Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 62(39): e202306691, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37455257

RESUMO

Life-like hierarchical architecture shows great potential for advancing intelligent biosensing, but modular expansion of its sensitivity and functionality remains a challenge. Drawing inspiration from intracellular liquid-liquid phase separation, we discovered that a DNA-encoded artificial cell with a liquid core (LAC) can enhance peroxidase-like activity of Hemin and its DNA G-quadruplex aptamer complex (DGAH) without substrate-selectivity, unlike its gelled core (GAC) counterpart. The LAC is easily engineered as an ultrasensitive biosensing system, benefiting from DNA's high programmability and unique signal amplification capability mediated by liquid-liquid phase separation. As proof of concept, its versatility was successfully demonstrated by coupling with two molecular recognition elements to monitor tumor-related microRNA and profile cancer cell phenotypes. This scalable design philosophy offers new insights into the design of next generation of artificial cells-based biosensors.


Assuntos
Aptâmeros de Nucleotídeos , Células Artificiais , Técnicas Biossensoriais , DNA Catalítico , Quadruplex G , MicroRNAs , Neoplasias , Humanos , DNA/genética , Hemina , DNA Catalítico/metabolismo
2.
ACS Nano ; 17(14): 13430-13440, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37410377

RESUMO

Although many smart probes for precise tumor recognition have been reported, the challenge of "on-target, off-tumor" remains. Therefore, we herein report the fabrication of a series of allosterically tunable DNA nanosensing-circles (NSCs). The recognition affinity of NSCs is programmed through sensitivity to tumor microenvironment (TME) hallmarks such as small molecules, acidity, or oncoproteins. Because of their special programming conditions and active targeting capabilities, NSCs can overcome the obstacles noted above, thus achieving precise tumor recognition. Results from in vitro analysis demonstrated that NSCs obtain their recognition ability through allosteric regulation after sensing TME hallmarks. Furthermore, in vivo imaging indicated that NSCs enable precise tumor imaging. These results demonstrate that our NSCs will be promising tools for precise tumor imaging and therapy.


Assuntos
Neoplasias , Humanos , Microambiente Tumoral
3.
Angew Chem Int Ed Engl ; 62(39): e202307656, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37423897

RESUMO

Nongenetic strategies that enable control over the cell-cell interaction network would be highly desired, particularly in T cell-based cancer immunotherapy. In this work, we developed an aptamer-functionalized DNA circuit to modulate the interaction between T cells and cancer cells. This DNA circuit was composed of recognition-then-triggering and aggregation-then-activation modules. Upon recognizing target cancer cells, the triggering strand was released to induce aggregation of immune receptors on the T cell surface, leading to an enhancement of T cell activity for effective cancer eradication. Our results demonstrated the feasibility of this DNA circuit for promoting target cancer cell-directed stimulation of T cells, which, consequently, enhanced their killing effect on cancer cells. This DNA circuit, as a modular strategy to modulate intercellular interactions, could lead to a new paradigm for the development of nongenetic T cell-based immunotherapy.


Assuntos
Aptâmeros de Nucleotídeos , Neoplasias , Linfócitos T/metabolismo , Aptâmeros de Nucleotídeos/metabolismo , DNA/metabolismo , Membrana Celular/metabolismo , Imunoterapia , Neoplasias/terapia , Neoplasias/metabolismo
4.
Angew Chem Int Ed Engl ; 62(23): e202302525, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-36930411

RESUMO

Carbon monoxide (CO) is an endogenous signaling molecule with broad therapeutic effects. Here, a multifunctional X-ray-triggered carbon monoxide (CO) and manganese dioxide (MnO2 ) generation nanoplatform based on metal carbonyl and scintillating nanoparticles (SCNPs) is reported. Attributed to the radioluminescent characteristic of SCNPs, UV-responsive Mn2 (CO)10 is not only indirectly activated to release CO by X-ray but can also be degraded into MnO2 . A high dose of CO can be used as a glycolytic inhibitor for tumor suppression; it will also sensitize tumor cells to radiotherapy. Meanwhile MnO2 , as the photolytic byproduct of Mn2 (CO)10 , has both glutathione (GSH) depletion and Fenton-like Mn2+ delivery properties to produce highly toxic hydroxyl radical (⋅OH) in tumors. Thus, this strategy can realize X-ray-activated CO release, GSH depletion, and ⋅OH generation for cascade cancer radiosensitization. Furthermore, X-ray-activated Mn2+ in vivo demonstrates an MRI contrast effect, making it a potential theranostic nanoplatform.


Assuntos
Nanopartículas , Neoplasias , Humanos , Compostos de Manganês/farmacologia , Compostos de Manganês/uso terapêutico , Óxidos/farmacologia , Monóxido de Carbono/farmacologia , Monóxido de Carbono/uso terapêutico , Raios X , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Linhagem Celular Tumoral , Glutationa/metabolismo , Peróxido de Hidrogênio/uso terapêutico
5.
ACS Nano ; 17(7): 6150-6164, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36942868

RESUMO

The selective removal of misfolded, aggregated, or aberrantly overexpressed protein plays an essential role in maintaining protein-dominated biological processes. In parallel, the precise knockout of abnormal proteins is inseparable from the accurate identification of proteins within complex environments. Guided by these precepts, small molecules, or antibodies, are commonly used as protein recognition tools for developing targeted protein degradation (TPD) technology. Indeed, TPD has shown tremendous prospects in chronic diseases, rare diseases, cancer research, and other fields. Meanwhile, aptamers are short RNA or DNA oligonucleotides that can bind to target proteins with high specificity and strong affinity. Accordingly, aptamers are actively used in designing and constructing TPD technology. In this perspective, we provide a brief introduction to TPD technology in its current progress, and we summarize its application challenges. Recent advances in aptamer-based TPD technology are reviewed, together with corresponding challenges and outlooks.


Assuntos
Aptâmeros de Nucleotídeos , Proteólise , RNA/metabolismo , DNA/metabolismo , Proteínas/metabolismo
6.
Anal Chem ; 95(2): 1446-1453, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36577081

RESUMO

An aptamer-based field-effect transistor (Apta-FET) is a well-developed assay method with high selectivity and sensitivity. Due to the limited information density that natural nucleotide library holds, the Apta-FET faces fundamental restriction in universality to detect various types of analytes. Herein, we demonstrate a type of Apta-FET sensors based on an artificial nucleotide aptamer (AN-Apta-FET). The introduction of an artificial nucleotide increases the diversity of the potential aptamer structure and expands the analyte category of the Apta-FET. The AN-Apta-FET specifically detects hepatoma exosomes, which traditional Apta-FET fails to discriminate from other tumor-derived exosomes, with a limit of detection down to 242 particles mL-1. The AN-Apta-FET distinguishes serum samples of hepatocellular carcinoma patients within 9 min from those of healthy people, showing the potential as a comprehensive assay tool in future disease diagnosis.


Assuntos
Aptâmeros de Nucleotídeos , Técnicas Biossensoriais , Carcinoma Hepatocelular , Exossomos , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/diagnóstico , Aptâmeros de Nucleotídeos/química , Técnicas Biossensoriais/métodos , Neoplasias Hepáticas/diagnóstico
7.
ACS Nano ; 16(12): 21129-21138, 2022 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-36484532

RESUMO

Cytotoxic T cells initiate antitumor effects mainly through direct interactions with tumor cells. As a counter to this, tumor cells can put the brakes on such T-cell activity via specific linkage between programmed death ligand 1 (PDL1) and its receptor programmed cell death protein 1 (PD1). Bispecific inhibitors that enabled synchronous blockade of PD1 and PDL1, thereby releasing the brakes on T-cell antitumor activity, should significantly improve the efficacy of immune checkpoint blockade (ICB) therapy. In this work, we identified a DNA aptamer, Ap3, that could specifically recognize PDL1 on tumor cells and competed with the binding of PD1. By integrating Ap3 with an anti-PD1 aptamer, the bispecific aptamer Ap3-7c was constructed, and it showed promise for improving the T-cell immune response. We further designed a dibenzocyclooctyne (DBCO)-labeled bispecific aptamer, D-Ap3-7c, allowing covalent conjugation of aptamers onto PD1 and PDL1 after specific cell recognition. Our in vivo studies showed that this recognition-then-conjugation strategy could induce a potent immunological effect against tumors. This work is expected to provide clues for antitumor immunotherapy.


Assuntos
Neoplasias , Receptor de Morte Celular Programada 1 , Humanos , Neoplasias/terapia , Antígeno B7-H1 , Imunoterapia
8.
Nano Lett ; 22(23): 9441-9449, 2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36442508

RESUMO

Approaches to DNA probe-mediated precision medicine have been extensively explored for the diagnosis and treatment of diverse types of cancer. Despite this, simple nanoscale devices with the required recognition specificity and sensitivity for clinical application have remained elusive until now. Here, we report a pH-driven covalent nanoscale device that integrates pH-responsive, switchable structure and proximity-driven covalent cross-linking. A tumor acidic, pH-driven mechanism eliminates "on-target, off-tumor" nonspecific recognition. By manipulating covalent binding to target molecule on the cell surface, this nanodevice avoids binding-then-shedding to improve the sensitivity of tumor recognition. We envision that this pH-driven covalent nanoscale device will inspire more clinical applications toward specific, long-term tumor imaging in the cancer microenvironment.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Microambiente Tumoral , Diagnóstico por Imagem , Concentração de Íons de Hidrogênio
9.
Angew Chem Int Ed Engl ; 61(31): e202201220, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35536294

RESUMO

Aptamers are emerging as promising molecular tools in cancer-targeted theranostics. Improving their in vivo stability has been a critical issue in promoting clinical translation, but such efforts could lead to more serious side effects resulting from prolonged retention in healthy organs. To address this problem, we developed an environment-responsive stabilization strategy for the selective enhancement of aptamer biostability in the tumor microenvironment (TME). Briefly, by means of the end extension of an ATP-responsive protection (ARP) module, the designed aptamer could be protected from nuclease degradation through the specific incorporation of ATP. Based on our in vivo results, this ARP-aptamer probe was effectively accumulated in tumors via aptamer-based molecular recognition. It showed selectively prolonged tumor retention time, but rapid digestion in healthy organs. Our strategy should provide a new paradigm for the development of organ-specific nucleic acid-based imaging and therapeutic agents.


Assuntos
Aptâmeros de Nucleotídeos , Neoplasias , Ácidos Nucleicos , Trifosfato de Adenosina , Humanos , Neoplasias/tratamento farmacológico , Microambiente Tumoral
10.
Methods Mol Biol ; 2504: 3-20, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35467275

RESUMO

Cancerous exosomes that carry multiple biomarkers are attractive targets for the early diagnosis and therapy of cancer. As one of the powerful molecular recognition tools, aptamers with excellent binding affinity and specificity toward biomarkers have been exploited to construct various aptamer-based biosensors (aptasensors) for exosome detection. Here, we review recent advances in aptasensors for the detection of cancerous exosomes. We first discuss the importance and potential of cancerous exosomes in cancer diagnosis and then summarize some conventional aptasensors from the perspective of biomarker recognition and signal collection strategies. Finally, we comment on the outlook for aptasensor research and new directions for cancerous exosome detection.


Assuntos
Aptâmeros de Nucleotídeos , Técnicas Biossensoriais , Exossomos , Neoplasias , Aptâmeros de Nucleotídeos/química , Biomarcadores/metabolismo , Exossomos/metabolismo , Humanos , Neoplasias/diagnóstico , Neoplasias/metabolismo
11.
ACS Appl Mater Interfaces ; 13(8): 9542-9560, 2021 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-33595277

RESUMO

In the past decades, various nanomaterials with unique properties have been explored for bioapplications. Meanwhile, aptamers, generated from the systematic evolution of ligands by exponential enrichment technology, are becoming an indispensable element in the design of functional nanomaterials because of their small size, high stability, and convenient modification, especially endowing nanomaterials with recognition capability to specific targets. Therefore, the incorporation of aptamers into nanomaterials offers an unprecedented opportunity in the research fields of diagnostics and therapeutics. Here, we focus on recent advances in aptamer-embedded nanomaterials for bioapplications. First, we briefly introduce the properties of nanomaterials that can be functionalized with aptamers. Then, the applications of aptamer-embedded nanomaterials in cellular analysis, imaging, targeted drug delivery, gene editing, and cancer diagnosis/therapy are discussed. Finally, we provide some perspectives on the challenges and opportunities that have arisen from this promising area.


Assuntos
Aptâmeros de Nucleotídeos/química , Ácidos Nucleicos Imobilizados/química , Nanopartículas Metálicas/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Sequência de Bases , Portadores de Fármacos/química , Humanos , Hidrogéis/química , Lipossomos/química , Estruturas Metalorgânicas/química , Micelas , Nanoporos
12.
Bioconjug Chem ; 31(5): 1289-1294, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32223180

RESUMO

Bispecific aptamer-drug conjugates (BsApDC) may improve the efficacy of drugs by enhancing cellular internalization and targeted delivery. Nevertheless, the synthesis of single-molecular BsApDC has not yet been reported, and it could be thwarted by synthetic challenges. Herein we report a general approach to synthesize a BsApDC hybridized chemical and biological method. Primers incorporated with 5-Fluorouracil (5-FU), 10-Hydroxycamptothecin, and Maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl-monomethyl auristatin E(vcMMAE) were prepared by chemical synthesis, which were converted to corresponding ApDCs efficiently by enzymatic reaction. Biological studies revealed that BsApDC binds with target cells with enhanced internalization and better inhibitory activity, demonstrating the potential of BsApDCs for targeted tumor therapy.


Assuntos
Aptâmeros de Nucleotídeos/química , Preparações Farmacêuticas/química , Aminobenzoatos/química , Camptotecina/análogos & derivados , Camptotecina/química , Linhagem Celular Tumoral , Fluoruracila/química , Humanos , Oligopeptídeos/química
13.
Nat Commun ; 11(1): 1347, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165631

RESUMO

Protein-dominant cellular processes cannot be fully decoded without precise manipulation of their activity and localization in living cells. Advances in optogenetics have allowed spatiotemporal control over cellular proteins with molecular specificity; however, these methods require recombinant expression of fusion proteins, possibly leading to conflicting results. Instead of modifying proteins of interest, in this work, we focus on design of a tunable recognition unit and develop an aptamer-based near-infrared (NIR) light-responsive nanoplatform for manipulating the subcellular localization of specific proteins in their native states. Our results demonstrate that this nanoplatform allows photocontrol over the cytoplasmic-nuclear shuttling behavior of the target RelA protein (a member of the NF-κß family), enabling regulation of RelA-related signaling pathways. With a modular design, this aptamer-based nanoplatform can be readily extended for the manipulation of different proteins (e.g., lysozyme and p53), holding great potential to develop a variety of label-free protein photoregulation strategies for studying complex biological events.


Assuntos
Aptâmeros de Nucleotídeos/genética , Fator de Transcrição RelA/metabolismo , Aptâmeros de Nucleotídeos/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Optogenética , Transporte Proteico , Transdução de Sinais , Fator de Transcrição RelA/genética
14.
ACS Appl Mater Interfaces ; 12(5): 5403-5412, 2020 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31916740

RESUMO

The deficiency of reactive oxygen species (ROS) is the main reason for the current poor efficiency of tumor photodynamic therapy (PDT). To solve this problem, a simple light-triggered core-satellite nanoplatform (UPSD@Au) has been developed by loading Au nanoparticles on the surface of mesoporous silica-coated upconversion nanoparticles. Small molecules DC50 (C17H14BrF2N3OS) and photosensitizer (silicon phthalocyanine dihydroxide, SPCD) were loaded into the silica shell to improve ROS production. Meanwhile, PDT can be triggered through facile near-infrared laser irradiation given the occurrence of a moderate photothermal transfer process between upconversion nanoparticles and Au. The reasonable increment in temperature induced by Au resulted in the timely release of DC50. The inhibition of copper transfer by DC50 results in reduced ROS scavenging and thus improves light-triggered ROS accumulation. Notably, the expression levels of the human copper-trafficking proteins Atox1 and CCS in cancerous cells exceed those in normal cells, and thus enhanced ROS accumulation effect was achieved in cancerous cells. In vitro and in vivo results demonstrate that the synergism between DC50 and SPCD coloaded in the UPSD@Au nanoplatform increases the efficiency of PDT. The UPSD@Au platform represents an efficient codelivery method for hydrophobic small molecules and improves sensitization to specific cancer therapy.


Assuntos
Antineoplásicos , Nanopartículas , Fotoquimioterapia , Fármacos Fotossensibilizantes , Espécies Reativas de Oxigênio/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Cobre/metabolismo , Sistemas de Liberação de Medicamentos , Células HEK293 , Humanos , Indóis , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/toxicidade , Compostos de Organossilício , Oxirredução , Fármacos Fotossensibilizantes/química
15.
Angew Chem Int Ed Engl ; 59(2): 663-668, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31650689

RESUMO

Expanding the number of nucleotides in DNA increases the information density of functional DNA molecules, creating nanoassemblies that cannot be invaded by natural DNA/RNA in complex biological systems. Here, we show how six-letter GACTZP DNA contributes this property in two parts of a nanoassembly: 1) in an aptamer evolved from a six-letter DNA library to selectively bind liver cancer cells; and 2) in a six-letter self-assembling GACTZP nanotrain that carries the drug doxorubicin. The aptamer-nanotrain assembly, charged with doxorubicin, selectively kills liver cancer cells in culture, as the selectivity of the aptamer binding directs doxorubicin into the aptamer-targeted cells. The assembly does not kill untransformed cells that the aptamer does not bind. This architecture, built with an expanded genetic alphabet, is reminiscent of antibodies conjugated to drugs, which presumably act by this mechanism as well, but with the antibody replaced by an aptamer.


Assuntos
Aptâmeros de Nucleotídeos/química , DNA/química , Doxorrubicina/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Humanos , Neoplasias Hepáticas/genética
16.
J Am Chem Soc ; 142(1): 382-391, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31801020

RESUMO

The inner region of solid tumors is found to be high-pressure, hypoxic, and immunosuppressive, providing a breeding ground for tumor aggressiveness and metastasis. While intratumoral accumulation of nanomedicines combined with immunomodulation would significantly enhance therapeutic efficacy, such potential is challenged by the compressed environment and distinct heterogeneity of the tumor bulk. By using an apoptotic body (AB) as the carrier, we develop an effective and universal intratumoral nanomedicine delivery system for the long-lasting remission of tumors. Our results show that the AB-encapsulated nanomedicine (using CpG immunoadjuvant-modified gold-silver nanorods as a model), after intravenous injection, can be specifically phagocytosed by inflammatory Ly-6C+ monocytes, which then actively infiltrate the tumor center via their natural tumor-homing tendency. With the integration of AB-facilitated intratumoral accumulation, the nanorod-based photothermal effect, and CpG-promoted immunostimulation, this cell-mediated delivery system can not only efficiently ablate primary tumors but also elicit a potent immunity to prevent tumors from metastasizing and recurring.


Assuntos
Macrófagos/metabolismo , Monócitos/metabolismo , Nanomedicina , Neoplasias/terapia , Adjuvantes Imunológicos/administração & dosagem , Animais , Sistemas de Liberação de Medicamentos , Ouro/química , Humanos , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/prevenção & controle , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias/metabolismo , Neoplasias/patologia , Fototerapia/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Sci China Chem ; 61(4): 497-504, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30271427

RESUMO

Drug administration customized to individual cells could intrinsically address cancer heterogeneity and provide a safe and effective method for delivering personalized treatment. To accomplish this, we developed a smart nanodrug delivery system characterized by cancer cell-targeted drug delivery and intracellular biomarker-responsive drug activation. This system was composed of a long-nicked DNA duplex formed by tandem hybridization of two extended antisense oligonucleotides whose ends were separately blocked with a cancer cell-specific aptamer, AS1411, and a replaceable anti-biomarker probe (ABP). We demonstrated that this DNA nanodrug was directed to cancer cells with the guidance power of AS1411 and then activated by the presence of a given intracellular biomarker. By using such a belt-and-braces strategy, this DNA nanodrug system could safely and efficiently accelerate apoptosis of target cancer cells. Moreover, since the expression level of biomarkers tends to indicate the specific physiological state of individual cells, biomarker-responsive activation of the nanodrug is expected to enable customized drug administration at the cellular level.

18.
J Am Chem Soc ; 140(1): 258-263, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29211455

RESUMO

DNA molecular machines show great promise in fields such as biomarker discovery and biological activity regulation, but operating DNA machines with specific functions within living systems remains extremely challenging. Although DNA machines have been engineered with exact molecular-level specifications, some intrinsic imperfections such as poor cell permeation and fragility in complex cytoplasmic milieu persist due to the well-established character of nucleic acid molecules. To circumvent these problems, we herein report a molecularly engineered, entropy-driven three-dimensional DNA amplifier (EDTD) that can operate inside living cells in response to a specific mRNA target. In particular, mRNA target/EDTD interaction can specifically initiate an autonomous DNA circuit inside living cells owing to the exclusive entropy-driven force, thus providing enormous signal amplification for ultrasensitive detection of the mRNA. Moreover, owing to molecular engineering of a unique DNA tetrahedral framework into the DNA amplifier, EDTD exhibits significantly enhanced biostability and cellular uptake efficiency, which are prerequisites for DNA machines used for in vivo applications. This programmable DNA machine presents a simple and modular amplification mechanism for the detection of intracellular biomarkers. Moreover, this study provides a potentially valuable molecular tool for understanding the chemistry of cellular systems and offers a design blueprint for further expansion of DNA nanotechnology in living systems.


Assuntos
DNA/química , RNA Mensageiro/química , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Imagem Óptica , Relação Estrutura-Atividade , Termodinâmica
19.
ACS Nano ; 11(4): 4060-4066, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28328200

RESUMO

Accurate detection and imaging of tumor-related mRNA in living cells hold great promise for early cancer detection. However, currently, most probes designed to image intracellular mRNA confront intrinsic interferences arising from complex biological matrices and resulting in inevitable false-positive signals. To circumvent this problem, an intracellular DNA nanoprobe, termed DNA tetrahedron nanotweezer (DTNT), was developed to reliably image tumor-related mRNA in living cells based on the FRET (fluorescence resonance energy transfer) "off" to "on" signal readout mode. DTNT was self-assembled from four single-stranded DNAs. In the absence of target mRNA, the respectively labeled donor and acceptor fluorophores are separated, thus inducing low FRET efficiency. However, in the presence of target mRNA, DTNT alters its structure from the open to closed state, thus bringing the dual fluorophores into close proximity for high FRET efficiency. The DTNT exhibited high cellular permeability, fast response and excellent biocompatibility. Moreover, intracellular imaging experiments showed that DTNT could effectively distinguish cancer cells from normal cells and, moreover, distinguish among changes of mRNA expression levels in living cells. The DTNT nanoprobe also exhibits minimal effect of probe concentration, distribution and laser power as other ratiometric probe. More importantly, as a result of the FRET "off" to "on" signal readout mode, the DTNT nanoprobe almost entirely avoids false-positive signals due to intrinsic interferences, such as nuclease digestion, protein binding and thermodynamic fluctuations in complex biological matrices. This design blueprint can be applied to the development of powerful DNA nanomachines for biomedical research and clinical early diagnosis.


Assuntos
DNA de Cadeia Simples/química , Transferência Ressonante de Energia de Fluorescência/métodos , Nanoestruturas/química , Neoplasias/química , RNA Mensageiro/análise , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Sobrevivência Celular , Corantes Fluorescentes/química , Humanos , Limite de Detecção , Neoplasias/diagnóstico , Imagem Óptica/métodos , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA