids Res ; 50(7): e40, 2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-34935962

RESUMO

There is considerable interest in creating a precise and sensitive strategy for in situ visualizing and profiling intracellular miRNA. Present here is a novel photocaged amplified FRET nanoflare (PAFN), which spatiotemporal controls of mRNA-powered nanomachine for precise and sensitive miRNA imaging in live cells. The PAFN could be activated remotely by light, be triggered by specific low-abundance miRNA and fueled by high-abundance mRNA. It offers high spatiotemporal control over the initial activity of nanomachine at desirable time and site, and a 'one-to-more' ratiometric signal amplification model. The PAFN, an unprecedented design, is quiescent during the delivery process. However, upon reaching the interest tumor site, it can be selectively activated by light, and then be triggered by specific miRNA, avoiding undesirable early activation and reducing nonspecific signals, allowing precise and sensitive detection of specific miRNA in live cells. This strategy may open new avenues for creating spatiotemporally controllable and endogenous molecule-powered nanomachine, facilitating application at biological and medical imaging.


Assuntos
Técnicas Biossensoriais , MicroRNAs , Diagnóstico por Imagem , Transferência Ressonante de Energia de Fluorescência , MicroRNAs/genética , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA