Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Theranostics ; 13(12): 4166-4181, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37554274

RESUMEN

Rationale: Although neoantigen-based cancer vaccines have shown promise in various solid tumors, limited immune responses and clinical outcomes have been reported in patients with advanced disease. Cytosolic transport of neoantigen and adjuvant is required for the activation of intracellular Toll-like receptors (TLRs) and cross-presentation to prime neoantigen-specific CD8+T cells but remains a significant challenge. Methods: In this study, we aimed to develop a virus-like silicon vaccine (V-scVLPs) with a unique spike topological structure, capable of efficiently co-delivering a hepatocellular carcinoma (HCC)-specific neoantigen and a TLR9 agonist to dendritic cells (DCs) to induce a robust CD8+T cell response to prevent orthotopic tumor growth. We evaluated the antitumor efficacy of V-scVLPs by examining tumor growth and survival time in animal models, as well as analyzing tumor-infiltrating CD8+T cells and cytokine responses in the tumor microenvironment (TME). To evaluate the synergistic efficacy of V-scVLPs in combination with α-TIM-3 in HCC, we used an orthotopic HCC mouse model, a lung metastasis model, and a tumor rechallenge model after hepatectomy. Results: We found that V-scVLPs can efficiently co-deliver the hepatocellular carcinoma (HCC)-specific neoantigen and the TLR9 agonist to DCs via caveolin-mediated endocytosis. This advanced delivery strategy results in efficient lymph node draining of V-scVLPs to activate lymphoid DC maturation for promoting robust CD8+T cells and central memory T cells responses, which effectively prevents orthotopic HCC tumor growth. However, in the established orthotopic liver tumor models, the inhibitory receptor of TIM-3 was significantly upregulated in tumor-infiltrating CD8+T cells after immunization with V-scVLPs. Blocking the TIM-3 signaling further restored the antitumor activity of V-scVLPs-induced CD8+T cells, reduced the proportion of regulatory T cells, and increased the levels of cytokines to alter the tumor microenvironment to efficiently suppress established orthotopic HCC tumor growth, and inhibit lung metastasis as well as recurrence after hepatectomy. Conclusion: Overall, the developed novel spike nanoparticles with efficient neoantigen and adjuvant intracellular delivery capability holds great promise for future clinical translation to improve HCC immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias Pulmonares , Ratones , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Receptor 2 Celular del Virus de la Hepatitis A/uso terapéutico , Receptor Toll-Like 9 , Citocinas/metabolismo , Linfocitos T CD8-positivos , Vacunas contra el Cáncer/uso terapéutico , Caveolina 1/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Microambiente Tumoral
2.
J Hepatocell Carcinoma ; 9: 127-140, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35300207

RESUMEN

Purpose: This study aimed to assess the efficacy and safety of adjuvant transarterial chemoembolization (TACE) plus tyrosine kinase inhibitor (TKI) treatment in patients with hepatocellular carcinoma (HCC) with a high risk of early recurrence after curative resection. Patients and Methods: Patients from multiple centres were divided into postoperative adjuvant TACE with (n=57) or without (n=142) TKI administration groups. The disease-free survival (DFS) curve was depicted by the Kaplan-Meier method, and the difference between the two groups was tested using the log rank test. Univariate and multivariate Cox analyses were performed to identify independent risk factors for DFS. Additionally, three propensity score analyses were performed to minimise the potential confounding factors to facilitate a more reliable conclusion. Adverse events (AEs) were assessed according to the Common Terminology Criteria for Adverse Events, version 4.0. Results: The 1-and 2-year DFS rates of the TACE plus TKI treatment group were 45.5% and 34.9%, respectively, which were significantly better than those of the TACE alone group (26.8% and 18.3%, respectively). Multivariate analysis identified adjuvant TACE plus TKI treatment as an independent prognostic factor for DFS (hazard ratio: 0.611, 95% confidence interval: 0.408-0.915, P=0.017). Further analysis based on the various propensity score methods yielded similar results. Subgroup analysis showed that patients with tumour diameter ≥5 cm, tumour number <3, absence of hepatic vein tumour thrombus and bile duct tumour thrombus, ruptured tumours, and stage IIIB could benefit more from TACE plus TKI treatment (all P<0.05). Some patients (33.33%) experienced grade ≥3 AEs in the TACE plus TKI group. Conclusion: TACE plus TKI treatment can reduce the incidence of early recurrence with tolerable adverse events in HCC patients at high risk of recurrence after hepatectomy and may be an appropriate option in postoperative anti-recurrence treatment.

3.
Adv Healthc Mater ; 11(11): e2102526, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35134273

RESUMEN

Combinatorial photothermal and immunotherapy have demonstrated great potential to remove primary tumors, suppress metastases, and prevent tumor recurrence. However, this strategy still confronts patients with many limitations, such as complex components, sophisticated construction, and inadequate therapeutic efficacy. In this work, small molecules of porphyrin derivatives (PPor) which can self-assemble into monodispersed nanoparticles without supplement of any other ingredients or surfactants are developed. The formed PPor nanoparticles (PPor NPs) exhibit highly photothermal conversion efficiency of 70% and NIR-II luminous abilities originate from the strong intramolecular charge transfer (ICT) effect of D-A structure under 808 nm laser irradiation, thus achieving NIR-II fluorescence imaging guided photothermal therapy (PTT) against primary tumors with a high cure rate. More importantly, tumor-associated antigens (TAAs), together with damage-associated molecular patterns (DAMPs) released from PTT-treated cancer cells, are proved to elicit immune responses to some degree. After combination with programmed cell death-1 (PD-1) antibodies, a robust systematic antitumor immunity is generated to restrain both primary and abscopal tumors growth, prolong survival, and prevent pulmonary metastasis on an aggressive 4T1 murine breast tumor model. Thus, this study provides a promising therapeutic paradigm with porphyrin derivatives nano-assembly as phototheranostic agents for the treatment of aggressive tumors with high efficiency.


Asunto(s)
Nanopartículas , Porfirinas , Animales , Línea Celular Tumoral , Humanos , Inmunoterapia , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Imagen Óptica/métodos , Fototerapia/métodos , Porfirinas/farmacología
4.
Nanoscale ; 13(3): 1745-1758, 2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33432957

RESUMEN

Recently, photothermal therapy (PTT) in the second near-infrared (NIR-II) biowindow has emerged as a promising treatment modality; however, its therapeutic outcomes are still limited by heterogeneous heat distribution and insufficient control of metastatic lesions. Tremendous efforts have been made to overcome the PTT's shortcomings by combining PTT with immunotherapy, but unfortunately current strategies still suffer from low response rates, primary/acquired resistance or severe immune-related adverse events. Herein, a novel photothermal agent and gene co-delivery nanoparticle (CSP), with CuS inside the SiO2 pore channels and PDMAEMA polycation on the outside of SiO2 surface, is explored for tumor localized NIR-II PTT and in situ immunotherapy through local generation of IL-12 cytokine. The resulting CSP integrated with the plasmid encoding IL-12 gene (CSP@IL-12) exhibited good gene transfection efficiency, outstanding NIR-II PTT effect and excellent therapeutic outcomes both in vitro and in vivo. Meanwhile, such an in situ joint therapy modality could significantly induce systemic immune responses including promoting DC maturation, CD8+ T cell proliferation and infiltration to efficiently eliminate possible metastatic lesions through abscopal effects. Hence, this creative combinational strategy of NIR-II PTT and IL-12 cytokine therapy might provide a more efficient, controllable and safer alternative strategy for future photo-immunotherapy.


Asunto(s)
Hipertermia Inducida , Neoplasias , Citocinas , Humanos , Inmunoterapia , Interleucina-12/genética , Neoplasias/terapia , Fototerapia , Dióxido de Silicio
5.
ACS Appl Mater Interfaces ; 11(10): 9804-9813, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30773883

RESUMEN

Recently reported black phosphorus quantum dots (BPQDs) possess unique photocatalysis activities. However, the environmental instability accompanied by a hypoxic tumor microenvironment (TME) seriously hindered the bioapplications of BPQDs, especially in oxygen-dependent photodynamic therapy (PDT). Here, we construct a hepatocellular carcinoma (HCC)-specific targeting aptamer "TLS11a"-decorated BPQDs-hybridized nanocatalyst, which can specifically target HCC tumor cells and self-compensate oxygen (O2) into hypoxic TME for enhancing PDT efficiency. The BPQD-hybridized mesoporous silica framework (BMSF) with in situ synthesized Pt nanoparticles (PtNPs) in the BMSF is simply prepared. After being decorated by TLS11a aptamer/Mal-PEG-NHS, the resultant nanosystem (refer as Apt-BMSF@Pt) exhibits excellent environmental stability, active targeting ability to HCC cells, and self-compensation ability of oxygen. Compared with the PEG-BMSF@Pt without H2O2 incubation, the PEG-BMSF@Pt nanocatalyst exhibits 4.2-folds O2 and 1.6-folds 1O2 generation ability in a mimetic closed-system in the presence of both H2O2 and near-infrared laser. In a mouse model, the Apt-BMSF@Pt can effectively accumulate into tumor sites, and the core of BMSF subsequently can act as a photosensitizer to generate reactive oxygen species, while the PtNPs can serve as a catalyst to convert H2O2 into O2 for enhancing PDT through self-compensation mechanisms in hypoxic TME. By comparison of the tumor volume/weight, H&E, and immunohistochemical analysis, the excellent antitumor effects with minimized side effects of our Apt-BMSF@Pt could be demonstrated in vivo. Taken together, the current study suggests that our Apt-BMSF@Pt could act as an active targeting nanocatalyst for programmable killing of cancer cells in hypoxic TME.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Animales , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Catálisis , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Humanos , Peróxido de Hidrógeno/química , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Oxígeno/metabolismo , Fósforo/química , Fósforo/farmacología , Fármacos Fotosensibilizantes/química , Puntos Cuánticos/administración & dosificación , Puntos Cuánticos/química , Dióxido de Silicio/química , Dióxido de Silicio/farmacología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Anal Chem ; 90(11): 7086-7093, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29775052

RESUMEN

A new double photosystems-based 'Z-scheme' photoelectrochemical (PEC) sensing platform is designed for ultrasensitive detection of prostate-specific antigen (PSA) by coupling with a three-dimensional (3D) DNA walker. Two photosystems consist of CdS quantum dots (photosystem I; PS I) and BiVO4 photoactive materials (photosystem II; PS II), whereas gold nanoparticles (AuNPs) photodeposited on high-active {010} facets of BiVO4 are used as the electron mediators to promote electron transfer from conduction band of PS II to valence band of PS I. 3D DNA walker-based amplification strategy is carried out between hairpin DNA1 conjugated onto the AuNP, hairpin DNA2 labeled with CdS quantum dot (QD-H2), and DNA walker complementary with the PSA aptamer modified to a magnetic bead (Apt-MB). Upon addition of target, DNA walker strand is displaced from DNA walker/Apt-MB to open hairpin DNA1 on AuNP@BiVO4. In the presence of QD-H2, DNA walker induces the hybridization of DNA1 with DNA2 on the gold nanoparticles step by step, thereby resulting in the assembly of CdS QDs on the AuNP@BiVO4 to form Z-scheme double photosystems with strong photocurrent. Under optimum conditions, the Z-scheme PEC sensing system exhibits good photocurrent responses toward target PSA within the working range of 0.01-50 ng mL-1 at a low detection limit of 1.5 pg mL-1. Good reproducibility and accuracy are acquired for analysis of target PSA and human serum specimens relative to the commercial PSA ELISA kit. Importantly, our strategy provides a new horizon for photoelectrochemical in vitro diagnostics.


Asunto(s)
Técnicas Biosensibles , ADN/química , Técnicas Electroquímicas , Fármacos Fotosensibilizantes/farmacología , Antígeno Prostático Específico/sangre , Puntos Cuánticos/química , Biomarcadores/sangre , Bismuto/química , Compuestos de Cadmio/química , Ensayo de Inmunoadsorción Enzimática , Oro/química , Humanos , Tamaño de la Partícula , Procesos Fotoquímicos , Fármacos Fotosensibilizantes/química , Sulfuros/química , Propiedades de Superficie , Vanadatos/química
7.
Nanoscale ; 9(30): 10861-10868, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28731107

RESUMEN

Herein, a novel dual-responsive two-color fluorescent nanoprobe has been designed for the fluorescence activation imaging of cell apoptosis in living cells. The nanoprobe consists of a gold nanoparticle core functionalized with a dense layer of DNA aptamers and peptides, which shows high affinity and specific response to cytochrome c (Cyt c) and caspase-3, respectively. The formation of the aptamer-Cyt c complex and the cleavage of the specific peptide by caspase-3 can liberate the dye labelled aptamers and peptides from the surface of gold nanoparticles, and then recover their fluorescence. The turn-on and specific recognition properties of our nanoprobe allow for the sensitive and selective detection of Cyt c concentration and caspase-3 activity both in solutions and in living cells. The here proposed nanoprobe with the abilities of real-time monitoring the cell apoptosis and evaluating the apoptosis-related drug efficacy might serve as a potential interesting tool for studying the molecular mechanisms of apoptosis regulation or screening the apoptosis-based drugs.


Asunto(s)
Apoptosis , Evaluación Preclínica de Medicamentos , Colorantes Fluorescentes , Nanopartículas del Metal , Aptámeros de Nucleótidos , Oro , Células HeLa , Humanos , Imagen Óptica
8.
J Mater Chem B ; 4(4): 589-599, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-32262941

RESUMEN

Incorporating multiple imaging modalities and simultaneous therapeutic functions together into one single nano-formulation is of great importance for developing high-performance clinical-translatable theranostic agents. Herein, we fabricated multifunctional lipid-micelles incorporated with semiconducting polymer dots and a photosensitizer (referred as Pdots/Ce6@lipid-Gd-DOTA micelles) for combined magnetic resonance imaging (MRI)/photoacoustic imaging (PAI) and photodynamic (PDT)/photothermal (PTT) dual-modal therapy that induced by a single laser to achieve enhanced cancer therapeutic efficiency. The Pdots/Ce6@lipid-Gd-DOTA micelles with excellent water dispersibility were comprised of a core with poly[2,6-(4,4-bis-(2-ethylhexyl)-4H-cyclopenta[2,1-b;3,4-b']-dithiophene)-alt-4,7-(2,1,3-benzo-thiadiazole)] dots (Pdots) and Ce6 molecules inside, and a lipid-PEG outlayer conjugated with gadolinium-1,4,7,10-tetraacetic acid. The prepared Pdots/Ce6@lipid-Gd-DOTA micelles exhibited extremely low cytotoxicity, and had excellent MR- and PA-imaging contrast-enhancement ability, which could synchronously offer anatomical information and morphological information of tumors. Moreover, both Pdots and Ce6 photosensitizer, encapsulated inside the lipid-Gd-DOTA micelles, exhibited high NIR absorption at 670 nm and were applied to combine photothermal and photodynamic therapy simultaneously to achieve enhanced synergistic cancer therapeutic efficiency both in vitro and in vivo. In summary, our studies demonstrated that Pdots/Ce6@lipid-Gd-DOTA micelles with multi-diagnosis modalities and simultaneous dual-modal photo-therapy functions might be a potential interesting theranostic platform for tumor treatment.

9.
ACS Appl Mater Interfaces ; 7(15): 8176-87, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25837008

RESUMEN

Photodynamic therapy (PDT), using a combination of chemical photosensitizers (PS) and light, has been successfully applied as a noninvasive therapeutic procedure to treat tumors by inducing apoptosis or necrosis of cancer cells. However, most current clinically used PS have suffered from the instability in physiological conditions which lead to low photodynamic therapy efficacy. Herein, a highly biocompatible poly(dopamine) (PDA) nanoparticle conjugated with Chlorin e6 (referenced as the PDA-Ce6 nanosphere) was designed as a nanotherapeutic agent to achieve simultaneous photodynamic/photothermal therapy (PDT/PTT). Compared to the free Ce6, the PDA-Ce6 nanosphere exhibited significantly higher PDT efficacy against tumor cells, because of the enhanced cellular uptake and subsequently greater reactive oxygen species (ROS) production upon laser irradiation at 670 nm. Meanwhile, the PDA-Ce6 nanosphere could be also used as a photoabsorbing agent for PTT, because of the excellent photothermal conversion ability of PDA nanoparticle under laser irradiation at 808 nm. Moreover, our prepared nanosphere had extremely low dark toxicity, while excellent phototoxicity under the combination laser irradiation of 670 and 808 nm, both in vitro and in vivo, compared to any single laser irradiation alone. Therefore, our prepared PDA-Ce6 nanosphere could be applied as a very promising dual-modal phototherapeutic agent for enhanced cancer therapy in future clinical applications.


Asunto(s)
Hipertermia Inducida/métodos , Indoles/uso terapéutico , Nanoconjugados/administración & dosificación , Nanosferas/administración & dosificación , Fotoquimioterapia/métodos , Polímeros/uso terapéutico , Porfirinas/uso terapéutico , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Clorofilidas , Terapia Combinada/métodos , Difusión , Células Hep G2 , Humanos , Indoles/química , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanoconjugados/química , Nanosferas/química , Fármacos Fotosensibilizantes/uso terapéutico , Polímeros/química
10.
Nanotechnology ; 26(11): 115102, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25721867

RESUMEN

In this paper, a core­shell nanocomposite of clusters of superparamagnetic iron oxide nanoparticles coated with poly(dopamine) (SPION clusters@PDA) is fabricated as a magnetic field-directed theranostic agent that combines the capabilities of highly sensitive magnetic resonance imaging (MRI) and photothermal cancer therapy. The highly concentrated SPION cluster core is suitable for sensitive MRI due to its superparamagnetic properties, and the poly(dopamine) coating layer can induce cancer cell death under near-infrared (NIR) laser irradiation because of the photothermal conversion ability of PDA. MRI scanning reveals that the nanocomposite has relatively high r2 and r2(*) relaxivities, and the r2(*) values are nearly threefold higher than the r2 values because of the clustering of the SPIONs in the nanocomposite core. Due to the rapid response to magnetic field gradients, enhanced cellular uptake of our nanocomposite mediated by an external magnetic field can be achieved, thus producing significantly enhanced local photothermal killing efficiency against cancer cells under NIR irritation.


Asunto(s)
Compuestos Férricos/química , Imagen por Resonancia Magnética/métodos , Nanopartículas del Metal/química , Nanotecnología/métodos , Neoplasias/patología , Fototerapia/métodos , Animales , Medios de Contraste/química , Dextranos/química , Dopamina/química , Ferrocianuros/química , Células HeLa , Células Hep G2 , Humanos , Indoles/química , Rayos Láser , Campos Magnéticos , Nanopartículas de Magnetita/química , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Células 3T3 NIH , Nanocompuestos/química , Polímeros/química , Espectroscopía Infrarroja Corta , Temperatura
11.
Nanotechnology ; 26(2): 025102, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25517859

RESUMEN

The combination of a multi-therapeutic mode with a controlled fashion is a key improvement in nanomedicine. Here, we synthesized polyethylene glycol (PEG)-modified doxorubicin (DOX)-loaded mesoporous silica nanoparticle (MSN) @CuS nanohybrids as efficient drug delivery carriers, combined with photothermal therapy and chemotherapy to enhance the therapeutic efficacy on hepatocellular carcinoma (HCC). The physical properties of the nanohybrids were characterized by transmission electron microscopy (TEM), N2 adsorption and desorption experiments and by the Vis-NIR absorption spectra. The results showed that the doxorubicin could be stored in the inner pores of mesoporous silica nanoparticles; the CuS nanoparticles, which are coated on the surface of a mesoporous silica nanoparticle, could serve as efficient photothermal therapy (PTT) agents; the loaded drug release could be easily triggered by NIR irradiation. The combination of the PTT treatment with controlled chemotherapy could further enhance the cancer ablation ability compared to any of the single approaches alone. Hence, the reported PEG-modified DOX-loaded mesoporous silica nanoparticle@CuS nanohybrids might be very promising therapeutic agents for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas/química , Polietilenglicoles/química , Dióxido de Silicio/química , Adsorción , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cobre/química , Doxorrubicina/farmacología , Células Hep G2 , Humanos , Hipertermia Inducida , Rayos Láser , Nanopartículas/ultraestructura , Fototerapia , Polietilenglicoles/síntesis química , Porosidad , Dióxido de Silicio/síntesis química , Espectroscopía Infrarroja Corta , Temperatura
12.
ACS Appl Mater Interfaces ; 6(16): 14266-77, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25090604

RESUMEN

Multifunctional theranostic nanoparticles represent an emerging agent with the potential to offer extremely sensitive diagnosis and targeted cancer therapy. Herein, we report the synthesis and characterization of a multifunctional theranostic agent (referred to as LA-LAPNHs) for targeted magnetic resonance imaging/computed X-ray tomography (MRI/CT) dual-mode imaging and photothermal therapy of hepatocellular carcinoma. The LA-LAPNHs were characterized as having a core-shell structure with the gold nanoparticles (AuNPs)@polydopamine (PDA) as the inner core, the indocyanine green (ICG), which is electrostatically absorbed onto the surface of PDA, as the photothermal therapeutic agent, and the lipids modified with gadolinium-1,4,7,10-tetraacetic acid and lactobionic acid (LA), which is self-assembled on the outer surface as the shell. The LA-LAPNHs could be selectively internalized into the hepatocellular cell line (HepG2 cells) but not into HeLa cells due to the specific recognition ability of LA to asialoglycoprotein receptor. Additionally, the dual-mode imaging ability of the LA-LAPNH aqueous solution was confirmed by enhanced MR and CT imaging showing a shorter T1 relaxation time and a higher Hounsfield unit value, respectively. In addition, the LA-LAPNHs showed significant photothermal cytotoxicity against liver cancer cells with near-infrared irradiation due to their strong absorbance in the region between 700 and 850 nm. In summary, this study demonstrates that LA-LAPNHs may be a promising candidate for targeted MR/CT dual-mode imaging and photothermal therapy of hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/terapia , Diagnóstico por Imagen/métodos , Oro/química , Indoles/química , Lípidos/química , Nanopartículas del Metal/química , Fototerapia/métodos , Polímeros/química , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular , Gadolinio/química , Células HeLa , Células Hep G2 , Humanos , Verde de Indocianina/química , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/terapia , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Rayos X/métodos
13.
Analyst ; 138(17): 4732-6, 2013 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-23814782

RESUMEN

We designed a novel aptamer based biosensor (aptasensor) for ultrasensitive detection of adenosine triphosphate (ATP) through resonance energy transfer (RET). The ATP aptamer was modified with Cy3 at the 3' end, and a green quantum dot (525) was attached to the 5' end of its complementary sequence respectively. The ATP aptamer and its complementary sequence could assemble into a duplex structure in the absence of target ATP, and then decrease the distance between the quantum dot and Cy3 which could produce significant RET signal. Upon ATP binding, the ATP aptamer could dissociate with its complementary sequence and then increase the distance between the quantum dot and Cy3 which would significantly decrease the RET signal. Therefore, the ATP detection could be easily achieved through detection of the fluorescence intensity ratio between 525 nm and 560 nm. The results show that the emission fluorescence intensity ratio of 525/560 is linearly related to the logarithmic concentration of ATP. The linear range of this aptasensor is from 0.1 nM to 1 µM, and the detection limit is lower down to 0.01 nM. Excellent selectivity of this aptasensor for ATP has been demonstrated through the detection of thymidine triphosphate (TTP), cytidine triphosphate (CTP), guanosine triphosphate (GTP) and adenosine diphosphate (ADP) respectively as control. The method we described here could easily detect ATP with excellent selectivity, linearity and sensitivity down to the nanomolar range, as well as avoid photobleaching.


Asunto(s)
Adenosina Trifosfato/análisis , Aptámeros de Nucleótidos/metabolismo , Técnicas Biosensibles/métodos , Transferencia Resonante de Energía de Fluorescencia , Puntos Cuánticos , Adenosina Trifosfato/metabolismo , Aptámeros de Nucleótidos/genética , Secuencia de Bases
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA